1
|
Zadeh FJ, Fateh A, Saffari H, Khodadadi M, Eslami Samarin M, Nikoubakht N, Dadgar F, Goodarzi V. The vaso-occlusive pain crisis in sickle cell patients: A focus on pathogenesis. Curr Res Transl Med 2025; 73:103512. [PMID: 40220659 DOI: 10.1016/j.retram.2025.103512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025]
Abstract
Vaso-occlusive pain crisis (VOC) is recognized as a prominent complication of sickle cell disease, accompanied by debilitating pain and serious consequences for patients, making it the primary cause of visits to hospital emergency departments. In the etiology of VOC, the intricate interaction of endothelial cells, hypoxia, inflammation, and the coagulation system is pivotal. Hemoglobin S polymerization under hypoxic conditions leads to the formation of rigid and adhesive red blood cells that interact with vascular endothelial cells and other blood cells, causing occlusion and subsequent inflammation. Hemolysis of red blood cells results in anemia and heightened inflammation, whereas oxidative stress and involvement of the coagulation system further complicate matters. In this review, we strive to examine the pathophysiology of VOC from these mentioned aspects by consolidating findings from various studies, as a comprehensive understanding of the causes of VOC is essential for the development of targeted therapeutic interventions and the prevention and management of pain, ultimately improving the quality of life for patients.
Collapse
Affiliation(s)
| | - Azadeh Fateh
- Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hamed Saffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammadamin Eslami Samarin
- Student Research Committee, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Universal Scientific Education and Research Network(USERN),Tehran,Iran
| | - Nasim Nikoubakht
- Department of Anesthesiology, Hazrat-e Rasool General Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Dadgar
- Department of Internal Medicine, Lorestan University of Medical Science, Khorramabad, Iran; Student Research Committe, Lorestan University of Medical Science, Khorramabad, Iran
| | - Vahid Goodarzi
- Department of Anesthesiology, Rasoul-Akram Medical Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
2
|
Elsherif L, Tang Y, Patillo KL, Wichlan D, Ogu UO, Landes K, McCune P, Scott LC, Gulledge W, Woodland WH, Nelson M, Loehr LR, Cronin RM, Desai PC, Zhou LY, Pollock DM, Zou F, Cai J, Derebail VK, Ataga KI. Association of biomarkers of endothelial function, coagulation activation and kidney injury with persistent albuminuria in sickle cell anaemia. Br J Haematol 2024; 205:1963-1973. [PMID: 39228027 DOI: 10.1111/bjh.19743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024]
Abstract
Persistent albuminuria (PA) is common in sickle cell anaemia (SCA). With the association of chronic kidney disease (CKD) with increased mortality, biomarkers that predict its development or progression are needed. We evaluated the association of select biomarkers with PA in adults with SCA using Kruskal-Wallis rank-sum test and logistic regression models, with adjustment for multiple testing. Of 280 subjects, 100 (35.7%) had PA. Median plasma levels of soluble vascular cell adhesion molecule-1 (VCAM-1) (1176.3 vs. 953.4 ng/mL, false discovery rate [FDR] q-value <0.003), thrombin-antithrombin complex (5.5 vs. 4.7 ng/mL, FDR q-value = 0.04), and urinary angiotensinogen (AGT) (12.2 vs. 5.3 ng/mg, FDR q-value <0.003), urinary nephrin (30.6 vs. 27.2 ng/mg, FDR q-value = 0.04), and urinary kidney injury molecule-1 (KIM-1) (0.8 vs. 0.5 ng/mg, FDR q-value <0.003), normalized to urine creatinine, were significantly higher in subjects with PA. In multivariable analysis, only urinary AGT (odds ratio = 1.058, FDR q-value <0.0001) remained a significant predictor of PA. In addition, soluble VCAM-1 (FDR q-value <0.0001), D-dimer (FDR q-value <0.0001), urinary AGT (FDR q-value <0.0001), KIM-1 (FDR q-value <0.0001), and nephrin (FDR q-value <0.0001) were significantly associated with urine albumin-creatinine ratio in multivariable analyses. Longitudinal studies to evaluate the predictive capacity of biomarkers for the development and progression of CKD in SCA are warranted.
Collapse
Affiliation(s)
- Laila Elsherif
- Center for Sickle Cell Disease, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Yihan Tang
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kammie L Patillo
- Office of Clinical Trials, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - David Wichlan
- Division of Hematology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Ugochi O Ogu
- Center for Sickle Cell Disease, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Kristina Landes
- Division of Hematology, The Ohio State University, Columbus, Ohio, USA
| | - Paula McCune
- Office of Clinical Trials, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Lara C Scott
- College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Whitney Gulledge
- College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Woodi H Woodland
- College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Marquita Nelson
- Center for Sickle Cell Disease, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Laura R Loehr
- Division of General Medicine and Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Robert M Cronin
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Payal C Desai
- Levine Cancer Institute - Atrium Health, Wake Forest School of Medicine, Charlotte, North Carolina, USA
| | - Laura Y Zhou
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - David M Pollock
- Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Fei Zou
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Vimal K Derebail
- Division of Nephrology and Hypertension, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kenneth I Ataga
- Center for Sickle Cell Disease, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
3
|
Youssry I, Mostafa AS, Hamed DH, Hafez YFA, Bishai IE, Selim YMM. Role of endothelial dysfunction in sleep-disordered breathing in egyptian children with sickle cell disease. BMC Pediatr 2024; 24:626. [PMID: 39354381 PMCID: PMC11443814 DOI: 10.1186/s12887-024-05066-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 09/09/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Endothelial dysfunction is an integral pathophysiologic mechanism in sickle cell disease (SCD), and can lead to many complications. Sleep-disordered breathing (SDB) is a SCD complication with diverse incidence and pathophysiology. This study aimed to determine the prevalence of SDB in children with SCD and to assess its relation to endothelial dysfunction. METHODS Sixty children with SCD and 60 healthy controls were enrolled. The levels of TNF-α, IL-6, and IL-17A were evaluated in the entire cohort using enzyme-linked immunosorbent assay (ELISA) kits. Polysomnography (PSG) was performed for all SCD patients after completion of the Pediatric Sleep Questionnaire (PSQ). RESULTS TNF-α, IL-6, and IL-17A levels were significantly greater in children with SCD than in controls (p-values < 0.001, < 0.001, and 0.006, respectively). The PSQ revealed symptoms suggestive of SDB in 50 children with SCD (83.3%), and PSG revealed obstructive sleep apnea (OSA) in 44 children with SCD (73.3%); 22 patients had mild OSA, and 22 had moderate-to-severe OSA according to the apnea-hypopnea index (AHI). TNF-α was significantly greater in SCD children who reported heavy or loud breathing, trouble breathing or struggle to breathe, and difficulty waking up in the morning (p-values = 0.002, 0.002, and 0.031, respectively). The IL-6 levels were significantly greater in SCD children who stopped growing normally (p-value = 0.002). The levels of IL-6 and IL-17A were significantly greater in SCD children with morning headaches (p-values = 0.007 and 0.004, respectively). CONCLUSION Children with SCD showed a high prevalence of SDB with significantly elevated levels of markers of endothelial function, highlighting the interplay of SDB and endothelial dysfunction in SCD.
Collapse
Affiliation(s)
- Ilham Youssry
- Department of Pediatric Hematology, Faculty ofMedicine, Cairo University, Giza, Egypt
| | - Abla S Mostafa
- Department of Pediatric Pulmonology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Dina H Hamed
- Department of Pediatric Pulmonology, Faculty of Medicine, Cairo University, Giza, Egypt
| | | | - Irene E Bishai
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Yasmeen M M Selim
- Department of Pediatric Hematology, Faculty ofMedicine, Cairo University, Giza, Egypt.
| |
Collapse
|
4
|
Pavan AR, Terroni B, Dos Santos JL. Endothelial dysfunction in Sickle Cell Disease: Strategies for the treatment. Nitric Oxide 2024; 149:7-17. [PMID: 38806107 DOI: 10.1016/j.niox.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/15/2024] [Accepted: 05/25/2024] [Indexed: 05/30/2024]
Abstract
Sickle Cell Anemia (SCA), is an inherited hemoglobinopathy characterized by the presence of an abnormal hemoglobin (HbS), being the most prevalent sickle cell disease (SCD). SCA is characterized by vascular endothelial dysfunction, which contributes significantly to various clinical conditions, including but not limited to pulmonary hypertension, priapism, cutaneous leg ulceration, and stroke. The pathophysiology of endothelial dysfunction (ED) in SCA is a multifaceted process involving a chronic inflammatory and hypercoagulable state. Key factors include hemolysis-associated elements like reduced arginine and nitric oxide (NO) availability, elevated levels of vascular adhesion molecules, the uncoupling effect of NO synthase, heightened arginase activity, an environment characterized by oxidative stress with the production of reactive oxygen and nitrogen species, and occurrences of ischemia-reperfusion injury, along with apolipoprotein A-1 depletion. The urgency for novel interventions addressing ED is evident. Presently, there is a focus on investigating small molecules that disrupt the arginine-nitric oxide pathway, exhibiting anti-inflammatory and antioxidant properties while diminishing levels of cellular and vascular adhesion molecules. In this mini-review article, we delve into the progress made in strategies for treating ED in SCD with the aim of cultivating insights for drug design.
Collapse
Affiliation(s)
- Aline Renata Pavan
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, Brazil; São Paulo State University (UNESP), Institute of Chemistry, Araraquara, Brazil.
| | - Barbara Terroni
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, Brazil
| | | |
Collapse
|
5
|
Peterson JM, Smith TA, Rock EP, Magnani JL. Selectins in Biology and Human Disease: Opportunity in E-selectin Antagonism. Cureus 2024; 16:e61996. [PMID: 38983984 PMCID: PMC11232095 DOI: 10.7759/cureus.61996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2024] [Indexed: 07/11/2024] Open
Abstract
Selectins are cell adhesion proteins discovered in the 1980s. As C-type lectins, selectins contain an essential calcium ion in the ligand-binding pocket and recognize the isomeric tetrasaccharides sialyl Lewisx (sLex) and sialyl Lewisa (sLea). Three selectins, E-selectin, P-selectin, and L-selectin, play distinct, complementary roles in inflammation, hematopoiesis, and tumor biology. They have been implicated in the pathology of diverse inflammatory disorders, and several selectin antagonists have been tested clinically. E-selectin plays a unique role in leukocyte activation, making it an attractive target for intervention, for example, in sickle cell disease (SCD). This review summarizes selectin biology and pathology, structure and ligand binding, and selectin antagonists that have reached clinical testing with an emphasis on E-selectin.
Collapse
Affiliation(s)
| | | | - Edwin P Rock
- Development, GlycoMimetics, Inc., Rockville, USA
| | - John L Magnani
- Research and Development, GlycoTech Corporation, Rockville, USA
| |
Collapse
|
6
|
Uy GL, DeAngelo DJ, Lozier JN, Fisher DM, Jonas BA, Magnani JL, Becker PS, Lazarus HM, Winkler IG. Targeting hematologic malignancies by inhibiting E-selectin: A sweet spot for AML therapy? Blood Rev 2024; 65:101184. [PMID: 38493006 PMCID: PMC11051645 DOI: 10.1016/j.blre.2024.101184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/06/2024] [Accepted: 02/26/2024] [Indexed: 03/18/2024]
Abstract
E-selectin, a cytoadhesive glycoprotein, is expressed on venular endothelial cells and mediates leukocyte localization to inflamed endothelium, the first step in inflammatory cell extravasation into tissue. Constitutive marrow endothelial E-selectin expression also supports bone marrow hematopoiesis via NF-κB-mediated signaling. Correspondingly, E-selectin interaction with E-selectin ligand (sialyl Lewisx) on acute myeloid leukemia (AML) cells leads to chemotherapy resistance in vivo. Uproleselan (GMI-1271) is a carbohydrate analog of sialyl Lewisx that blocks E-selectin binding. A Phase 2 trial of MEC chemotherapy combined with uproleselan for relapsed/refractory AML showed a median overall survival of 8.8 months and low (2%) rates of severe oral mucositis. Clinical trials seek to confirm activity in AML and mitigation of neutrophil-mediated adverse events (mucositis and diarrhea) after intensive chemotherapy. In this review we summarize E-selectin biology and the rationale for uproleselan in combination with other therapies for hematologic malignancies. We also describe uproleselan pharmacology and ongoing clinical trials.
Collapse
Affiliation(s)
- Geoffrey L Uy
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel J DeAngelo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | - Brian A Jonas
- Department of Internal Medicine, Division of Malignant Hematology/Cellular Therapy and Transplantation, University of California Davis, Davis, CA, USA
| | | | - Pamela S Becker
- Leukemia Division, Department of Hematology and Hematopoietic Cell Transplantation, Department of Hematologic Malignancies Translational Science, City of Hope National Medical Center, Duarte, CA, USA
| | - Hillard M Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ingrid G Winkler
- Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, Woolloongabba, QLD, Australia
| |
Collapse
|
7
|
Delgadinho M, Veiga L, Ginete C, Santos B, Miranda A, de Vasconcelos JN, Brito M. Differential expression of adhesion molecules in sickle cell anemia and gut microbiome effect. Ann Hematol 2024; 103:409-419. [PMID: 38153527 PMCID: PMC10799142 DOI: 10.1007/s00277-023-05589-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/12/2023] [Indexed: 12/29/2023]
Abstract
Sickle cell anemia (SCA) causes a long-standing vascular inflammation state, leading to endothelial dysfunction and chronic overexpression of several adhesion molecules, which contributes to acute and constant vaso-occlusive (VOC) episodes. It has been demonstrated that hydroxyurea (HU) can reduce VOC events, organ damage, blood transfusions, and even the adhesion properties to endothelial cells of SCA subjects. Due to VOC episodes, these patients are also more susceptible to recurrent bacterial translocation and dysbiosis. Given this, our study aimed to uncover the interplay between adhesion molecules, gut microbiome, and hydroxyurea in a population of Angolan SCA children. Serum and fecal samples were obtained before and after HU treatment in 35 children. After HU, four of these adhesion molecules were significantly reduced: sE-selectin (p = 0.002), ADAMTS13 (p = 0.023), sICAM-1 (p = 0.003), and sVCAM-1 (p = 0.018). A positive correlation was observed between the number of neutrophils and sICAM-1, platelets, and sP-selectin, and also between leukocytes, sICAM-1, and sVCAM-1. Most taxa showing a significant correlation mainly belonged to the Clostridiales order. Specifically, from the Clostridium genera, the groups g19, g21, and g34 were all negatively correlated with HbF levels; g19, g21, and g24 positively correlated with leukocytes; g19 positively with neutrophils and sVCAM-1; and g34 positively with E- and P-selectin. Serratia, an opportunistic pathogen, was positively correlated with sE-selectin and sICAM-1 levels. Additionally, a negative correlation was observed between sP-selectin and Bifidobacterium. Research studies in this area could improve our understanding and contribute to finding new prognostic biomarkers to guarantee precise SCA patient stratification and predict severe complications.
Collapse
Affiliation(s)
- Mariana Delgadinho
- H&TRC - Health & Technology Research Center, ESTeSL - Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Luísa Veiga
- H&TRC - Health & Technology Research Center, ESTeSL - Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Catarina Ginete
- H&TRC - Health & Technology Research Center, ESTeSL - Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Brígida Santos
- Centro de Investigação em Saúde de Angola (CISA), Caxito, Bengo, Angola
- Hospital Pediátrico David Bernardino (HPDB), Luanda, Angola
| | - Armandina Miranda
- Instituto Nacional de Saúde Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | | | - Miguel Brito
- H&TRC - Health & Technology Research Center, ESTeSL - Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal.
- Centro de Investigação em Saúde de Angola (CISA), Caxito, Bengo, Angola.
| |
Collapse
|
8
|
Lizarralde-Iragorri MA, Parachalil Gopalan B, Merriweather B, Brooks J, Hill M, Lovins D, Pierre-Charles R, Cullinane A, Dulau-Florea A, Lee DY, Villasmil R, Jeffries N, Shet AS. Isoquercetin for thromboinflammation in sickle cell disease: a randomized double-blind placebo-controlled trial. Blood Adv 2024; 8:172-182. [PMID: 38157227 PMCID: PMC10787266 DOI: 10.1182/bloodadvances.2023011542] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/07/2023] [Indexed: 01/03/2024] Open
Abstract
ABSTRACT Data from a small trial in patients with cancer suggest that isoquercetin (IQ) treatment lowered thrombosis biomarkers and prevented clinical thrombosis, but, to our knowledge, no studies of IQ have been conducted to target thromboinflammation in adults with sickle cell disease (SCD). We conducted a randomized, double-blind, placebo-controlled trial in adults with steady-state SCD (hemoglobin SS [HbSS], HbSβ0thal, HbSβ+thal, or HbSC). The primary outcome was the change in plasma soluble P-selectin (sP-selectin) after treatment compared with baseline, analyzed in the intention-to-treat population. Between November 2019 and July 2022, 46 patients (aged 40 ± 11 years, 56% female, 75% under hydroxyurea treatment) were randomized to receive IQ (n = 23) or placebo (n = 23). IQ was well tolerated and all the adverse events (AEs; n = 21) or serious AEs (n = 14) recorded were not attributable to the study drug. The mean posttreatment change for sP-selectin showed no significant difference between the treatment groups (IQ, 0.10 ± 6.53 vs placebo, 0.74 ± 4.54; P = .64). In patients treated with IQ, whole-blood coagulation (P = .03) and collagen-induced platelet aggregation (P = .03) were significantly reduced from the baseline. Inducible mononuclear cell tissue factor gene expression and plasma protein disulfide isomerase reductase activity were also significantly inhibited (P = .003 and P = .02, respectively). Short-term fixed-dose IQ in patients with SCD was safe with no off-target bleeding and was associated with changes from the baseline in the appropriate direction for several biomarkers of thromboinflammation. The trial was registered at www.clinicaltrials.gov as #NCT04514510.
Collapse
Affiliation(s)
- Maria A Lizarralde-Iragorri
- Sickle Thrombosis and Vascular Biology Lab, Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Bindu Parachalil Gopalan
- Sickle Thrombosis and Vascular Biology Lab, Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Brenda Merriweather
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Jennifer Brooks
- Office of the Clinical Director, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Mai Hill
- Office of the Clinical Director, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Dianna Lovins
- Office of the Clinical Director, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Ruth Pierre-Charles
- Office of the Clinical Director, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Ann Cullinane
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Alina Dulau-Florea
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Duck-Yeon Lee
- Biochemistry Core Facility, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Rafael Villasmil
- Flow Cytometry Core Facility, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Neal Jeffries
- Office of Biostatistics Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Arun S Shet
- Sickle Thrombosis and Vascular Biology Lab, Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
9
|
Hakami F, Alhazmi E, Busayli WM, Althurwi S, Darraj AM, Alamir MA, Hakami A, Othman RA, Moafa AI, Mahasi HA, Madkhali MA. Overview of the Association Between the Pathophysiology, Types, and Management of Sickle Cell Disease and Stroke. Cureus 2023; 15:e50577. [PMID: 38107212 PMCID: PMC10723021 DOI: 10.7759/cureus.50577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2023] [Indexed: 12/19/2023] Open
Abstract
Sickle cell disease (SCD) is a genetic blood disorder that affects hemoglobin and increases stroke risk, particularly in childhood. This review examines the pathophysiological association between SCD and stroke, the classification of stroke types, risk factors, diagnosis, management, prevention, and prognosis. A comprehensive literature search was conducted via PubMed, Scopus, and Cochrane databases. Relevant studies on SCD and stroke pathophysiology, classification, epidemiology, diagnosis, treatment, and prevention were identified. Sickle cell disease causes red blood cells to become rigid and sickle-shaped, obstructing blood vessels. Recurrent sickling alters cerebral blood flow and damages vessel walls, often leading to ischemic or hemorrhagic strokes (HS). These occur most frequently in childhood, with ischemic strokes (IS) being more common. Key risk factors include a prior transient ischemic attack (TIA), low hemoglobin, and a high leukocyte count. Neuroimaging is essential for diagnosis and determining stroke type. Primary prevention centers on blood transfusions and hydroxyurea for those at high risk. Acute treatment involves promptly restoring blood flow and managing complications. However, significant knowledge gaps remain regarding stroke mechanisms, optimizing screening protocols, and improving long-term outcomes. This review synthesizes current evidence on SCD and stroke to highlight opportunities for further research and standardizing care protocols across institutions. Ultimately, a holistic perspective is critical for mitigating the high risk of debilitating strokes in this vulnerable patient population.
Collapse
Affiliation(s)
- Faisal Hakami
- Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Essam Alhazmi
- Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Wafa M Busayli
- Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | | | | | | | - Alyaj Hakami
- Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Renad A Othman
- Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | - Amal I Moafa
- Medicine, Faculty of Medicine, Jazan University, Jazan, SAU
| | | | - Mohammed Ali Madkhali
- Internal Medicine, and Hematology and Oncology, Faculty of Medicine, Jazan University, Jazan, SAU
| |
Collapse
|
10
|
Silva M, Faustino P. From Stress to Sick(le) and Back Again-Oxidative/Antioxidant Mechanisms, Genetic Modulation, and Cerebrovascular Disease in Children with Sickle Cell Anemia. Antioxidants (Basel) 2023; 12:1977. [PMID: 38001830 PMCID: PMC10669666 DOI: 10.3390/antiox12111977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Sickle cell anemia (SCA) is a genetic disease caused by the homozygosity of the HBB:c.20A>T mutation, which results in the production of hemoglobin S (HbS). In hypoxic conditions, HbS suffers autoxidation and polymerizes inside red blood cells, altering their morphology into a sickle shape, with increased rigidity and fragility. This triggers complex pathophysiological mechanisms, including inflammation, cell adhesion, oxidative stress, and vaso-occlusion, along with metabolic alterations and endocrine complications. SCA is phenotypically heterogeneous due to the modulation of both environmental and genetic factors. Pediatric cerebrovascular disease (CVD), namely ischemic stroke and silent cerebral infarctions, is one of the most impactful manifestations. In this review, we highlight the role of oxidative stress in the pathophysiology of pediatric CVD. Since oxidative stress is an interdependent mechanism in vasculopathy, occurring alongside (or as result of) endothelial dysfunction, cell adhesion, inflammation, chronic hemolysis, ischemia-reperfusion injury, and vaso-occlusion, a brief overview of the main mechanisms involved is included. Moreover, the genetic modulation of CVD in SCA is discussed. The knowledge of the intricate network of altered mechanisms in SCA, and how it is affected by different genetic factors, is fundamental for the identification of potential therapeutic targets, drug development, and patient-specific treatment alternatives.
Collapse
Affiliation(s)
- Marisa Silva
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge (INSA), Av. Padre Cruz, 1649-016 Lisboa, Portugal;
| | - Paula Faustino
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge (INSA), Av. Padre Cruz, 1649-016 Lisboa, Portugal;
- Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental (ISAMB), Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
- Laboratório Associado TERRA, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
11
|
An R, Man Y, Cheng K, Zhang T, Chen C, Wang F, Abdulla F, Kucukal E, Wulftange WJ, Goreke U, Bode A, Nayak LV, Vercellotti GM, Belcher JD, Little JA, Gurkan UA. Sickle red blood cell-derived extracellular vesicles activate endothelial cells and enhance sickle red cell adhesion mediated by von Willebrand factor. Br J Haematol 2023; 201:552-563. [PMID: 36604837 PMCID: PMC10121869 DOI: 10.1111/bjh.18616] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/14/2022] [Accepted: 12/06/2022] [Indexed: 01/07/2023]
Abstract
Endothelial activation and sickle red blood cell (RBC) adhesion are central to the pathogenesis of sickle cell disease (SCD). Quantitatively, RBC-derived extracellular vesicles (REVs) are more abundant from SS RBCs compared with healthy RBCs (AA RBCs). Sickle RBC-derived REVs (SS REVs) are known to promote endothelial cell (EC) activation through cell signalling and transcriptional regulation at longer terms. However, the SS REV-mediated short-term non-transcriptional response of EC is unclear. Here, we examined the impact of SS REVs on acute microvascular EC activation and RBC adhesion at 2 h. Compared with AA REVs, SS REVs promoted human pulmonary microvascular ECs (HPMEC) activation indicated by increased von Willebrand factor (VWF) expression. Under microfluidic conditions, we found abnormal SS RBC adhesion to HPMECs exposed to SS REVs. This enhanced SS RBC adhesion was reduced by haeme binding protein haemopexin or VWF cleaving protease ADAMTS13 to a level similar to HPMECs treated with AA REVs. Consistent with these observations, haemin- or SS REV-induced microvascular stasis in SS mice with implanted dorsal skin-fold chambers that was inhibited by ADAMTS13. The adhesion induced by SS REVs was variable and was higher with SS RBCs from patients with increased markers of haemolysis (lactate dehydrogenase and reticulocyte count) or a concomitant clinical diagnosis of deep vein thrombosis. Our results emphasise the critical contribution made by REVs to the pathophysiology of SCD by triggering acute microvascular EC activation and abnormal RBC adhesion. These findings may help to better understand acute pathophysiological mechanism of SCD and thereby the development of new treatment strategies using VWF as a potential target.
Collapse
Affiliation(s)
- Ran An
- Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, OH, USA
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
- Department of Biomedical Sciences, University of Houston, Houston, TX, USA
- indicates equal contribution
| | - Yuncheng Man
- Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, OH, USA
- indicates equal contribution
| | - Kevin Cheng
- Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, OH, USA
| | - Tianyi Zhang
- Physiology and Biophysics Department, Case Western Reserve University, Cleveland, OH, USA
| | - Chunsheng Chen
- Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| | - Fang Wang
- Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, OH, USA
| | - Fuad Abdulla
- Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| | - Erdem Kucukal
- Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, OH, USA
| | - William J. Wulftange
- Biomedical Engineering Department, Case Western Reserve University, Cleveland, OH, USA
| | - Utku Goreke
- Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, OH, USA
| | - Allison Bode
- Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, OH, USA
- Department of Hematology and Oncology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Lalitha V. Nayak
- Department of Hematology and Oncology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Gregory M. Vercellotti
- Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| | - John D. Belcher
- Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| | - Jane A. Little
- Divison of Hematology & UNC Blood Research Center, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Umut A. Gurkan
- Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, OH, USA
- Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
12
|
Abstract
While neutrophils are the main effectors of protective innate immune responses, they are also key players in inflammatory pathologies. Sickle cell disease (SCD) is a genetic blood disorder in which red blood cells (RBCs) are constantly destroyed in the circulation which generates a highly inflammatory environment that culminates in vascular occlusions. Vaso-occlusion is the hallmark of SCD and a predictor of disease severity. Neutrophils initiate and propagate SCD-related vaso-occlusion through adhesive interactions with the activated and dysfunctional endothelium, sickle RBCs, and platelets, leading to acute and chronic complications that progress to irreversible organ damage and ultimately death. The use of SCD humanized mouse models, in combination with in vivo imaging techniques, has emerged as a fundamental tool to understand the dynamics of neutrophils under complex inflammatory contexts and their contribution to vascular injury in SCD. In this review, we discuss the various mechanisms by which circulating neutrophils sense and respond to the wide range of stimuli present in the blood of SCD patients and mice. We argue that the central role of neutrophils in SCD can be rationalized to develop targets for the management of clinical complications in SCD patients.
Collapse
Affiliation(s)
- Lidiane S Torres
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Andrés Hidalgo
- Area of Cell and Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
13
|
Agouti I, Masson E, Loundou A, Jean E, Arnaud L, Abdili E, Berenger P, Lavoipierre V, Séguier J, Dignat-George F, Lacroix R, Bernit E. Plasma levels of E-selectin are associated with retinopathy in sickle cell disease. Eur J Haematol 2023; 110:271-279. [PMID: 36409296 PMCID: PMC10100354 DOI: 10.1111/ejh.13902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND The vascular endothelium is markedly disrupted in sickle cell disease (SCD) and is the converging cascade of the complex pathophysiologic processes linked to sickle cell vasculopathy. Circulating endothelial activation and/or apoptotic markers may reflect this endothelial activation/damage that contributes to the pathophysiology of the SCD vascular complications. METHODS Plasmatic levels of circulating endothelial cells (CECs), E-selectin, progenitor's endothelial cells (EPCs), and circulating extracellular vesicles (EVs) were evaluated in 50 SCD patients, 16 with vasculopathy. The association between these markers and the occurrence of disease-related microvascular injuries of the eye (retinopathy), kidney (nephropathy), and skin (chronic active ulcers) was explored. RESULTS Among the endothelial activation markers studied, only higher plasma levels of E-selectin were found in SCD patients with vasculopathy (p = .015). Increased E-selectin levels were associated with retinopathy (p < .001) but not with nephropathy or leg ulcers. All patients, at steady state, with or without vasculopathy, did not display a high count of CEC and EPC, markers of endothelial injury and repair. We did not show any significant differences in EVs levels between vasculopathy and not vasculopathy SCD patients. CONCLUSIONS Further studies will be required to determine whether the E-selectin could be used as an early biomarker of retinopathy sickle cell development.
Collapse
Affiliation(s)
- Imane Agouti
- Centre de référence des syndromes drépanocytaires majeurs, thalassémies et autres pathologies rare du globule rouge et de l'érythropoïèse, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Elodie Masson
- Département de médecine interne, Hôpital de la Timone, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Anderson Loundou
- Centre d'Etudes et de Recherche sur les services de santé et la qualité de vie. Unité de recherche EA 3279. Faculté de médecine, université Aix Marseille, Marseille, France
| | - Estelle Jean
- Centre de référence des syndromes drépanocytaires majeurs, thalassémies et autres pathologies rare du globule rouge et de l'érythropoïèse, Assistance Publique des Hôpitaux de Marseille, Marseille, France.,Département de médecine interne, Hôpital de la Timone, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Laurent Arnaud
- Département d'Hématologie et de Biologie vasculaire. Biogénopôle, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Evelyne Abdili
- Département d'Hématologie et de Biologie vasculaire. Biogénopôle, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Patricia Berenger
- Département d'Hématologie et de Biologie vasculaire. Biogénopôle, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Virginie Lavoipierre
- Département de médecine interne, Hôpital de la Timone, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Julie Séguier
- Centre de référence des syndromes drépanocytaires majeurs, thalassémies et autres pathologies rare du globule rouge et de l'érythropoïèse, Assistance Publique des Hôpitaux de Marseille, Marseille, France.,Département de médecine interne, Hôpital de la Timone, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Françoise Dignat-George
- Département d'Hématologie et de Biologie vasculaire. Biogénopôle, Assistance Publique des Hôpitaux de Marseille, Marseille, France.,C2VN, INSERM, INRAE, université Aix Marseille, Marseille, France
| | - Romaric Lacroix
- Département d'Hématologie et de Biologie vasculaire. Biogénopôle, Assistance Publique des Hôpitaux de Marseille, Marseille, France.,C2VN, INSERM, INRAE, université Aix Marseille, Marseille, France
| | - Emmanuelle Bernit
- Centre de référence des syndromes drépanocytaires majeurs, thalassémies et autres pathologies rare du globule rouge et de l'érythropoïèse, Assistance Publique des Hôpitaux de Marseille, Marseille, France.,Unité transversale de la drépanocytose, centre de référence des syndromes drépanocytaires majeurs, thalassémies et autres pathologies rare du globule rouge et de l'érythropoïèse, CHU de la Guadeloupe, Guadeloupe, France
| |
Collapse
|
14
|
Abi Rached NM, Gbotosho OT, Archer DR, Jones JA, Sterling MS, Hyacinth HI. Adhesion molecules and cerebral microvascular hemodynamic abnormalities in sickle cell disease. Front Neurol 2022; 13:976063. [PMID: 36570439 PMCID: PMC9767957 DOI: 10.3389/fneur.2022.976063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Cerebrovascular abnormalities are a common feature of sickle cell disease that may be associated with risk of vaso-occlusive pain crises, microinfarcts, and cognitive impairment. An activated endothelium and adhesion factors, VCAM-1 and P-selectin, are implicated in sickle cell vasculopathy, including abnormal hemodynamics and leukocyte adherence. This study examined the association between cerebral expression of these adhesion factors and cortical microvascular blood flow dynamics by using in-vivo two-photon microscopy. We also examined the impact of blood transfusion treatment on these markers of vasculopathy. Results showed that sickle cell mice had significantly higher maximum red blood cell (RBC) velocity (6.80 ± 0.25 mm/sec, p ≤ 0.01 vs. 5.35 ± 0.35 mm/sec) and more frequent blood flow reversals (18.04 ± 0.95 /min, p ≤ 0.01 vs. 13.59 ± 1.40 /min) in the cortical microvasculature compared to controls. In addition, sickle cell mice had a 2.6-fold (RFU/mm2) increase in expression of VCAM-1 and 17-fold (RFU/mm2) increase in expression of P-selectin compared to controls. This was accompanied by an increased frequency in leukocyte adherence (4.83 ± 0.57 /100 μm/min vs. 2.26 ± 0.37 /100 μm/min, p ≤ 0.001). We also found that microinfarcts identified in sickle cell mice were 50% larger than in controls. After blood transfusion, many of these parameters improved, as results demonstrated that sickle cell mice had a lower post-transfusion maximum RBC velocity (8.30 ± 0.98 mm/sec vs. 11.29 ± 0.95 mm/sec), lower frequency of blood flow reversals (12.80 ± 2.76 /min vs. 27.75 ± 2.09 /min), and fewer instances of leukocyte adherence compared to their pre-transfusion imaging time point (1.35 ± 0.32 /100 μm/min vs. 3.46 ± 0.58 /100 μm/min). Additionally, we found that blood transfusion was associated with lower expression of adhesion factors. Our results suggest that blood transfusion and adhesion factors, VCAM-1 and P-selectin, are potential therapeutic targets for addressing cerebrovascular pathology, such as vaso-occlusion, in sickle cell disease.
Collapse
Affiliation(s)
- Noor Mary Abi Rached
- Neuroscience and Behavioral Biology Undergraduate Program, Emory University, Atlanta, GA, United States
| | - Oluwabukola T. Gbotosho
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - David R. Archer
- Aflac Cancer and Blood Disorders Center, Emory University Department of Pediatrics and Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Jayre A. Jones
- Aflac Cancer and Blood Disorders Center, Emory University Department of Pediatrics and Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Morgan S. Sterling
- Aflac Cancer and Blood Disorders Center, Emory University Department of Pediatrics and Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Hyacinth I. Hyacinth
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
15
|
The oral ferroportin inhibitor vamifeport improves hemodynamics in a mouse model of sickle cell disease. Blood 2022; 140:769-781. [PMID: 35714304 PMCID: PMC9389634 DOI: 10.1182/blood.2021014716] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 05/20/2022] [Indexed: 12/02/2022] Open
Abstract
Sickle cell disease (SCD) is an inherited hemolytic anemia caused by a single point mutation in the β-globin gene of hemoglobin that leads to synthesis of sickle hemoglobin (HbS) in red blood cells (RBCs). HbS polymerizes in hypoxic conditions, leading to intravascular hemolysis, release of free hemoglobin and heme, and increased adhesion of blood cells to the endothelial vasculature, which causes painful vaso-occlusion and organ damage. HbS polymerization kinetics are strongly dependent on the intracellular HbS concentration; a relatively small reduction in cellular HbS concentration may prevent HbS polymerization and its sequelae. We hypothesized that iron restriction via blocking ferroportin, the unique iron transporter in mammals, might reduce HbS concentration in RBCs, thereby decreasing hemolysis, improving blood flow, and preventing vaso-occlusive events. Indeed, vamifeport (also known as VIT-2763), a clinical-stage oral ferroportin inhibitor, reduced hemolysis markers in the Townes model of SCD. The RBC indices of vamifeport-treated male and female Townes mice exhibited changes attributable to iron-restricted erythropoiesis: decreased corpuscular hemoglobin concentration mean and mean corpuscular volume, as well as increased hypochromic and microcytic RBC fractions. Furthermore, vamifeport reduced plasma soluble VCAM-1 concentrations, which suggests lowered vascular inflammation. Accordingly, intravital video microscopy of fluorescently labeled blood cells in the microvasculature of Townes mice treated with vamifeport revealed diminished adhesion to the endothelium and improved hemodynamics. These preclinical data provide a strong proof-of-concept for vamifeport in the Townes model of SCD and support further development of this compound as a potential novel therapy in SCD.
Collapse
|
16
|
Hemin-Induced Endothelial Dysfunction and Endothelial to Mesenchymal Transition in the Pathogenesis of Pulmonary Hypertension Due to Chronic Hemolysis. Int J Mol Sci 2022; 23:ijms23094763. [PMID: 35563154 PMCID: PMC9104708 DOI: 10.3390/ijms23094763] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 01/08/2023] Open
Abstract
Pulmonary hypertension in sickle cell disease is an independent predictor of mortality, yet the pathogenesis of pulmonary vascular disease in chronic hemolytic disorders remains incompletely understood and treatment options are limited primarily to supportive care. The release of extracellular hemoglobin has been implicated in the development of pulmonary hypertension, and in this study we explored the direct effects of hemin, the oxidized moiety of heme, on the pulmonary artery endothelium. We found that low dose hemin exposure leads to significantly increased endothelial cell proliferation, migration, and cytokine release as markers of endothelial dysfunction. Protein expression changes in our pulmonary artery endothelial cells showed upregulation of mesenchymal markers after hemin treatment in conjunction with a decrease in endothelial markers. Endothelial to mesenchymal transition (EndoMT) resulting from hemin exposure was further confirmed by showing upregulation of the transcription factors SNAI1 and SLUG, known to regulate EndoMT. Lastly, given the endothelial dysfunction and phenotypic transition observed, the endothelial cytoskeleton was considered a potential novel target. Inhibiting myosin light chain kinase, to prevent phosphorylation of myosin light chain and cytoskeletal contraction, attenuated hemin-induced endothelial hyper-proliferation, migration, and cytokine release. The findings in this study implicate hemin as a key inducer of endothelial dysfunction through EndoMT, which may play an important role in pulmonary vascular remodeling during the development of pulmonary hypertension in chronic hemolytic states.
Collapse
|
17
|
Kengne Fotsing CB, Pieme CA, Biapa Nya PC, Chedjou JP, Dabou S, Nguemeni C, Teto G, Mbacham WF, Gatsing D. Relation between haptoglobin polymorphism and oxidative stress status, lipid profile, and cardiovascular risk in sickle cell anemia patients. Health Sci Rep 2022; 5:e465. [PMID: 35229039 PMCID: PMC8865067 DOI: 10.1002/hsr2.465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE The haptoglobin (Hp) gene located on chromosome 16q22 exhibits a polymorphism that can impact its capacity to inhibit the deleterious oxidative activity of free hemoglobin. We aimed to determine the influence of Hp polymorphism on oxidative stress, lipid profile, and cardiovascular risk in Cameroonian sickle cell anemia patients (SCA patients). METHOD The Hp genotypes of 102 SCA patients (SS), 60 healthy individuals (AA), and 55 subjects with sickle cell trait (AS) were determined by allele-specific PCR, and the blood parameters were assessed using standard methods. RESULTS Hp2-2 genotype was significantly (P < .05) present in SS (54%) than in AS (42%) and AA (38%). Levels of catalase and cell reactive protein were higher, while levels of total antioxidant capacity, triglycerides, low-density lipoprotein cholestetol, blood pressure, Framingham score, and body mass index were lower in the SCA patients. These parameters appeared to be unrelated to the haptoglobin genotypes. SCA patients with Hp1-1 genotype presented a higher oxidative stress index (0.53 ± 0.31) than those with Hp2-1 (0.33 ± 0.18). Lipid profile and cardiovascular risk were not significantly different between various Hp genotypes in SCA patients. CONCLUSION Haptoglobin polymorphism did not affect lipid profile, cardiovascular risk, and oxidative stress status of SCA patients. Nevertheless, SCA patients with Hp1-1 genotype tended to be more prone to oxidative stress than those with Hp2-1.
Collapse
Affiliation(s)
- Christian Bernard Kengne Fotsing
- Research Unit of Microbiology and Antimicrobial Substances, Department of Biochemistry, Faculty of ScienceUniversity of DschangDschangCameroon
- Research Unit of Biochemistry of Medicinal Plants, Food Science and Nutrition, Department of Biochemistry, Faculty of ScienceUniversity of DschangDschangCameroon
| | - Constant Anatole Pieme
- Laboratory of Biochemistry, Department of Biochemistry and Physiological Science, Faculty of Medicine and Biomedical ScienceUniversity of Yaounde IYaoundeCameroon
| | - Prosper Cabral Biapa Nya
- Research Unit of Biochemistry of Medicinal Plants, Food Science and Nutrition, Department of Biochemistry, Faculty of ScienceUniversity of DschangDschangCameroon
| | - Jean Paul Chedjou
- Laboratory for Public Health Research Biotechnology, Department of BiochemistryUniversity of Yaounde IYaoundeCameroon
| | - Solange Dabou
- Research Unit of Microbiology and Antimicrobial Substances, Department of Biochemistry, Faculty of ScienceUniversity of DschangDschangCameroon
| | - Carine Nguemeni
- Department of NeurologyUniversity Hospital of WürzburgWürzburgGermany
| | - Georges Teto
- Centre International de Recherche Chantal BiyaYaoundeCameroon
| | - Wilfred Fon Mbacham
- Laboratory for Public Health Research Biotechnology, Department of BiochemistryUniversity of Yaounde IYaoundeCameroon
| | - Donatien Gatsing
- Research Unit of Microbiology and Antimicrobial Substances, Department of Biochemistry, Faculty of ScienceUniversity of DschangDschangCameroon
| |
Collapse
|
18
|
de Azevedo JTC, Costa TCDM, Lima KC, Maciel TT, Palma PVB, Darrigo-Júnior LG, Setanni Grecco CE, Stracieri ABPL, Elias JB, Pieroni F, Guerino-Cunha RL, Pinto ACS, De Santis GC, Covas DT, Hermine O, Simões BP, Oliveira MC, Malmegrim KCR. Long-Term Effects of Allogeneic Hematopoietic Stem Cell Transplantation on Systemic Inflammation in Sickle Cell Disease Patients. Front Immunol 2021; 12:774442. [PMID: 34956203 PMCID: PMC8696202 DOI: 10.3389/fimmu.2021.774442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/08/2021] [Indexed: 11/26/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only currently available curative treatment for sickle cell disease (SCD). However, the effects of HSCT on SCD pathophysiology are poorly elucidated. Here, we assessed red blood cell (RBC) adhesiveness, intensity of hemolysis, vascular tone markers and systemic inflammation, in SCD patients treated with allogeneic HSCT. Thirty-two SCD patients were evaluated before and on long-term follow-up after HSCT. Overall survival was 94% with no severe (grade III-IV) graft-vs-host disease and a 22% rejection rate (graft failure). Hematological parameters, reticulocyte counts, and levels of lactate dehydrogenase (LDH), endothelin-1 and VCAM-1 normalized in SCD patients post-HSCT. Expression of adhesion molecules on reticulocytes and RBC was lower in patients with sustained engraftment. Levels of IL-18, IL-15 and LDH were higher in patients that developed graft failure. Increased levels of plasma pro-inflammatory cytokines, mainly TNF-α, were found in SCD patients long-term after transplantation. SCD patients with sustained engraftment after allo-HSCT showed decreased reticulocyte counts and adhesiveness, diminished hemolysis, and lower levels of vascular tonus markers. Nevertheless, systemic inflammation persists for at least five years after transplantation, indicating that allo-HSCT does not equally affect all aspects of SCD pathophysiology.
Collapse
Affiliation(s)
- Júlia Teixeira Cottas de Azevedo
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Graduate Program in Basic and Applied Immunology of the Ribeirão Preto Medicinal School, University of São Paulo, Ribeirão Preto, Brazil
| | - Thalita Cristina de Mello Costa
- Bone Marrow Transplantation and Cellular Therapy Unit, University Hospital, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Keli Cristina Lima
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Graduate Program in Bioscience and Biotechnology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Thiago Trovati Maciel
- Institut national de la santé et de la recherche médicale (INSERM) Unité mixte de recherche (UMR) 1163, Centre national de la recherche scientifique (CNRS) Equipe de Recherche Labellisée (ERL) 8254, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutical Implications, Imagine Institute, Paris, France.,Imagine Institute, Université Paris Descartes, Sorbonne Paris-Cité et Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Patrícia Vianna Bonini Palma
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Luiz Guilherme Darrigo-Júnior
- Bone Marrow Transplantation and Cellular Therapy Unit, University Hospital, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Ana Beatriz P L Stracieri
- Bone Marrow Transplantation and Cellular Therapy Unit, University Hospital, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Juliana Bernardes Elias
- Bone Marrow Transplantation and Cellular Therapy Unit, University Hospital, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fabiano Pieroni
- Bone Marrow Transplantation and Cellular Therapy Unit, University Hospital, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Renato Luiz Guerino-Cunha
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Ana Cristina Silva Pinto
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gil Cunha De Santis
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Dimas Tadeu Covas
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Olivier Hermine
- Institut national de la santé et de la recherche médicale (INSERM) Unité mixte de recherche (UMR) 1163, Centre national de la recherche scientifique (CNRS) Equipe de Recherche Labellisée (ERL) 8254, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutical Implications, Imagine Institute, Paris, France.,Imagine Institute, Université Paris Descartes, Sorbonne Paris-Cité et Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Belinda Pinto Simões
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maria Carolina Oliveira
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Internal Medicine, Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Kelen Cristina Ribeiro Malmegrim
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
19
|
Characterization of a murine model of endothelial dysfunction induced by chronic intraperitoneal administration of angiotensin II. Sci Rep 2021; 11:21193. [PMID: 34707201 PMCID: PMC8551243 DOI: 10.1038/s41598-021-00676-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/14/2021] [Indexed: 02/05/2023] Open
Abstract
Endothelial dysfunction (ED) is a key factor for the development of cardiovascular diseases. Due to its chronic, life-threatening nature, ED only can be studied experimentally in animal models. Therefore, this work was aimed to characterize a murine model of ED induced by a daily intraperitoneal administration of angiotensin II (AGII) for 10 weeks. Oxidative stress, inflammation, vascular remodeling, hypertension, and damage to various target organs were evaluated in treated animals. The results indicated that a chronic intraperitoneal administration of AGII increases the production of systemic soluble VCAM, ROS and ICAM-1 expression, and the production of TNFα, IL1β, IL17A, IL4, TGFβ, and IL10 in the kidney, as well as blood pressure levels; it also promotes vascular remodeling and induces non-alcoholic fatty liver disease, glomerulosclerosis, and proliferative retinopathy. Therefore, the model herein proposed can be a representative model for ED; additionally, it is easy to implement, safe, rapid, and inexpensive.
Collapse
|
20
|
Wang Q, Zennadi R. The Role of RBC Oxidative Stress in Sickle Cell Disease: From the Molecular Basis to Pathologic Implications. Antioxidants (Basel) 2021; 10:antiox10101608. [PMID: 34679742 PMCID: PMC8533084 DOI: 10.3390/antiox10101608] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 01/14/2023] Open
Abstract
Sickle cell disease (SCD) is an inherited monogenic disorder and the most common severe hemoglobinopathy in the world. SCD is characterized by a point mutation in the β-globin gene, which results in hemoglobin (Hb) S production, leading to a variety of mechanistic and phenotypic changes within the sickle red blood cell (RBC). In SCD, the sickle RBCs are the root cause of the disease and they are a primary source of oxidative stress since sickle RBC redox state is compromised due to an imbalance between prooxidants and antioxidants. This imbalance in redox state is a result of a continuous production of reactive oxygen species (ROS) within the sickle RBC caused by the constant endogenous Hb autoxidation and NADPH oxidase activation, as well as by a deficiency in the antioxidant defense system. Accumulation of non-neutralized ROS within the sickle RBCs affects RBC membrane structure and function, leading to membrane integrity deficiency, low deformability, phosphatidylserine exposure, and release of micro-vesicles. These oxidative stress-associated RBC phenotypic modifications consequently evoke a myriad of physiological changes involved in multi-system manifestations. Thus, RBC oxidative stress in SCD can ultimately instigate major processes involved in organ damage. The critical role of the sickle RBC ROS production and its regulation in SCD pathophysiology are discussed here.
Collapse
|
21
|
Lance EI, Faulcon LM, Fu Z, Yang J, Whyte-Stewart D, Strouse JJ, Barron-Casella E, Jones K, Van Eyk JE, Casella JF, Everett AD. Proteomic discovery in sickle cell disease: Elevated neurogranin levels in children with sickle cell disease. Proteomics Clin Appl 2021; 15:e2100003. [PMID: 33915030 PMCID: PMC8666096 DOI: 10.1002/prca.202100003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/06/2021] [Accepted: 04/26/2021] [Indexed: 01/12/2023]
Abstract
PURPOSE Sickle cell disease (SCD) is an inherited hemoglobinopathy that causes stroke and silent cerebral infarct (SCI). Our aim was to identify markers of brain injury in SCD. EXPERIMENTAL DESIGN Plasma proteomes were analyzed using a sequential separation approach of hemoglobin (Hb) and top abundant plasma protein depletion, followed by reverse phase separation of intact proteins, trypsin digestion, and tandem mass spectrometry. We compared plasma proteomes of children with SCD with and without SCI in the Silent Cerebral Infarct Multi-Center Clinical Trial (SIT Trial) to age-matched, healthy non-SCD controls. RESULTS From the SCD group, 1172 proteins were identified. Twenty-five percent (289/1172) were solely in the SCI group. Twenty-five proteins with enriched expression in the human brain were identified in the SCD group. Neurogranin (NRGN) was the most abundant brain-enriched protein in plasma of children with SCD. Using a NRGN sandwich immunoassay and SIT Trial samples, median NRGN levels were higher at study entry in children with SCD (0.28 ng/mL, N = 100) compared to control participants (0.12 ng/mL, N = 25, p < 0.0004). CONCLUSIONS AND CLINICAL RELEVANCE NRGN levels are elevated in children with SCD. NRGN and other brain-enriched plasma proteins identified in plasma of children with SCD may provide biochemical evidence of neurological injury.
Collapse
Affiliation(s)
- Eboni I. Lance
- Department of Neurodevelopmental Medicine, Kennedy Krieger Institute, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Zongming Fu
- Division of Pediatric Hematology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jun Yang
- Division of Pediatric Cardiology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Donna Whyte-Stewart
- Division of Pediatric Hematology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John J. Strouse
- Division of Pediatric Hematology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Emily Barron-Casella
- Division of Pediatric Hematology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kimberly Jones
- Division of Pediatric Hematology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jennifer E. Van Eyk
- Division of Cardiology, Department of Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - James F. Casella
- Division of Pediatric Hematology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Allen D. Everett
- Division of Pediatric Cardiology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
22
|
Plasma Lipids and Lipoproteins in Sickle Cell Disease Patients in the Northern West Bank, Palestine. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6640956. [PMID: 34395622 PMCID: PMC8360734 DOI: 10.1155/2021/6640956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 02/02/2023]
Abstract
Background Lipid metabolism may be altered in red cell genetic disorders. The erythrocyte and plasma lipids are defected which may increase the risk of cardiovascular disease. In the present study, we hypothesized a possible link between severity of anemia and altered lipid profile in SCD. Methods A total of 151 SCD patients, including 62 patients with sickle cell anemia (SS), 54 patients with sickle β-thalassemia (ST), and 35 individuals with sickle cell trait (AS), were studied. The control group consisted of 160 healthy individuals. Total cholesterol (TC), triglyceride (TG), and high-density lipoprotein cholesterol (HDL-C) were enzymatically measured. Results Total cholesterol and LDL-C were significantly lower (P value < 0.001) in SS and ST patients compared to AS individuals and AA controls. However, LDL-C was significantly lower in AS individuals (both males and female) compared to AA controls (P value < 0.001). The HDL-C in SS and ST patients (both males and females) was significantly lower than that in AS individuals (P value < 0.001). In addition, the HDL-C was significantly higher in SS and ST males and AS (males and females) compared to AA controls (P value < 0.001). The HDL-C was also significantly higher in SS males (P value < 0.001) and females (P value < 0.05) compared to ST patients. The HDL-C was significantly higher in AS individuals (P value < 0.001) compared to AA controls. The triglycerides in SS males was significantly lower than that in ST patients (P value < 0.001), but there was no significant difference when compared to AS individuals and AA controls. In contrast, triglycerides in SS females were significantly lower than those in ST (P value < 0.05), AS (P value < 0.001), and AA controls (P value < 0.001). In males of ST patients, triglycerides were significantly higher than those observed in AS males and AA males (P value < 0.001). In contrast, females of ST patients have a significantly lower triglycerides compared to AS and AA females (P value < 0.001). Conclusions In SCD, the plasma is affected in some way, especially the plasma cholesterol that was investigated in this study. Further prospective studies should examine the contribution of an altered lipid profile to the severity and clinical complications in SCD patients.
Collapse
|
23
|
Suriany S, Xu I, Liu H, Ulker P, Fernandez GE, Sposto R, Borzage M, Wenby R, Meiselman HJ, Forman HJ, Coates TD, Detterich JA. Individual red blood cell nitric oxide production in sickle cell anemia: Nitric oxide production is increased and sickle shaped cells have unique morphologic change compared to discoid cells. Free Radic Biol Med 2021; 171:143-155. [PMID: 33974976 DOI: 10.1016/j.freeradbiomed.2021.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 10/21/2022]
Abstract
Sickle cell anemia (SCA) is characterized by decreased red blood cell (RBC) deformability due to polymerization of deoxygenated hemoglobin, leading to abnormal mechanical properties of RBC, increased cellular adhesion, and microcirculatory obstruction. Prior work has demonstrated that NO• influences RBC hydration and deformability and is produced at a basal rate that increases under shear stress in normal RBC. Nevertheless, the origin and physiological relevance of nitric oxide (NO•) production and scavenging in RBC remains unclear. We aimed to assess the basal and shear-mediated production of NO• in RBC from SCA patients and control (CTRL) subjects. RBCs loaded with a fluorescent NO• detector, DAF-FM (4-Amino-5-methylamino- 2',7'-difluorofluorescein diacetate), were imaged in microflow channels over 30-min without shear stress, followed by a 30-min period under 0.5Pa shear stress. We utilized non-specific nitric oxide synthase (NOS) blockade and carbon monoxide (CO) saturation of hemoglobin to assess the contribution of NOS and hemoglobin, respectively, to NO• production. Quantification of DAF-FM fluorescence intensity in individual RBC showed an increase in NO• in SCA RBC at the start of the basal period; however, both SCA and CTRL RBC increased NO• by a similar quantity under shear. A subpopulation of sickle-shaped RBC exhibited lower basal NO• production compared to discoid RBC from SCA group, and under shear became more circular in the direction of shear when compared to discoid RBC from SCA and CTRL, which elongated. Both CO and NOS inhibition caused a decrease in basal NO• production. Shear-mediated NO• production was decreased by CO in all RBC, but was decreased by NOS blockade only in SCA. In conclusion, total NO• production is increased and shear-mediated NO• production is preserved in SCA RBC in a NOS-dependent manner. Sickle shaped RBC with inclusions have higher NO• production and they become more circular rather than elongated with shear.
Collapse
Affiliation(s)
- Silvie Suriany
- Division of Cardiology, Children's Hospital of Los Angeles, USA
| | - Iris Xu
- Division of Hematology, Children's Hospital of Los Angeles, USA
| | - Honglei Liu
- Division of Cardiology, Children's Hospital of Los Angeles, USA
| | - Pinar Ulker
- Department of Physiology, Akdeniz University, Turkey
| | | | - Richard Sposto
- Division of Hematology, Children's Hospital of Los Angeles, USA
| | - Matthew Borzage
- Fetal and Neonatal Institute, Division of Neonatology Children's Hospital Los Angeles, USA
| | - Rosalinda Wenby
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, USA
| | - Herbert J Meiselman
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, USA
| | - Henry Jay Forman
- Leonard Davis School of Gerontology, University of Southern California, USA
| | - Thomas D Coates
- Division of Hematology, Children's Hospital of Los Angeles, USA
| | - Jon A Detterich
- Division of Cardiology, Children's Hospital of Los Angeles, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, USA.
| |
Collapse
|
24
|
Madany E, Okwan-Duodu D, Balbuena-Merle R, Hendrickson JE, Gibb DR. Potential Implications of a Type 1 Interferon Gene Signature on COVID-19 Severity and Chronic Inflammation in Sickle Cell Disease. Front Med (Lausanne) 2021; 8:679030. [PMID: 34368185 PMCID: PMC8339405 DOI: 10.3389/fmed.2021.679030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/25/2021] [Indexed: 11/18/2022] Open
Abstract
At the onset of the corona virus disease 19 (COVID-19) pandemic, there were concerns that patients with sickle cell disease (SCD) might be especially vulnerable to severe sequelae of SARS-CoV-2 infection. While two reports support this conclusion, multiple studies have reported unexpectedly favorable outcomes in patients with SCD. However, mechanisms explaining these disparate conclusions are lacking. Here, we review recent studies indicating that the majority of patients with SCD express elevated levels of anti-viral type 1 interferons (IFNα/β) and interferon stimulated genes, independent of COVID-19, during their baseline state of health. We also present our data from the pre-COVID-19 era, illustrating elevated expression of a well-characterized interferon stimulated gene in a cohort of patients with SCD, compared to race-matched controls. These type 1 interferons and interferon stimulated genes have the potential to contribute to the variable progression of COVID-19 and other viral infections in patients with SCD. While the majority of evidence supports a protective role, the role of IFNα/β in COVID-19 severity in the general population remains an area of current investigation. We conclude that type 1 interferon responses in patients with SCD may contribute to the variable COVID-19 responses reported in prior studies. Additional studies investigating the mechanisms underlying IFNα/β production and other clinical consequences of IFNα/β-mediated inflammation in SCD disease are warranted.
Collapse
Affiliation(s)
- Emaan Madany
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Derick Okwan-Duodu
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Raisa Balbuena-Merle
- Department of Laboratory Medicine, Yale New Haven Hospital, New Haven, CT, United States
| | - Jeanne E. Hendrickson
- Department of Laboratory Medicine, Yale New Haven Hospital, New Haven, CT, United States
| | - David R. Gibb
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
25
|
Longitudinal effect of disease-modifying therapy on tricuspid regurgitant velocity in children with sickle cell anemia. Blood Adv 2021; 5:89-98. [PMID: 33570630 DOI: 10.1182/bloodadvances.2020003197] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/09/2020] [Indexed: 01/26/2023] Open
Abstract
Elevated tricuspid regurgitant velocity (TRV) ≥2.5 m/s is a predictor of disease severity in adults and children with sickle cell anemia (SCA), but how disease-modifying therapies (DMTs) affect this biomarker is incompletely understood. We investigated the effect of DMTs on TRV elevation in children. In a prospective single-center study, 204 subjects with HbSS or HbSβ0 thalassemia (mean age, 10.6 years; range, 5-18) had echocardiograms with assessment of TRV, with repeat evaluations after 2 years of observation. One-hundred and twelve participants received DMTs (hydroxyurea, n = 72; monthly erythrocyte transfusions, n = 40), 58 did not receive any DMT, and 34 were begun on hydroxyurea during this observation period. In the entire cohort, an increase in hemoglobin of 1.0 g/dL was associated with a 0.03-m/s decrease in TRV (P = .024), and a decrease in absolute reticulocyte count of 1.0 × 106/mL was associated with a 0.34-m/s decrease in TRV (P = .034). Compared with baseline, hydroxyurea exposure (continuous or newly started) was associated with an average 5% decline in mean TRV at the 2-year evaluation. Among participants newly started on hydroxyurea (mean treatment duration 1.2 ± 0.6 years), an increase in hemoglobin of 1.0 g/dL was associated with a 0.06-m/s decrease in TRV (P = .05). We conclude that hydroxyurea therapy may mitigate TRV elevation in children with SCA, possibly as a result of a reduction in hemolysis and improvement in anemia.
Collapse
|
26
|
PPARγ increases HUWE1 to attenuate NF-κB/p65 and sickle cell disease with pulmonary hypertension. Blood Adv 2021; 5:399-413. [PMID: 33496741 DOI: 10.1182/bloodadvances.2020002754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 12/07/2020] [Indexed: 12/17/2022] Open
Abstract
Sickle cell disease (SCD)-associated pulmonary hypertension (PH) causes significant morbidity and mortality. Here, we defined the role of endothelial specific peroxisome proliferator-activated receptor γ (PPARγ) function and novel PPARγ/HUWE1/miR-98 signaling pathways in the pathogenesis of SCD-PH. PH and right ventricular hypertrophy (RVH) were increased in chimeric Townes humanized sickle cell (SS) mice with endothelial-targeted PPARγ knockout (SSePPARγKO) compared with chimeric littermate control (SSLitCon). Lung levels of PPARγ, HUWE1, and miR-98 were reduced in SSePPARγKO mice compared with SSLitCon mice, whereas SSePPARγKO lungs were characterized by increased levels of p65, ET-1, and VCAM1. Collectively, these findings indicate that loss of endothelial PPARγ is sufficient to increase ET-1 and VCAM1 that contribute to endothelial dysfunction and SCD-PH pathogenesis. Levels of HUWE1 and miR-98 were decreased, and p65 levels were increased in the lungs of SS mice in vivo and in hemin-treated human pulmonary artery endothelial cells (HPAECs) in vitro. Although silencing of p65 does not regulate HUWE1 levels, the loss of HUWE1 increased p65 levels in HPAECs. Overexpression of PPARγ attenuated hemin-induced reductions of HUWE1 and miR-98 and increases in p65 and endothelial dysfunction. Similarly, PPARγ activation attenuated baseline PH and RVH and increased HUWE1 and miR-98 in SS lungs. In vitro, hemin treatment reduced PPARγ, HUWE1, and miR-98 levels and increased p65 expression, HPAEC monocyte adhesion, and proliferation. These derangements were attenuated by pharmacological PPARγ activation. Targeting these signaling pathways can favorably modulate a spectrum of pathobiological responses in SCD-PH pathogenesis, highlighting novel therapeutic targets in SCD pulmonary vascular dysfunction and PH.
Collapse
|
27
|
Interplay of erythropoietin, fibroblast growth factor 23, and erythroferrone in patients with hereditary hemolytic anemia. Blood Adv 2021; 4:1678-1682. [PMID: 32324886 DOI: 10.1182/bloodadvances.2020001595] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/23/2020] [Indexed: 12/13/2022] Open
Abstract
Recently, erythropoietin (EPO) was identified as regulator of fibroblast growth factor 23 (FGF23). Proteolytic cleavage of biologically active intact FGF23 (iFGF23) results in the formation of C-terminal fragments (cFGF23). An increase in cFGF23 relative to iFGF23 suppresses FGF receptor signaling by competitive inhibition. EPO lowers the i:cFGF23 ratio, thereby overcoming iFGF23-mediated suppression of erythropoiesis. We investigated EPO-FGF23 signaling and levels of erythroferrone (ERFE) in 90 patients with hereditary hemolytic anemia (www.trialregister.nl [NL5189]). We show, for the first time, the importance of EPO-FGF23 signaling in hereditary hemolytic anemia: there was a clear correlation between total FGF23 and EPO levels (r = +0.64; 95% confidence interval [CI], 0.09-0.89), which persisted after adjustment for iron load, inflammation, and kidney function. There was no correlation between iFGF23 and EPO. Data are consistent with a low i:cFGF23 ratio. Therefore, as expected, we report a correlation between EPO and ERFE in a diverse set of hereditary hemolytic anemias (r = +0.47; 95% CI, 0.14-0.69). There was no association between ERFE and total FGF23 or iFGF23, which suggests that ERFE does not contribute to the connection between FGF23 and EPO. These findings open a new area of research and might provide potentially new druggable targets with the opportunity to ameliorate ineffective erythropoiesis and the development of disease complications in hereditary hemolytic anemias.
Collapse
|
28
|
Morikis VA, Hernandez AA, Magnani JL, Sperandio M, Simon SI. Targeting Neutrophil Adhesive Events to Address Vaso-Occlusive Crisis in Sickle Cell Patients. Front Immunol 2021; 12:663886. [PMID: 33995392 PMCID: PMC8113856 DOI: 10.3389/fimmu.2021.663886] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/29/2021] [Indexed: 11/13/2022] Open
Abstract
Neutrophils are essential to protect the host against invading pathogens but can promote disease progression in sickle cell disease (SCD) by becoming adherent to inflamed microvascular networks in peripheral tissue throughout the body. During the inflammatory response, leukocytes extravasate from the bloodstream using selectin adhesion molecules and migrate to sites of tissue insult through activation of integrins that are essential for combating pathogens. However, during vaso-occlusion associated with SCD, neutrophils are activated during tethering and rolling on selectins upregulated on activated endothelium that line blood vessels. Recently, we reported that recognition of sLex on L-selectin by E-selectin during neutrophil rolling initiates shear force resistant catch-bonds that facilitate tethering to endothelium and activation of integrin bond clusters that anchor cells to the vessel wall. Evidence indicates that blocking this important signaling cascade prevents the congestion and ischemia in microvasculature that occurs from neutrophil capture of sickled red blood cells, which are normally deformable ellipses that flow easily through small blood vessels. Two recently completed clinical trials of therapies targeting selectins and their effect on neutrophil activation in small blood vessels reveal the importance of mechanoregulation that in health is an immune adaption facilitating rapid and proportional leukocyte adhesion, while sustaining tissue perfusion. We provide a timely perspective on the mechanism underlying vaso-occlusive crisis (VOC) with a focus on new drugs that target selectin mediated integrin adhesive bond formation.
Collapse
Affiliation(s)
- Vasilios A. Morikis
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, United States
| | - Alfredo A. Hernandez
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, United States
| | | | - Markus Sperandio
- Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine Biomedical Center, Ludwig Maximilians University, Walter Brendel Center, Munich, Germany
| | - Scott I. Simon
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, United States
| |
Collapse
|
29
|
Tsou PY, Cielo CM, Xanthopoulos MS, Wang YH, Kuo PL, Tapia IE. The burden of obstructive sleep apnea in pediatric sickle cell disease: a Kids' inpatient database study. Sleep 2021; 44:5896596. [PMID: 32835382 DOI: 10.1093/sleep/zsaa157] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 08/10/2020] [Indexed: 11/13/2022] Open
Abstract
STUDY OBJECTIVES Obstructive sleep apnea (OSA) is associated with cardiovascular and cerebrovascular morbidity. Patients with sickle cell disease (SCD) are at increased risk for both neurologic complications (NC) and OSA. However, the relationship between OSA and SCD complications is unclear. We hypothesized that there would be an association between OSA diagnosis and SCD complications. METHODS Hospital discharge records of patients with SCD aged < 19 years were obtained for the years 1997, 2000, 2003, 2006, 2009, and 2012 from the Kid's Inpatient Database. The primary outcome, NC, a composite of stroke, transient ischemic attack, and seizures. Secondary outcomes included acute chest syndrome (ACS), vaso-occlusive crisis, length of hospital stay, and inflation-adjusted cost of hospitalization. Multivariable regression was conducted to ascertain the association of OSA with primary and secondary outcomes. Analyses were adjusted for the use of noninvasive mechanical ventilation (NIMV) to determine its role as NC risk modifier. RESULTS There were 203,705 SCD discharges included in the analysis, of which 2,820 (1.4%) and 4,447 (2.2%) also included OSA and NC diagnoses. Multivariable logistic regression indicated that OSA was associated with NC (adjusted odds ratio [OR], 1.50 [95% CI 1.02-2.21], p = 0.039) and ACS (OR, 1.34 [95% CI 1.08-1.67], p = 0.009) in children with SCD. In the multivariable analysis adjusted for NIMV, the significant association between OSA and NC was no longer observed (OR, 1.39 [95% CI 0.94-2.05], p = 0.100). CONCLUSIONS OSA is associated with a 50% increase of odds of NC in children with SCD in this nationwide dataset. The use of NIMV to treat OSA may modify the risk of OSA-associated NC.
Collapse
Affiliation(s)
- Po-Yang Tsou
- Department of Pediatrics, Driscoll Children's Hospital, Corpus Christi, TX.,Sleep Center, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Christopher M Cielo
- Sleep Center, Children's Hospital of Philadelphia, Philadelphia, PA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Yu-Hsun Wang
- Department of Pediatrics, Driscoll Children's Hospital, Corpus Christi, TX
| | - Pei-Lun Kuo
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Ignacio E Tapia
- Sleep Center, Children's Hospital of Philadelphia, Philadelphia, PA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
30
|
Vinchi F, Sparla R, Passos ST, Sharma R, Vance SZ, Zreid HS, Juaidi H, Manwani D, Yazdanbakhsh K, Nandi V, Silva AMN, Agarvas AR, Fibach E, Belcher JD, Vercellotti GM, Ghoti H, Muckenthaler MU. Vasculo-toxic and pro-inflammatory action of unbound haemoglobin, haem and iron in transfusion-dependent patients with haemolytic anaemias. Br J Haematol 2021; 193:637-658. [PMID: 33723861 PMCID: PMC8252605 DOI: 10.1111/bjh.17361] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
Increasing evidence suggests that free haem and iron exert vasculo‐toxic and pro‐inflammatory effects by activating endothelial and immune cells. In the present retrospective study, we compared serum samples from transfusion‐dependent patients with β‐thalassaemia major and intermedia, hereditary spherocytosis and sickle cell disease (SCD). Haemolysis, transfusions and ineffective erythropoiesis contribute to haem and iron overload in haemolytic patients. In all cohorts we observed increased systemic haem and iron levels associated with scavenger depletion and toxic ‘free’ species formation. Endothelial dysfunction, oxidative stress and inflammation markers were significantly increased compared to healthy donors. In multivariable logistic regression analysis, oxidative stress markers remained significantly associated with both haem‐ and iron‐related parameters, while soluble vascular cell adhesion molecule 1 (sVCAM‐1), soluble endothelial selectin (sE‐selectin) and tumour necrosis factor α (TNFα) showed the strongest association with haem‐related parameters and soluble intercellular adhesion molecule 1 (sICAM‐1), sVCAM‐1, interleukin 6 (IL‐6) and vascular endothelial growth factor (VEGF) with iron‐related parameters. While hereditary spherocytosis was associated with the highest IL‐6 and TNFα levels, β‐thalassaemia major showed limited inflammation compared to SCD. The sVCAM1 increase was significantly lower in patients with SCD receiving exchange compared to simple transfusions. The present results support the involvement of free haem/iron species in the pathogenesis of vascular dysfunction and sterile inflammation in haemolytic diseases, irrespective of the underlying haemolytic mechanism, and highlight the potential therapeutic benefit of iron/haem scavenging therapies in these conditions.
Collapse
Affiliation(s)
- Francesca Vinchi
- Iron Research Program, New York Blood Center, New York, NY, USA.,Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA.,Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory (EMBL), Heidelberg University, Heidelberg, Germany
| | - Richard Sparla
- Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University of Heidelberg, Heidelberg, Germany
| | - Sara T Passos
- Iron Research Program, New York Blood Center, New York, NY, USA
| | - Richa Sharma
- Iron Research Program, New York Blood Center, New York, NY, USA
| | - S Zebulon Vance
- Iron Research Program, New York Blood Center, New York, NY, USA
| | - Hala S Zreid
- Department of Internal Medicine, Al Shifa Hospital, Gaza, Palestine
| | - Hesham Juaidi
- Department of Internal Medicine, Al Shifa Hospital, Gaza, Palestine
| | - Deepa Manwani
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA.,Pediatric Hematology, The Children's Hospital at Montefiore, New York, NY, USA
| | | | - Vijay Nandi
- Laboratory of Data Analytic Services, New York Blood Center, New York, NY, USA
| | - André M N Silva
- REQUIMTE-LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências, University of Porto, Porto, Portugal
| | - Anand R Agarvas
- Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University of Heidelberg, Heidelberg, Germany
| | - Eitan Fibach
- Department of Hematology, The Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - John D Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| | - Gregory M Vercellotti
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| | - Husam Ghoti
- European Center for Cancer and Cell Therapy (ECCT), Nicosia, Cyprus
| | - Martina U Muckenthaler
- Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory (EMBL), Heidelberg University, Heidelberg, Germany.,Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, University of Heidelberg, Heidelberg, Germany.,German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg, Germany.,Translational Lung Research Center (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
31
|
Gbotosho OT, Taylor M, Malik P. Cardiac pathophysiology in sickle cell disease. J Thromb Thrombolysis 2021; 52:248-259. [PMID: 33677791 DOI: 10.1007/s11239-021-02414-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/16/2021] [Indexed: 11/24/2022]
Affiliation(s)
- Oluwabukola Temitope Gbotosho
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, Cincinnati, OH, USA
| | - Michael Taylor
- Division of Cardiology, Heart Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, Cincinnati, OH, USA
| | - Punam Malik
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, Cincinnati, OH, USA. .,Division of Hematology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, 3333 Burnet Ave, Cincinnati, OH, 45229, USA.
| |
Collapse
|
32
|
Topchieva LV, Korneva VA, Kurbatova IV. The relationship of the carriership of allelic variations in rs2228145 (A > C) of the IL6R gene with the levels of VCAM1 and ICAM1 gene transcripts in patients with essential hypertension. Vavilovskii Zhurnal Genet Selektsii 2021; 24:96-101. [PMID: 33659786 PMCID: PMC7716534 DOI: 10.18699/vj20.600] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The levels of plasma interleukin 6 and its soluble receptors were found to be elevated in subjects with cardiovascular diseases, which points to amplification of the IL-6-mediated trans-signaling pathway in cells and the development of chronic inflammation. The allelic variation in the rs2228145 IL6R gene is associated with a change in the contents of the soluble and membrane-bound receptor forms mediating the biological activity of IL-6. Cytokine IL-6 is involved in the development of endothelial dysfunction by regulating the expression of the VCAM1 and ICAM1 genes, encoding intercellular adhesion molecules. Prior to this work, no data on the association of essential arterial hypertension (EAH) with rs2228145 allelic variations of the IL6R gene have been reported. The aim of our work was to study the relationship of the carriership of rs2228145 (A > C) allelic variations with the development of EAH and the VCAM1 and ICAM1 transcript levels. We analyzed samples of DNA isolated from the whole blood of 148 healthy donors and 152 patients with EAH (stages I–II). The genotyping was performed by PCR-RFLP. The level of transcripts in peripheral blood leukocytes (PBL) was assessed by real-time PCR. Differences in the frequency distributions of rs2228145 (A > C) genotypes between the control group and the group of patients with EAH (χ2 = 9.303) were found. The frequency of the CC genotype in EAH patients was higher than in healthy people (0.191 and 0.095, respectively). The risk of EAH (I–II stages) development was shown to be 2.3 times higher in CC genotype carriers as compared to individuals with other genotypes (OR = 2.257, 95 % confidence interval 1.100–4.468).
The levels of VCAM1 and ICAM1 gene transcripts in PBL of patients with EAH were significantly higher than in healthy people. The level of ICAM1 gene transcripts was almost 4 times higher in patients with CC genotype. The Kruskal–Wallis analysis of variance revealed an effect of rs2228145 (A > C) genotype on the transcriptional activity of ICAM1, which argues for its role in the pathogenesis of endothelial dysfunction and essential hypertension.
Collapse
Affiliation(s)
- L V Topchieva
- Institute of Biology of the Karelian Research Centre of the Russian Academy of Sciences, Petrozavodsk, Russia
| | - V A Korneva
- Petrozavodsk State University, Petrozavodsk, Russia
| | - I V Kurbatova
- Institute of Biology of the Karelian Research Centre of the Russian Academy of Sciences, Petrozavodsk, Russia
| |
Collapse
|
33
|
Gbotosho OT, Kapetanaki MG, Kato GJ. The Worst Things in Life are Free: The Role of Free Heme in Sickle Cell Disease. Front Immunol 2021; 11:561917. [PMID: 33584641 PMCID: PMC7873693 DOI: 10.3389/fimmu.2020.561917] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 12/04/2020] [Indexed: 12/15/2022] Open
Abstract
Hemolysis is a pathological feature of several diseases of diverse etiology such as hereditary anemias, malaria, and sepsis. A major complication of hemolysis involves the release of large quantities of hemoglobin into the blood circulation and the subsequent generation of harmful metabolites like labile heme. Protective mechanisms like haptoglobin-hemoglobin and hemopexin-heme binding, and heme oxygenase-1 enzymatic degradation of heme limit the toxicity of the hemolysis-related molecules. The capacity of these protective systems is exceeded in hemolytic diseases, resulting in high residual levels of hemolysis products in the circulation, which pose a great oxidative and proinflammatory risk. Sickle cell disease (SCD) features a prominent hemolytic anemia which impacts the phenotypic variability and disease severity. Not only is circulating heme a potent oxidative molecule, but it can act as an erythrocytic danger-associated molecular pattern (eDAMP) molecule which contributes to a proinflammatory state, promoting sickle complications such as vaso-occlusion and acute lung injury. Exposure to extracellular heme in SCD can also augment the expression of placental growth factor (PlGF) and interleukin-6 (IL-6), with important consequences to enthothelin-1 (ET-1) secretion and pulmonary hypertension, and potentially the development of renal and cardiac dysfunction. This review focuses on heme-induced mechanisms that are implicated in disease pathways, mainly in SCD. A special emphasis is given to heme-induced PlGF and IL-6 related mechanisms and their role in SCD disease progression.
Collapse
Affiliation(s)
- Oluwabukola T. Gbotosho
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Maria G. Kapetanaki
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Gregory J. Kato
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
34
|
Adekile A. The Genetic and Clinical Significance of Fetal Hemoglobin Expression in Sickle Cell Disease. Med Princ Pract 2021; 30:201-211. [PMID: 32892201 PMCID: PMC8280415 DOI: 10.1159/000511342] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/03/2020] [Indexed: 01/19/2023] Open
Abstract
Sickle cell disease (SCD) is phenotypically heterogeneous. One major genetic modifying factor is the patient's fetal hemoglobin (HbF) level. The latter is determined by the patient's β-globin gene cluster haplotype and cis- and trans-acting single nucleotide polymorphisms (SNPs) at other distant quantitative trait loci (QTL). The Arab/India haplotype is associated with persistently high HbF levels and also a relatively mild phenotype. This haplotype carries the Xmn1 (C/T) SNP, rs7482144, in the HBG2 locus. The major identified trans-acting QTL contain SNPs residing in the BCL11A on chromosome 2 and the HMIP locus on chromosome 6. These collectively account for 15-30% of HbF expression in different world populations and in patients with SCD or β-thalassemia. Patients with SCD in Kuwait and Eastern Saudi Arabia uniformly carry the Arab/India haplotype, but despite this, the HbF and clinical phenotypes show considerable heterogeneity. Pain episodes and avascular necrosis of the femoral head are particularly common, but severe bacterial infections, stroke, priapism, and leg ulcers are uncommon. Moreover, the HbF modifiers appear to be different; the reported BCL11A and HMIP SNPs appear to play insignificant roles. There are probably novel modifiers to be discovered in this population. This review examines the common clinical phenotypes in Kuwaiti patients with elevated HbF and the available information on HbF modifiers. The response of the patients to hydroxyurea is discussed. The presentation of patients with other sickle compound heterozygotes (Sβthal and HbSD), vis-à-vis their HbF levels, is also addressed critically.
Collapse
Affiliation(s)
- Adekunle Adekile
- Department of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait,
| |
Collapse
|
35
|
Huang J, Zhang T, Zou X, Wu S, Zhu J. Panton-valentine leucocidin carrying Staphylococcus aureus causing necrotizing pneumonia inactivates the JAK/STAT signaling pathway and increases the expression of inflammatory cytokines. INFECTION GENETICS AND EVOLUTION 2020; 86:104582. [PMID: 33017689 DOI: 10.1016/j.meegid.2020.104582] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/13/2020] [Accepted: 10/01/2020] [Indexed: 11/17/2022]
Abstract
PURPOSE Methicillin-resistant Staphylococcus aureus (MRSA) carrying Panton-Valentine leukocidin, a pore-forming toxin, is a common cause of necrotizing pneumonia. However, the early pulmonary inflammatory response following PVL(+) MRSA infection is unknown. The purpose of this study was to use a murine model to determine the effect of PVL(+) MRSA on lung tissues and the expression of cytokines and JAK and STAT mRNA and protein. METHODS Mice were randomly divided into 3 groups and intra-nasally treated with PBS (control group), recombinant PVL (rPVL group), and PVL(+) MRSA (PVL group). At 24 and 48 h after inoculation, bronchoalveolar lavage fluid (BALF) was tested for cytokine levels, and lung tissues were tested for JAK and STAT mRNA and protein expression, and examined after hematoxylin and eosin staining. RESULTS Mice infected with the PVL(+) strain became ill, characterized by impaired mobility, hunched posture, ruffled fur, and labored breathing. Lung tissue exhibited tissue necrosis and hemorrhage. BALF levels of IL-8, TNF-α, IFN-γ, IL-12, sICAM-1, and sVCAM-1 were increased in the rPVL or PVL groups, while levels of IL-10 and IL-4 levels were similar among the groups. JAK1 and STAT1 mRNA expression and protein levels were increased in lung tissue from mice infected with PVL(+) MRSA and rPVL. CONCLUSIONS PVL is a significant S. aureus virulence factor, and upregulates the expression of proinflammatory cytokines but does not affect the expression of anti-inflammatory cytokines. The effect of PVL may be due to JAK/STAT pathway activation. Blockade of the JAK/STAT pathway may decrease the severity of PVL(+) MRSA pneumonia.
Collapse
Affiliation(s)
- Jing Huang
- Department of Pulmonary and Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, Guangdong, China.
| | - Tiantuo Zhang
- Department of Pulmonary and Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, Guangdong, China
| | - Xiaoling Zou
- Department of Pulmonary and Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, Guangdong, China
| | - Shaozhu Wu
- Department of Pulmonary and Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, Guangdong, China
| | - Jiaxin Zhu
- Department of Pulmonary and Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, Guangdong, China
| |
Collapse
|
36
|
Li J, Jeong SY, Xiong B, Tseng A, Mahon AB, Isaacman S, Gordeuk VR, Cho J. Repurposing pyridoxamine for therapeutic intervention of intravascular cell-cell interactions in mouse models of sickle cell disease. Haematologica 2020; 105:2407-2419. [PMID: 33054081 PMCID: PMC7556679 DOI: 10.3324/haematol.2019.226720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/29/2019] [Indexed: 12/25/2022] Open
Abstract
Adherent neutrophils on vascular endothelium positively contribute to cell-cell aggregation and vaso-occlusion in sickle cell disease. In the present study, we demonstrated that pyridoxamine, a derivative of vitamin B6, might be a therapeutic agent to alleviate intravascular cell-cell aggregation in sickle cell disease. Using real-time intravital microscopy, we found that one oral administration of pyridoxamine dose-dependently increased the rolling influx of neutrophils and reduced neutrophil adhesion to endothelial cells in cremaster microvessels of sickle cell disease mice challenged with hypoxia-reoxygenation. Short-term treatment also mitigated neutrophil-endothelial cell and neutrophil-platelet interactions in the microvessels and improved the survival of sickle cell disease mice challenged with tumor necrosis factor-α. The inhibitory effects of pyridoxamine on intravascular cell-cell interactions were potentiated by co-treatment with hydroxyurea. We observed that long-term (5.5 months) oral treatment with pyridoxamine significantly diminished the adhesive function of neutrophils and platelets and down-regulated the expression of E-selectin and intercellular adhesion molecule-1 on the vascular endothelium in tumor necrosis factor-α-challenged sickle cell disease mice. Ex vivo studies revealed that the surface amount of αMβ2 integrin was significantly decreased in stimulated neutrophils isolated from sickle cell disease mice treated with pyridoxamine-containing water. Studies using platelets and neutrophils from sickle cell disease mice and patients suggested that treatment with pyridoxamine reduced the activation state of platelets and neutrophils. These results suggest that pyridoxamine may be a novel therapeutic and a supplement to hydroxyurea to prevent and treat vaco-occlusion events in sickle cell disease.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Si-Yeon Jeong
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Bei Xiong
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Alan Tseng
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | | | | | - Victor R. Gordeuk
- Section of Hematology/Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
- Comprehensive Sickle Cell Center, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Jaehyung Cho
- Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| |
Collapse
|
37
|
Changes in Blood Profile from Steady State in Patients with Sickle Cell Anemia Admitted for Vaso-occlusive Crisis and Acute Chest Syndrome. Adv Hematol 2020; 2020:3656717. [PMID: 32908517 PMCID: PMC7468598 DOI: 10.1155/2020/3656717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/30/2020] [Accepted: 08/07/2020] [Indexed: 02/02/2023] Open
Abstract
Close to half of all patients with sickle cell disease (SCD) will have at least one episode of acute chest syndrome (ACS) during their lifetime. Multiple cells and molecules involved with the inflammatory cascade play a role in the development of ACS. We found that patients with SCD who developed ACS as a complication of a vaso-occlusive crisis (VOC) had a significant increase in leukocytes and decrease in platelets from their steady state when compared with a separate admission for VOC without ACS development. No significant change from steady state hemoglobin or reticulocyte count was noted between the two admissions. These results indicate that trending laboratory markers may be useful to predict patients at risk for ACS development.
Collapse
|
38
|
Decoding the role of SOD2 in sickle cell disease. Blood Adv 2020; 3:2679-2687. [PMID: 31506286 DOI: 10.1182/bloodadvances.2019000527] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022] Open
Abstract
Sickle cell disease (SCD) is an inherited hemoglobinopathy caused by a single point mutation in the β-globin gene. As a consequence, deoxygenated hemoglobin polymerizes triggering red blood cell sickling and hemolysis, vaso-occlusion, and ischemia/reperfusion. Allied to these pathologies is the overproduction of reactive oxygen species driven by hemoglobin Fenton chemistry and peroxidase reactions as well as by secondary activation of vascular oxidases, including NAD(P)H oxidase and xanthine oxidase. In addition, hypoxia, produced by sickle red blood cell occlusion, disrupts mitochondrial metabolism and generates excess superoxide through electron leak from the mitochondrial respiratory chain. Superoxide dismutase 2 (SOD2) is a mitochondrial-specific antioxidant enzyme that dismutates superoxide to hydrogen peroxide, which is then converted to water by catalase and glutathione peroxidase. In SCD, the antioxidant defense system is significantly diminished through decreased expression and activity levels of antioxidant enzymes, including superoxide dismutase, catalase, and glutathione peroxidase. From a translational perspective, genetic variants including a missense variant in SOD2 (valine to alanine at position 16) are present in 45% of people with African ancestry and are associated with increased sickle complications. While it is known that there is an imbalance between oxidative species and antioxidant defenses in SCD, much more investigation is warranted. This review summarizes our current understanding of antioxidant defense systems in SCD, particularly focused on SOD2, and provides insight into challenges and opportunities as the field moves forward.
Collapse
|
39
|
Karafin MS, Simpson P, Field JJ. Chronic Pain Does Not Impact Baseline Circulating Cytokine Levels in Adults with Sickle Cell Disease. Acta Haematol 2020; 144:111-116. [PMID: 32403100 DOI: 10.1159/000507563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/29/2020] [Indexed: 11/19/2022]
Abstract
Chronic pain affects 50% of adults with sickle cell disease (SCD). Although inflammation is thought to contribute to the pathogenesis of chronic pain, no studies have examined the differences in circulating cytokines between patients with SCD with and without chronic pain. We performed an observational cohort study using blood and urine samples from adults with SCD with and without chronic pain at their usual state of health. We tested the hypothesis that, compared to those without chronic pain, those with chronic pain would have significantly higher baseline circulating proinflammatory cytokines. A total of 61 adults with SCD, 40 with chronic pain and 21 without chronic pain were tested. When SCD patients with chronic pain were compared to those without chronic pain, no significant differences in cytokine levels were noted. The variables most associated with the diagnosis of chronic pain in this population were opioid dose and subject age.
Collapse
Affiliation(s)
- Matthew S Karafin
- Medical Sciences Institute, Versiti, Milwaukee, Wisconsin, USA,
- Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA,
| | - Pippa Simpson
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Joshua J Field
- Medical Sciences Institute, Versiti, Milwaukee, Wisconsin, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
40
|
Abstract
INTRODUCTION In sickle cell disease (SCD), hemoglobin S (HbS) red blood cells (RBCs) are characteristically deformed and inflexible. Often breaking down in the circulation, they exhibit increased adhesive properties with the endothelium and activated neutrophils and platelets, increasing the risk of occlusion of the microcirculation. SCD is categorized into two sub-phenotypes: hyperhemolytic, associated with priapism, leg ulcers, pulmonary hypertension, and stroke, and high hemoglobin/viscosity, which may promote vaso-occlusion-associated pain, acute chest syndrome, and osteonecrosis. AREAS COVERED The sub-phenotypes are not completely distinct. Hemolysis may trigger vaso-occlusion, contributing to vascular complications. Targeting P-selectin, a key mediator of cross-talk between hyperhemolysis and vaso-occlusion, may be beneficial for vascular and vaso-occlusion-associated complications. English-language articles from PubMed on the topic of SCD and vaso-occlusive crises (VOCs) were reviewed from 1 January 2000 to 1 January 2019 using the search terms 'sickle cell disease,' 'vaso-occlusive crises,' and 'selectin.' EXPERT OPINION Besides targeting P-selectin, other strategies to counter VOCs and RBC sickling are being pursued. These include platelet inhibition to counter aggregation, intercellular adhesion, and thrombosis during VOCs; gene therapy to correct the homozygous missense mutation in the β-globin gene, causing polymerization of HbS; L-glutamine, possibly reducing oxidative stress in sickled RBCs; and fetal hemoglobin inducers.
Collapse
Affiliation(s)
- Solomon F Ofori-Acquah
- Department of Medicine, University of Pittsburgh , Pittsburgh, USA.,School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana , Accra, Ghana.,West African Genetic Medicine Centre (WAGMC), College of Health Sciences, University of Ghana , Accra, Ghana
| |
Collapse
|
41
|
Nader E, Romana M, Connes P. The Red Blood Cell-Inflammation Vicious Circle in Sickle Cell Disease. Front Immunol 2020; 11:454. [PMID: 32231672 PMCID: PMC7082402 DOI: 10.3389/fimmu.2020.00454] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/27/2020] [Indexed: 12/31/2022] Open
Abstract
Sickle cell disease (SCD) is a genetic disease caused by a single mutation in the β-globin gene, leading to the production of an abnormal hemoglobin called hemoglobin S (HbS), which polymerizes under deoxygenation, and induces the sickling of red blood cells (RBCs). Sickled RBCs are very fragile and rigid, and patients consequently become anemic and develop frequent and recurrent vaso-occlusive crises. However, it is now evident that SCD is not only a RBC rheological disease. Accumulating evidence shows that SCD is also characterized by the presence of chronic inflammation and oxidative stress, participating in the development of chronic vasculopathy and several chronic complications. The accumulation of hemoglobin and heme in the plasma, as a consequence of enhanced intravascular hemolysis, decreases nitric oxide bioavailability and enhances the production of reactive oxygen species (ROS). Heme and hemoglobin also represent erythrocytic danger-associated molecular pattern molecules (eDAMPs), which may activate endothelial inflammation through TLR-4 signaling and promote the development of complications, such as acute chest syndrome. It is also suspected that heme may activate the innate immune complement system and stimulate neutrophils to release neutrophil extracellular traps. A large amount of microparticles (MPs) from various cellular origins (platelets, RBCs, white blood cells, endothelial cells) is also released into the plasma of SCD patients and participate in the inflammation and oxidative stress in SCD. In turn, this pro-inflammatory and oxidative stress environment further alters the RBC properties. Increased pro-inflammatory cytokine concentrations promote the activation of RBC NADPH oxidase and, thus, raise the production of intra-erythrocyte ROS. Such enhanced oxidative stress causes deleterious damage to the RBC membrane and further alters the deformability of the cells, modifying their aggregation properties. These RBC rheological alterations have been shown to be associated to specific SCD complications, such as leg ulcers, priapism, and glomerulopathy. Moreover, RBCs positive for the Duffy antigen receptor for chemokines may be very sensitive to various inflammatory molecules that promote RBC dehydration and increase RBC adhesiveness to the vascular wall. In summary, SCD is characterized by a vicious circle between abnormal RBC rheology and inflammation, which modulates the clinical severity of patients.
Collapse
Affiliation(s)
- Elie Nader
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team Vascular Biology and Red Blood Cell, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| | - Marc Romana
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France.,Université des Antilles, UMR_S1134, BIGR, Pointe-à-Pitre, France.,Université de Paris, UMR_S1134, BIGR, INSERM, Paris, France
| | - Philippe Connes
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team Vascular Biology and Red Blood Cell, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| |
Collapse
|
42
|
Ochocinski D, Dalal M, Black LV, Carr S, Lew J, Sullivan K, Kissoon N. Life-Threatening Infectious Complications in Sickle Cell Disease: A Concise Narrative Review. Front Pediatr 2020; 8:38. [PMID: 32154192 PMCID: PMC7044152 DOI: 10.3389/fped.2020.00038] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/27/2020] [Indexed: 12/19/2022] Open
Abstract
Sickle cell disease (SCD) results in chronic hemolytic anemia, recurrent vascular occlusion, insidious vital organ deterioration, early mortality, and diminished quality of life. Life-threatening acute physiologic crises may occur on a background of progressive diminishing vital organ function. Sickle hemoglobin polymerizes in the deoxygenated state, resulting in erythrocyte membrane deformation, vascular occlusion, and hemolysis. Vascular occlusion and increased blood viscosity results in functional asplenia and immune deficiency in early childhood, resulting in life-long increased susceptibility to serious bacterial infections. Infection remains a main cause of overall mortality in patients with SCD in low- and middle-income countries due to increased exposure to pathogens, increased co-morbidities such as malnutrition, lower vaccination rates, and diminished access to definitive care, including antibiotics and blood. Thus, the greatest gains in preventing infection-associated mortality can be achieved by addressing these factors for SCD patients in austere environments. In contrast, in high-income countries, perinatal diagnosis of SCD, antimicrobial prophylaxis, vaccination, aggressive use of antibiotics for febrile episodes, and the availability of contemporary critical care resources have resulted in a significant reduction in deaths from infection; however, chronic organ injury is problematic. All clinicians, regardless of their discipline, who assume the care of SCD patients must understand the importance of infectious disease as a contributor to death and disability. In this concise narrative review, we summarize the data that describes the importance of infectious diseases as a contributor to death and disability in SCD and discuss pathophysiology, prevalent organisms, prevention, management of acute episodes of critical illness, and ongoing care.
Collapse
Affiliation(s)
- Dominik Ochocinski
- Department of Anesthesiology, University of Florida, Gainesville, FL, United States
| | - Mansi Dalal
- Division of Pediatric Hematology/Oncology, University of Florida, Gainesville, FL, United States
| | - L Vandy Black
- Division of Pediatric Hematology/Oncology, University of Florida, Gainesville, FL, United States
| | - Silvana Carr
- Division of Pediatric Infectious Disease, University of Florida, Gainesville, FL, United States
| | - Judy Lew
- Division of Pediatric Infectious Disease, University of Florida, Gainesville, FL, United States
| | - Kevin Sullivan
- Department of Anesthesiology, University of Florida, Gainesville, FL, United States.,Congenital Heart Center, University of Florida, Gainesville, FL, United States
| | - Niranjan Kissoon
- Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| |
Collapse
|
43
|
Matte A, Cappellini MD, Iolascon A, Enrica F, De Franceschi L. Emerging drugs in randomized controlled trials for sickle cell disease: are we on the brink of a new era in research and treatment? Expert Opin Investig Drugs 2019; 29:23-31. [PMID: 31847604 DOI: 10.1080/13543784.2020.1703947] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Sickle cell disease (SCD) is caused by a mutation in the HBB gene which is key for making a component of hemoglobin. The mutation leads to the formation of an abnormal hemoglobin molecule called sickle hemoglobin (HbS). SCD is a chronic, complex disease with a multiplicity of pathophysiological targets; it has high morbidity and mortality.Hydroxyurea has for many years been the only approved drug for SCD; hence, the development of new therapeutics is critical.Areas covered: This article offers an overview of the key studies of new therapeutic options for SCD. We searched the PubMed database and Cochrane Database of Systemic Reviews for agents in early phase clinic trials and preclinical development.Expert opinion: Although knowledge of SCD has progressed, patient survival and quality of life must be improved. Phase II and phase III clinical trials investigating pathophysiology-based novel agents show promising results in the clinical management of SCD acute events. The design of long-term clinical studies is necessary to fully understand the clinical impact of these new therapeutics on the natural history of the disease. Furthermore, the building of global collaborations will enhance the clinical management of SCD and the design of primary outcomes of future clinical trials.
Collapse
Affiliation(s)
- Alessandro Matte
- Department of Medicine, University of Verona and AOUI Verona, Policlinico GB Rossi, Verona, Italy
| | - Maria Domenica Cappellini
- Ca Granda Foundation IRCCS, Dept of Clinical Science and Community, University of Milan, Milan, Italy
| | - Achille Iolascon
- Dept of Chemical Sciences, University Federico II, Naples, Italy
| | - Federti Enrica
- Department of Medicine, University of Verona and AOUI Verona, Policlinico GB Rossi, Verona, Italy
| | - Lucia De Franceschi
- Department of Medicine, University of Verona and AOUI Verona, Policlinico GB Rossi, Verona, Italy
| |
Collapse
|
44
|
Alkindi S, Almufargi SS, Pathare A. Clinical and laboratory parameters, risk factors predisposing to the development of priapism in sickle cell patients. Exp Biol Med (Maywood) 2019; 245:79-83. [PMID: 31810382 DOI: 10.1177/1535370219892846] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Although sickle cell disease is very common in Oman, priapism is a relatively rare complication of this disease. This study was aimed to identify the clinical and laboratory risk factors that predispose sickle cell disease patients to priapism. In a retrospective, case-control study, data on 21 male sickle cell disease patients, with priapism, were compared to 20 age and sex-matched sickle cell disease patients without priapism from the hospital medical records. Specifically, the demographical, hematological, biochemical, and clinical parameters including complications attributable to priapism were studied. Means for continuous measures and independent t-test were used to evaluate the association between the parameter studied and the occurrence of priapism. Priapism occurrence was associated with low basal hemoglobin (Hb), along with an increase in other parameters such as white blood cell count (WBC) ( P = 0.010), platelet count ( P = 0.001), reticulocyte count ( P < 0.001), mean corpuscular volume (MCV) ( P < 0.001), and mean corpuscular hemoglobin (MCH) ( P < 0.001). Biochemical laboratory parameters revealed an association with an increased total bilirubin ( P < 0.001). Patients with priapism were more likely to have acute chest syndrome, stroke, pulmonary hypertension, asplenia ( P = 0.006), gallbladder stones, and consequently cholecystectomy. Blood exchange and Penile aspiration were the most frequent medical therapies ( P = 0.010 and P < 0.001 respectively). Patients with sickle cell disease who presented with priapism were young adults with low Hb, high WBC’s, platelets, reticulocytes, MCV, MCH, bilirubin, and LDH. These findings suggest an association of priapism with increased hemolysis. Impact statement Sickle cell disease is prevalent throughout the world with a 5.8% incidence of sickle cell trait among the Omani population, yet priapism was a relatively rare complication. Penile erection and detumescence are complex physiological processes, which require a delicate neurohormonal and cardiovascular response. In this cohort, young SCD patients developed priapism with a low incidence of complications. These patients had high WBCs, platelets, bilirubin, LDH, and reticulocytes with low hemoglobin as compared to controls, suggesting an increased hemolytic process. Favorable outcome was dependent on early intervention, with almost half of these patients needing surgical intervention.
Collapse
Affiliation(s)
- Salam Alkindi
- Department of Haematology, Sultan Qaboos University Hospital, Muscat PC123, Oman.,College of Medicine & Health Sciences, Muscat, PC 123, Oman
| | - Said S Almufargi
- Department of Haematology, Sultan Qaboos University Hospital, Muscat PC123, Oman
| | - Anil Pathare
- Department of Haematology, Sultan Qaboos University Hospital, Muscat PC123, Oman
| |
Collapse
|
45
|
Potoka KP, Wood KC, Baust JJ, Bueno M, Hahn SA, Vanderpool RR, Bachman T, Mallampalli GM, Osei-Hwedieh DO, Schrott V, Sun B, Bullock GC, Becker-Pelster EM, Wittwer M, Stampfuss J, Mathar I, Stasch JP, Truebel H, Sandner P, Mora AL, Straub AC, Gladwin MT. Nitric Oxide-Independent Soluble Guanylate Cyclase Activation Improves Vascular Function and Cardiac Remodeling in Sickle Cell Disease. Am J Respir Cell Mol Biol 2019; 58:636-647. [PMID: 29268036 DOI: 10.1165/rcmb.2017-0292oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Sickle cell disease (SCD) is associated with intravascular hemolysis and oxidative inhibition of nitric oxide (NO) signaling. BAY 54-6544 is a small-molecule activator of oxidized soluble guanylate cyclase (sGC), which, unlike endogenous NO and the sGC stimulator, BAY 41-8543, preferentially binds and activates heme-free, NO-insensitive sGC to restore enzymatic cGMP production. We tested orally delivered sGC activator, BAY 54-6544 (17 mg/kg/d), sGC stimulator, BAY 41-8543, sildenafil, and placebo for 4-12 weeks in the Berkeley transgenic mouse model of SCD (BERK-SCD) and their hemizygous (Hemi) littermate controls (BERK-Hemi). Right ventricular (RV) maximum systolic pressure (RVmaxSP) was measured using micro right-heart catheterization. RV hypertrophy (RVH) was determined using Fulton's index and RV corrected weight (ratio of RV to tibia). Pulmonary artery vasoreactivity was tested for endothelium-dependent and -independent vessel relaxation. Right-heart catheterization revealed higher RVmaxSP and RVH in BERK-SCD versus BERK-Hemi, which worsened with age. Treatment with the sGC activator more effectively lowered RVmaxSP and RVH, with 90-day treatment delivering superior results, when compared with other treatments and placebo groups. In myography experiments, acetylcholine-induced (endothelium-dependent) and sodium-nitroprusside-induced (endothelium-independent NO donor) relaxation of the pulmonary artery harvested from placebo-treated BERK-SCD was impaired relative to BERK-Hemi but improved after therapy with sGC activator. By contrast, no significant effect for sGC stimulator or sildenafil was observed in BERK-SCD. These findings suggest that sGC is oxidized in the pulmonary arteries of transgenic SCD mice, leading to blunted responses to NO, and that the sGC activator, BAY 54-6544, may represent a novel therapy for SCD-associated pulmonary arterial hypertension and cardiac remodeling.
Collapse
Affiliation(s)
- Karin P Potoka
- 1 Division of Newborn Medicine, Department of Pediatrics.,2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | - Katherine C Wood
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine.,3 University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jeffrey J Baust
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | - Marta Bueno
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine.,4 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Scott A Hahn
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | | | - Tim Bachman
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | - Grace M Mallampalli
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine.,3 University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Valerie Schrott
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | - Bin Sun
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | - Grant C Bullock
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine
| | | | | | | | | | | | - Hubert Truebel
- 5 Bayer AG, Wuppertal, Germany.,6 University of Witten/Herdecke, Witten, Germany
| | - Peter Sandner
- 5 Bayer AG, Wuppertal, Germany.,8 Hannover Medical School, Hannover, Germany; and
| | - Ana L Mora
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine.,4 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adam C Straub
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine.,9 Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- 2 Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine.,4 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
46
|
West J, Chen X, Yan L, Gladson S, Loyd J, Rizwan H, Talati M. Adverse effects of BMPR2 suppression in macrophages in animal models of pulmonary hypertension. Pulm Circ 2019; 10:2045894019856483. [PMID: 31124398 PMCID: PMC7074495 DOI: 10.1177/2045894019856483] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/17/2019] [Indexed: 01/11/2023] Open
Abstract
Inflammatory cells contribute to irreversible damage in pulmonary arterial hypertension (PAH). We hypothesized that in PAH, dysfunctional BMPR2 signaling in macrophages contributes to pulmonary vascular injury and phenotypic changes via proinflammatory cytokine production. Studies were conducted in: (1) Rosa26-rtTA2 3 X TetO7-Bmpr2delx4 FVB/N mice (mutant Bmpr2 is universally expressed, BMPR2delx4 mice) given a weekly intra-tracheal liposomal clodronate injections for four weeks; and (2) LysM-Cre X floxed BMPR2 X floxed eGFP monocyte lineage-specific BMPR2 knockout (KO) mouse model (Bmpr2 gene expression knockdown in monocytic lineage cells) (BMPR2KO) following three weeks of sugen/hypoxia treatment. In the BMPR2delx4 mice, increased right ventricular systolic pressure (RVSP; P < 0.05) was normalized by clodronate, and in monocyte lineage-specific BMPR2KO mice sugen hypoxia treatment increased (P < 0.05) RVSP compared to control littermates, suggesting that suppressed BMPR2 in macrophages modulate RVSP in animal models of PH. In addition, in these mouse models, muscularized pulmonary vessels were increased (P < 0.05) and surrounded by an increased number of macrophages. Elimination of macrophages in BMPR2delx4 mice reduced the number of muscularized pulmonary vessels and macrophages surrounding these vessels. Further, in monocyte lineage-specific BMPR2KO mice, there was significant increase in proinflammatory cytokines, including C-X-C Motif Chemokine Ligand 12 (CXCL12), complement component 5 a (C5a), Interleukin-16 (IL-16), and secretory ICAM. C5a positive inflammatory cells present in and around the pulmonary vessels in the PAH lung could potentially be involved in pulmonary vessel remodeling. In summary, our data indicate that, in BMPR2-related PAH, macrophages with dysfunctional BMPR2 influence pulmonary vascular remodeling and phenotypic outcomes via proinflammatory cytokine production.
Collapse
Affiliation(s)
- James West
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Xinping Chen
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ling Yan
- Division of Medical Genetics and Genomic Medicine, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Santhi Gladson
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - James Loyd
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Hamid Rizwan
- Division of Medical Genetics and Genomic Medicine, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Megha Talati
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
47
|
Iranmehr A, Stobdan T, Zhou D, Poulsen O, Strohl KP, Aldashev A, Telenti A, Wong EHM, Kirkness EF, Venter JC, Bafna V, Haddad GG. Novel insight into the genetic basis of high-altitude pulmonary hypertension in Kyrgyz highlanders. Eur J Hum Genet 2019; 27:150-159. [PMID: 30254217 PMCID: PMC6303266 DOI: 10.1038/s41431-018-0270-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/09/2018] [Accepted: 08/30/2018] [Indexed: 02/07/2023] Open
Abstract
The Central Asian Kyrgyz highland population provides a unique opportunity to address genetic diversity and understand the genetic mechanisms underlying high-altitude pulmonary hypertension (HAPH). Although a significant fraction of the population is unaffected, there are susceptible individuals who display HAPH in the absence of any lung, cardiac or hematologic disease. We report herein the analysis of the whole-genome sequencing of healthy individuals compared with HAPH patients and other controls (total n = 33). Genome scans reveal selection signals in various regions, encompassing multiple genes from the first whole-genome sequences focusing on HAPH. We show here evidence of three candidate genes MTMR4, TMOD3 and VCAM1 that are functionally associated with well-known molecular and pathophysiological processes and which likely lead to HAPH in this population. These processes are (a) dysfunctional BMP signaling, (b) disrupted tissue repair processes and (c) abnormal endothelial cell function. Whole-genome sequence of well-characterized patients and controls and using multiple statistical tools uncovered novel candidate genes that belong to pathways central to the pathogenesis of HAPH. These studies on high-altitude human populations are pertinent to the understanding of sea level diseases involving hypoxia as a main element of their pathophysiology.
Collapse
Affiliation(s)
- Arya Iranmehr
- Department of Electrical & Computer Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Tsering Stobdan
- Division of Respiratory Medicine, Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Dan Zhou
- Division of Respiratory Medicine, Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Orit Poulsen
- Division of Respiratory Medicine, Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Kingman P Strohl
- Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Almaz Aldashev
- National Academy of Sciences, Bishkek, 720071, Kyrgyz Republic
| | - Amalio Telenti
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, La Jolla, CA, 92037, USA
| | | | | | - J Craig Venter
- Human Longevity Inc., San Diego, CA, 92121, USA
- J. Craig Venter Institute, La Jolla, CA, 92037, USA
| | - Vineet Bafna
- Department of Computer Science & Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Gabriel G Haddad
- Division of Respiratory Medicine, Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA.
- Department of Pediatrics, Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093, USA.
- Rady Children's Hospital, San Diego, CA, 92123, USA.
| |
Collapse
|
48
|
Correlation Between Soluble Endothelial Adhesion Molecules and Nitric Oxide Metabolites in Sickle Cell Disease. Med Sci (Basel) 2018; 7:medsci7010001. [PMID: 30577523 PMCID: PMC6359114 DOI: 10.3390/medsci7010001] [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] [Received: 10/31/2018] [Revised: 12/04/2018] [Accepted: 12/13/2018] [Indexed: 11/17/2022] Open
Abstract
Nitric Oxide (NO) and soluble adhesion molecules are promising biomarkers, which predict endothelial dysfunction in sickle cell disease (SCD). Several studies have investigated the relationship between NO (as well as its metabolites) and endothelial adhesion molecules in SCD. However, these studies were done mainly in the developed world, and it is difficult to extrapolate the findings to SCD populations in other geographical regions such as Africa due to significant disparities in the results. The aim of the current study was to determine the correlation between levels of nitric oxide metabolites (NOx) and adhesion molecules in SCD patients in a tertiary hospital in Ghana. A case control cross-sectional study involving 100 SCD (made up of HbSS and HbSC patients) and 60 healthy controls was conducted. Concentrations of NOx and soluble endothelial adhesion molecules (ICAM-1, VCAM-1 and E-selectin) were measured in all the study participants (n = 160) by the Griess reagent system and enzyme-linked immunosorbent assay (ELISA). Correlation analysis was performed to determine a possible link between the variables. Levels of soluble adhesion molecules were higher in the HbSS patients. Correlation of NOx with ICAM-1 almost approached significance (r = 0.565, p = 0.058) in the HbSS patients. There were no correlations between NOx and E-selectin in both HbSS and HbSC patients. There were no significant correlations between NOx and VCAM-1 in all the study participants (p > 0.05). Of the soluble adhesion molecules, ICAM-1 showed a significant positive correlation with VCAM-1 in the HbSC patients. There were no significant differences between the adhesion molecules and the age of participants in the various study groups. Whether or not a significant correlation exists between NOx and soluble adhesion molecules may not depend on the sickle cell genotype. The expression of adhesion molecules may not depend on age.
Collapse
|
49
|
Antwi-Boasiako C, Dzudzor B, Kudzi W, Doku A, Dale CA, Sey F, Otu KH, Boatemaa GD, Ekem I, Ahenkorah J, Achel DG, Aboagye ET, Donkor ES. Association between eNOS Gene Polymorphism (T786C and VNTR) and Sickle Cell Disease Patients in Ghana. Diseases 2018; 6:E90. [PMID: 30274269 PMCID: PMC6313677 DOI: 10.3390/diseases6040090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 09/21/2018] [Accepted: 09/27/2018] [Indexed: 01/01/2023] Open
Abstract
Endothelial nitric oxide synthase (eNOS) variants have been found to be associated with several vascular disorders as well as the pathogenesis of sickle cell disease (SCD) complications such as vaso-occlusive crises (VOC). Studies on eNOS gene variants among SCD patients are rare in Ghana and several other African countries. The current study aimed to determine a possible association between variants of the eNOS gene (variable number of tandem repeats in intron 4 and T786C) in SCD complications among Ghanaian patients. This was a cross-sectional study involving 89 HbSS patients with complications and 46 HbSS patients without complications. Genomic DNA was extracted from leukocytes in the buffy coat and separated from collected whole blood samples of the study participants. PCR amplification, followed by restriction fragment length polymorphism (RFLP) was used to genotype T786C (rs2070744) variants. Variable number of tandem repeats (VNTR) in intron 4 was genotyped by PCR and direct electrophoresis. There was a significant difference in the genotype frequency of the T786C variant between HbSS patients with complications and those without complications (p = 0.0165). However, there was no significant difference in the VNTR intron 4 variant of the eNOS gene between patients with complications and those without complications (p > 0.05). The study shows an association between the eNOS gene variant (T786C) and complications in SCD.
Collapse
Affiliation(s)
- Charles Antwi-Boasiako
- Department of Physiology, School of Biomedical and Allied Health Sciences, University of Ghana, Accra +233, Ghana.
| | - Bartholomew Dzudzor
- Department of Medical Biochemistry, School of Biomedical and Allied Health Sciences, University of Ghana, Accra +233, Ghana.
| | - William Kudzi
- Centre for Tropical Clinical Pharmacology and Therapeutics School of Biomedical and Allied Health Sciences, University of Ghana, Accra +233, Ghana.
| | - Alfred Doku
- Department of Internal Medicine, School of Medicine and Dentistry, University of Ghana, Accra +233, Ghana.
| | - Campbell Andrew Dale
- Center for Cancer and Blood Disorders Children's National Medical Center George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA.
| | - Fredericka Sey
- Center for Clinical Genetics, Korle-Bu Teaching Hospital, Accra +233, Ghana.
| | - Kate Hgar Otu
- Department of Nursing and Midwifery, Greenhills School of Health Sciences, Accra +233, Ghana.
| | - Gifty Dankwah Boatemaa
- Department of Physiology, School of Biomedical and Allied Health Sciences, University of Ghana, Accra +233, Ghana.
| | - Ivy Ekem
- Department of Haematology, School of Medical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast +233, Ghana.
| | - John Ahenkorah
- Department of Anatomy, School of Biomedical and Allied Health Sciences, University of Ghana, Accra +233, Ghana.
| | - Daniel Gyingiri Achel
- Applied Radiation Biology Center, Radiological and Medical Sciences Research, Ghana Atomic Energy Commission, Accra +233, Ghana.
| | - Elvis Twumasi Aboagye
- Centre for Tropical Clinical Pharmacology and Therapeutics School of Biomedical and Allied Health Sciences, University of Ghana, Accra +233, Ghana.
| | - Eric S Donkor
- Department of Medical Microbiology, School of Biomedical and Allied Health Sciences, University of Ghana, Accra +233, Ghana.
| |
Collapse
|
50
|
Connes P, Renoux C, Romana M, Abkarian M, Joly P, Martin C, Hardy-Dessources MD, Ballas SK. Blood rheological abnormalities in sickle cell anemia. Clin Hemorheol Microcirc 2018; 68:165-172. [PMID: 29614630 DOI: 10.3233/ch-189005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This review focuses on the contribution of abnormal blood rheology in the pathophysiology of sickle cell anemia (SCA). SCA is characterized by a reduction of red blood cell (RBC) deformability but this reduction is very heterogeneous among patients. Recent works have shown that patients with the lowest RBC deformability (measured by ektacytometry) have enhanced hemolysis and would be more prone to develop several complications such as priapism, leg ulcers and glomerulopathy. In contrast, patients with the highest deformability, and not under hydroxyurea therapy, seem to develop more frequently vaso-occlusive like events. Although less studied, RBC aggregation properties are very different between SCA and healthy individuals and it was demonstrated that increased RBC aggregates strength could be involved in some complications. Finally, several studies have established that the vascular system of SCA patients could not fully compensate any increase in blood viscosity because of the loss of vascular reactivity, which may result in vaso-occlusive crises.
Collapse
Affiliation(s)
- Philippe Connes
- Laboratoire LIBM EA7424, Team"Vascular Biology and Red Blood Cell", Université Claude Bernard Lyon 1, University of Lyon, 69100 Villeurbanne, France.,Laboratory of Excellence GR-Ex « The red cell: from genesis to death », PRES Sorbonne Paris Cité, 75015, Paris, France.,Institut Universitaire de France, Paris, France
| | - Céline Renoux
- Laboratoire LIBM EA7424, Team"Vascular Biology and Red Blood Cell", Université Claude Bernard Lyon 1, University of Lyon, 69100 Villeurbanne, France.,Laboratory of Excellence GR-Ex « The red cell: from genesis to death », PRES Sorbonne Paris Cité, 75015, Paris, France.,Laboratoire de biochimie des pathologies érythrocytaires, Centre de Biologie Est, Hospices Civils de Lyon, France
| | - Marc Romana
- Laboratory of Excellence GR-Ex « The red cell: from genesis to death », PRES Sorbonne Paris Cité, 75015, Paris, France.,Inserm UMR 1134, Hôpital Ricou, CHU de Pointe-à-Pitre, 97157 Pointe-à-Pitre, Guadeloupe
| | - Manouk Abkarian
- CNRS UMR 5048, Université de Montpellier, Centre de Biochimie Structurale, 34090 Montpellier, France
| | - Philippe Joly
- Laboratoire LIBM EA7424, Team"Vascular Biology and Red Blood Cell", Université Claude Bernard Lyon 1, University of Lyon, 69100 Villeurbanne, France.,Laboratory of Excellence GR-Ex « The red cell: from genesis to death », PRES Sorbonne Paris Cité, 75015, Paris, France.,Laboratoire de biochimie des pathologies érythrocytaires, Centre de Biologie Est, Hospices Civils de Lyon, France
| | - Cyril Martin
- Laboratoire LIBM EA7424, Team"Vascular Biology and Red Blood Cell", Université Claude Bernard Lyon 1, University of Lyon, 69100 Villeurbanne, France.,Laboratory of Excellence GR-Ex « The red cell: from genesis to death », PRES Sorbonne Paris Cité, 75015, Paris, France
| | - Marie-Dominique Hardy-Dessources
- Laboratory of Excellence GR-Ex « The red cell: from genesis to death », PRES Sorbonne Paris Cité, 75015, Paris, France.,Inserm UMR 1134, Hôpital Ricou, CHU de Pointe-à-Pitre, 97157 Pointe-à-Pitre, Guadeloupe
| | - Samir K Ballas
- Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|