1
|
Gupta P, Shrivastava S, Kumar R. Musculoskeletal complications in sickle cell disease: Pathophysiology, diagnosis and management. Best Pract Res Clin Rheumatol 2025:102033. [PMID: 39824706 DOI: 10.1016/j.berh.2025.102033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/20/2025]
Abstract
Sickle cell disease (SCD) is a mono-genic disorder causing chronic hemolysis, anemia, and vaso-occlusion, leading to musculoskeletal complications such as osteonecrosis, osteoporosis, and bone fractures affecting 50-70% SCD patients. These complications result from a complex interplay of genetic and physiological factors, including abnormal hemoglobin production, chronic inflammation, and oxidative stress. This review discusses the pathophysiology, pre-clinical symptoms, and clinical manifestations of musculoskeletal complications in SCD, as well as current treatment options, including pharmacological interventions, surgical procedures, and bone marrow transplantation. Early detection of pre-clinical symptoms is crucial to prevent progression. Pharmacological interventions (analgesics, anti-inflammatory agents, bone-modifying agents and hydroxyurea), surgical interventions (core decompression, bone grafting, joint replacement and osteotomy) and supportive measures enhance mobility, strength and well-being. A multidisciplinary approach is essential for optimal care, and early diagnosis and management are crucial to prevent long-term damage and improve outcomes. Future research directions include targeted therapies, biomarker investigation and infrastructure development to improve outcomes for SCD individuals with musculoskeletal complications.
Collapse
Affiliation(s)
- Parul Gupta
- ICMR-National Institute of Research in Tribal Health, Jabalpur, India
| | - Suyesh Shrivastava
- ICMR-National Institute of Research in Tribal Health, Jabalpur, India; Model Rural Health Research Unit, Badoni, Datia, India
| | - Ravindra Kumar
- ICMR-National Institute of Research in Tribal Health, Jabalpur, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India; Model Rural Health Research Unit, Jheet, Durg, India.
| |
Collapse
|
2
|
Gupta P, Kumar R. Targeting ICAM1 to Ameliorate Vaso-Occlusion and Inflammation in Sickle Cell Disease. Eur J Haematol 2024; 113:730-737. [PMID: 39354752 DOI: 10.1111/ejh.14313] [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: 07/10/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024]
Abstract
Sickle cell disease (SCD) is a hereditary disorder characterized by vaso-occlusion, inflammation, and tissue damage. Intercellular adhesion molecule 1 (ICAM-1) plays a crucial role in the pathophysiology of SCD by promoting the adhesion of sickle cells to the endothelium, contributing to vaso-occlusion and tissue damage. The ICAM-1 gene encodes a glycoprotein that interacts with lymphocyte function-associated antigen 1 (LFA-1) and macrophage 1-antigen (Mac-1) receptors, perpetuating inflammation, and oxidative stress. The NF-κB signaling pathway regulates ICAM-1 expression, which is elevated in patients with SCD, leading to increased endothelial cell activation and damage. Targeting ICAM-1 and its interactions with sickle cells and the endothelium has emerged as a potential therapeutic strategy for managing SCD. This review highlights the complex interplay between ICAM-1, sickle cells, and the endothelium, and discusses the potential of ICAM-1-targeted therapies for mitigating VOC and improving the quality of life for patients with SCD.
Collapse
Affiliation(s)
- Parul Gupta
- Division of Genetic Disorders, ICMR-National Institute of Research in Tribal Health, Jabalpur, India
| | - Ravindra Kumar
- Division of Genetic Disorders, ICMR-National Institute of Research in Tribal Health, Jabalpur, India
| |
Collapse
|
3
|
Gupta P, Choudhari V, Kumar R. Exploring the genetic mechanisms: SELP gene's contribution to alleviating vaso-occlusive crisis in sickle cell disease. Gene 2024; 928:148805. [PMID: 39079562 DOI: 10.1016/j.gene.2024.148805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/09/2024] [Accepted: 07/26/2024] [Indexed: 08/03/2024]
Abstract
Sickle cell disease is a catastrophic inflammatory disorder characterized by microvascular vaso-occlusion, leading to high morbidity and increased mortality. P-selectin, a cell adhesion molecule, plays a crucial role in the pathogenesis and severity of sickle cell disease. Its expression and binding with its ligand PSGL-1 is involved in various mechanisms that contribute to inflammation and immune response, resulting in complications in sickle cell disease. Preclinical data have verified the efficacy of P-Selectin inhibition in mitigating vaso-occlusive events and severity of disease. Currently clinical trials are ongoing to evaluate the safety and efficiency of P-Selectin-targeted therapies and concede the challenges and limitations associated with their use. Despite of its proven role in reducing severity in sickle cell disease, future research should focus on identifying other novel targets within the adhesion cascade and explore combination therapies. Conducting trials and addressing concerns about accessibility are crucial steps towards fully harnessing the potential of P selectin inhibitors as a groundbreaking treatment option. This review focuses on understanding the role of p selectin and its interactions with molecules involved in inflammation providing insights about the molecular etiology, pathophysiology, and potential therapeutic targets in sickle cell disease.
Collapse
Affiliation(s)
- Parul Gupta
- ICMR-National Institute of Research in Tribal Health, India
| | | | - Ravindra Kumar
- ICMR-National Institute of Research in Tribal Health, India.
| |
Collapse
|
4
|
Connes P, Stauffer E, Liem RI, Nader E. Exercise and training in sickle cell disease: Safety, potential benefits, and recommendations. Am J Hematol 2024; 99:1988-2001. [PMID: 39132839 DOI: 10.1002/ajh.27454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/28/2024] [Accepted: 07/23/2024] [Indexed: 08/13/2024]
Abstract
Sickle cell disease (SCD) is a genetic disorder characterized by complex pathophysiological mechanisms leading to vaso-occlusive crisis, chronic pain, chronic hemolytic anemia, and vascular complications, which require considerations for exercise and physical activity. This review aims to elucidate the safety, potential benefits, and recommendations regarding exercise and training in individuals with SCD. SCD patients are characterized by decreased exercise capacity and tolerance. Acute intense exercise may be accompanied by biological changes (acidosis, increased oxidative stress, and dehydration) that could increase the risk of red blood cell sickling and acute clinical complications. However, recent findings suggest that controlled exercise training is safe and well tolerated by SCD patients and could confer benefits in disease management. Regular endurance exercises of submaximal intensity or exercise interventions incorporating resistance training have been shown to improve cardiorespiratory and muscle function in SCD, which may improve quality of life. Recommendations for exercise prescription in SCD should be based on accurate clinical and functional evaluations, taking into account disease phenotype and cardiorespiratory status at rest and in response to exercise. Exercise programs should include gradual progression, incorporating adequate warm-up, cool-down, and hydration strategies. Exercise training represents promising therapeutic strategy in the management of SCD. It is now time to move through the investigation of long-term biological, physiological, and clinical effects of regular physical activity in SCD patients.
Collapse
Affiliation(s)
- 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
| | - Emeric Stauffer
- 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
- Explorations Fonctionnelles Respiratoires, Médecine du sport et de l'Activité Physique, Hospices Civils de Lyon, Hôpital Croix Rousse, Lyon, France
| | - Robert I Liem
- Division of Hematology, Oncology and Stem Cell Transplant, Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - 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
| |
Collapse
|
5
|
Kaltsas A, Zikopoulos A, Dimitriadis F, Sheshi D, Politis M, Moustakli E, Symeonidis EN, Chrisofos M, Sofikitis N, Zachariou A. Oxidative Stress and Erectile Dysfunction: Pathophysiology, Impacts, and Potential Treatments. Curr Issues Mol Biol 2024; 46:8807-8834. [PMID: 39194738 DOI: 10.3390/cimb46080521] [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: 07/10/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Erectile dysfunction (ED) is a prevalent condition affecting men's sexual health, with oxidative stress (OS) having recently been identified as a significant contributing causative factor. This narrative review aims to elucidate the role of OS in the pathophysiology of ED, focusing on impact, mechanisms, and potential therapeutic interventions. Key findings indicate that OS disrupts endothelial function and nitric oxide (NO) signaling, crucial for erectile function. Various sources of reactive oxygen species (ROS) and their detrimental effects on penile tissue are discussed, including aging, diabetes mellitus, hypertension, hyperlipidemia, smoking, obesity, alcohol consumption, psychological stress, hyperhomocysteinemia, chronic kidney disease, and sickle cell disease. Major sources of ROS, such as NADPH oxidase, xanthine oxidase, uncoupled endothelial NO synthase (eNOS), and mitochondrial electron transport, are identified. NO is scavenged by these ROS, leading to endothelial dysfunction characterized by reduced NO availability, impaired vasodilation, increased vascular tone, and inflammation. This ultimately results in ED due to decreased blood flow to penile tissue and the inability to achieve or maintain an erection. Furthermore, ROS impact the transmission of nitrergic neurotransmitters by causing the death of nitrergic neurons and reducing the signaling of neuronal NO synthase (nNOS), exacerbating ED. Therapeutic approaches targeting OS, including antioxidants and lifestyle modifications, show promise in ameliorating ED symptoms. The review underscores the need for further research to develop effective treatments, emphasizing the interplay between OS and vascular health in ED. Integrating pharmacological and non-pharmacological strategies could enhance clinical outcomes for ED patients, advocating for OS management in ED treatment protocols to improve patient quality of life.
Collapse
Affiliation(s)
- Aris Kaltsas
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | | | - Fotios Dimitriadis
- Department of Urology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Danja Sheshi
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Magdalena Politis
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Efthalia Moustakli
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Evangelos N Symeonidis
- Department of Urology II, European Interbalkan Medical Center, 55535 Thessaloniki, Greece
| | - Michael Chrisofos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Nikolaos Sofikitis
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Athanasios Zachariou
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
6
|
Rizvi I, Solipuram D, Kaur N, Komel A, Batool S, Wang J. The enigma of sickle cell hepatopathy: Pathophysiology, clinical manifestations and therapy. Br J Haematol 2024. [PMID: 38978231 DOI: 10.1111/bjh.19620] [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: 03/27/2024] [Accepted: 06/19/2024] [Indexed: 07/10/2024]
Abstract
Sickle cell disease (SCD) is one of the most common genetic disorders in the world predominantly affecting economically disadvantaged populations. There is a notable discrepancy between the growing adult SCD population and available diagnostic and therapeutic interventions for SCD. Sickle cell hepatopathy (SCH) is an all-inclusive term to describe the acute and chronic liver manifestations of SCD. The pathophysiology of SCH follows no defined pattern or sequence that poses challenges to clinicians and researchers alike. Evidence is lacking for this underreported disease at various levels from diagnostic to therapeutic options. This paper reviews the basic pathophysiology, clinical features, biochemical and radiological findings of various SCH manifestations and outlines the management of each condition. Old and new therapy options in SCD including hydroxyurea, red blood cell exchange transfusion, ursodeoxycholic acid, voxelotor, l-glutamine and crizanlizumab have been reviewed to investigate the role of these options in treating SCH. The role of liver transplant, haematopoietic stem cell transplant and gene therapy in SCH patients have been reviewed.
Collapse
Affiliation(s)
- Insia Rizvi
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Divya Solipuram
- Internal Medicine, Nassau University Medical Center, East Meadow, New York, USA
| | - Navneet Kaur
- Internal Medicine, North Alabama Medical Center, Florence, Alabama, USA
| | - Aqsa Komel
- Internal Medicine, Nishtar Medical College and Hospital, Multan, Punjab, Pakistan
| | - Saba Batool
- Internal Medicine, Carle Health Methodist Hospital, Peoria, Illinois, USA
| | - Jennifer Wang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
7
|
Pavitra E, Acharya RK, Gupta VK, Verma HK, Kang H, Lee JH, Sahu T, Bhaskar L, Raju GSR, Huh YS. Impacts of oxidative stress and anti-oxidants on the development, pathogenesis, and therapy of sickle cell disease: A comprehensive review. Biomed Pharmacother 2024; 176:116849. [PMID: 38823275 DOI: 10.1016/j.biopha.2024.116849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/17/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024] Open
Abstract
Sickle cell disease (SCD) is the most severe monogenic hemoglobinopathy caused by a single genetic mutation that leads to repeated polymerization and depolymerization of hemoglobin resulting in intravascular hemolysis, cell adhesion, vascular occlusion, and ischemia-reperfusion injury. Hemolysis causes oxidative damage indirectly by generating reactive oxygen species through various pathophysiological mechanisms, which include hemoglobin autoxidation, endothelial nitric oxide synthase uncoupling, reduced nitric oxide bioavailability, and elevated levels of asymmetric dimethylarginine. Red blood cells have a built-in anti-oxidant system that includes enzymes like sodium dismutase, catalase, and glutathione peroxidase, along with free radical scavenging molecules, such as vitamin C, vitamin E, and glutathione, which help them to fight oxidative damage. However, these anti-oxidants may not be sufficient to prevent the effects of oxidative stress in SCD patients. Therefore, in line with a recent FDA request that the focus to be placed on the development of innovative therapies for SCD that address the root cause of the disease, there is a need for therapies that target oxidative stress and restore redox balance in SCD patients. This review summarizes the current state of knowledge regarding the role of oxidative stress in SCD and the potential benefits of anti-oxidant therapies. It also discusses the challenges and limitations of these therapies and suggests future directions for research and development.
Collapse
Affiliation(s)
- Eluri Pavitra
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea; 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea
| | - Rakesh Kumar Acharya
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh 495009, India
| | - Vivek Kumar Gupta
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of lungs health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Neuherberg, Munich 85764, Germany
| | - Haneul Kang
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Jeong-Hwan Lee
- 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea
| | - Tarun Sahu
- Department of Physiology, All Indian Institute of Medical Science, Raipur, Chhattisgarh, India
| | - Lvks Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh 495009, India.
| | - Ganji Seeta Rama Raju
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 04620, Republic of Korea.
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
8
|
Catella J, Guillot N, Nader E, Skinner S, Poutrel S, Hot A, Connes P, Fromy B. Controversies in the pathophysiology of leg ulcers in sickle cell disease. Br J Haematol 2024; 205:61-70. [PMID: 38867511 DOI: 10.1111/bjh.19584] [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: 12/22/2023] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Patients with sickle cell disease (SCD) often experience painful vaso-occlusive crises and chronic haemolytic anaemia, as well as various acute and chronic complications, such as leg ulcers. Leg ulcers are characterized by their unpredictability, debilitating pain and prolonged healing process. The pathophysiology of SCD leg ulcers is not well defined. Known risk factors include male gender, poor social conditions, malnutrition and a lack of compression therapy when oedema occurs. Leg ulcers typically start with spontaneous pain, followed by induration, hyperpigmentation, blister formation and destruction of the epidermis. SCD is characterized by chronic haemolysis, increased oxidative stress and decreased nitric oxide bioavailability, which promote ischaemia and inflammation and consequently impair vascular function in the skin. This cutaneous vasculopathy, coupled with venostasis around the ankle, creates an ideal environment for local vaso-occlusive crises, which can result in the development of leg ulcers that resemble arterial ulcers. Following the development of the ulcer, healing is hindered as a result of factors commonly observed in venous ulceration, including venous insufficiency, oedema and impaired angiogenesis. All of these factors are modulated by genetic factors. However, our current understanding of these genetic factors remains limited and does not yet enable us to accurately predict ulceration susceptibility.
Collapse
Affiliation(s)
- Judith Catella
- Service de Médecine Interne et Vasculaire, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI UMR 5305), CNRS/Université Claude Bernard Lyon 1, Lyon, France
| | - Nicolas Guillot
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Elie Nader
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Sarah Skinner
- Clinical Research and Epidemiology Unit, Montpellier University, Montpellier, France
| | - Solène Poutrel
- Service de Médecine Interne et Vasculaire, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Arnaud Hot
- Service de Médecine Interne et Vasculaire, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Philippe Connes
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Berengère Fromy
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI UMR 5305), CNRS/Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
9
|
Bhatt S, Argueta DA, Gupta K, Kundu S. Red Blood Cells as Therapeutic Target to Treat Sickle Cell Disease. Antioxid Redox Signal 2024; 40:1025-1049. [PMID: 37975291 DOI: 10.1089/ars.2023.0348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Significance: Sickle cell disease (SCD) is the most common inherited diathesis affecting mostly underserved populations globally. SCD is characterized by chronic pain and fatigue, severe acute painful crises requiring hospitalization and opioids, strokes, multiorgan damage, and a shortened life span. Symptoms may appear shortly after birth, and, in less developed countries, most children with SCD die before attaining age 5. Hematopoietic stem cell transplant and gene therapy offer a curative therapeutic approach, but, due to many challenges, are limited in their availability and effectiveness for a majority of persons with SCD. A critical unmet need is to develop safe and effective novel targeted therapies. A wide array of drugs currently undergoing clinical investigation hold promise for an expanded pharmacological armamentarium against SCD. Recent Advances: Hydroxyurea, the most widely used intervention for SCD management, has improved the survival in the Western world and more recently, voxelotor (R-state-stabilizer), l-glutamine, and crizanlizumab (anti-P-selectin antibody) have been approved by the Food and Drug Administration (FDA) for use in SCD. The recent FDA approval emphasizes the need to revisit the advances in understanding the core pathophysiology of SCD to accelerate novel evidence-based strategies to treat SCD. The biomechanical breakdown of erythrocytesis, the core pathophysiology of SCD, is associated with intrinsic factors, including the composition of hemoglobin, membrane integrity, cellular volume, hydration, andoxidative stress. Critical Issues and Future Directions: In this context, this review focuses on advances in emerging nongenetic interventions directed toward the therapeutic targets intrinsic to sickle red blood cells (RBCs), which can prevent impaired rheology of RBCs to impede disease progression and reduce the sequelae of comorbidities, including pain, vasculopathy, and organ damage. In addition, given the intricate pathophysiology of the disease, it is unlikely that a single pharmacotherapeutic intervention will comprehensively ameliorate the multifaceted complications associated with SCD. However, the availability of multiple drug options affords the opportunity for individualized therapeutic regimens tailored to specific SCD-related complications. Furthermore, it opens avenues for combination drug therapy, capitalizing on distinct mechanisms of action and profiles of adverse effects.
Collapse
Affiliation(s)
- Shruti Bhatt
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Donovan A Argueta
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, California, USA
| | - Kalpna Gupta
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, California, USA
| | - Suman Kundu
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, KK Birla Goa Campus, Goa, India
| |
Collapse
|
10
|
Bolarinwa AB, Oduwole O, Okebe J, Ogbenna AA, Otokiti OE, Olatinwo AT. Antioxidant supplementation for sickle cell disease. Cochrane Database Syst Rev 2024; 5:CD013590. [PMID: 38775255 PMCID: PMC11110109 DOI: 10.1002/14651858.cd013590.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
BACKGROUND Sickle cell disease (SCD) refers to a group of genetic disorders characterized by the presence of an abnormal haemoglobin molecule called haemoglobin S (HbS). When subjected to oxidative stress from low oxygen concentrations, HbS molecules form rigid polymers, giving the red cell the typical sickle shape. Antioxidants have been shown to reduce oxidative stress and improve outcomes in other diseases associated with oxidative stress. Therefore, it is important to review and synthesize the available evidence on the effect of antioxidants on the clinical outcomes of people with SCD. OBJECTIVES To assess the effectiveness and safety of antioxidant supplementation for improving health outcomes in people with SCD. SEARCH METHODS We used standard, extensive Cochrane search methods. The latest search date was 15 August 2023. SELECTION CRITERIA We included randomized and quasi-randomized controlled trials comparing antioxidant supplementation to placebo, other antioxidants, or different doses of antioxidants, in people with SCD. DATA COLLECTION AND ANALYSIS Two authors independently extracted data, assessed the risk of bias and certainty of the evidence, and reported according to Cochrane methodological procedures. MAIN RESULTS The review included 1609 participants in 26 studies, with 17 comparisons. We rated 13 studies as having a high risk of bias overall, and 13 studies as having an unclear risk of bias overall due to study limitations. We used GRADE to rate the certainty of evidence. Only eight studies reported on our important outcomes at six months. Vitamin C (1400 mg) plus vitamin E (800 mg) versus placebo Based on evidence from one study in 83 participants, vitamin C (1400 mg) plus vitamin E (800 mg) may not be better than placebo at reducing the frequency of crisis (risk ratio (RR) 1.18, 95% confidence interval (CI) 0.64 to 2.18), the severity of pain (RR 1.33, 95% CI 0.40 to 4.37), or adverse effects (AE), of which the most common were headache, nausea, fatigue, diarrhoea, and epigastric pain (RR 0.56, 95% CI 0.31 to 1.00). Vitamin C plus vitamin E may increase the risk of SCD-related complications (acute chest syndrome: RR 2.66, 95% CI 0.77 to 9.13; 1 study, 83 participants), and increase haemoglobin level (median (interquartile range) 90 (81 to 96) g/L versus 93.5 (84 to 105) g/L) (1 study, 83 participants) compared to placebo. However, the evidence for all the above effects is very uncertain. The study did not report on quality of life (QoL) of participants and their caregivers, nor on frequency of hospitalization. Zinc versus placebo Zinc may not be better than placebo at reducing the frequency of crisis at six months (rate ratio 0.62, 95% CI 0.17 to 2.29; 1 study, 36 participants; low-certainty evidence). We are uncertain whether zinc is better than placebo at improving sickle cell-related complications (complete healing of leg ulcers at six months: RR 2.00, 95% CI 0.60 to 6.72; 1 study, 34 participants; very low-certainty evidence). Zinc may be better than placebo at increasing haemoglobin level (g/dL) (MD 1.26, 95% CI 0.44 to 1.26; 1 study, 36 participants; low-certainty evidence). The study did not report on severity of pain, QoL, AE, and frequency of hospitalization. N-acetylcysteine versus placebo N-acetylcysteine (NAC) 1200 mg may not be better than placebo at reducing the frequency of crisis in SCD, reported as pain days (rate ratio 0.99 days, 95% CI 0.53 to 1.84; 1 study, 96 participants; low-certainty evidence). Low-certainty evidence from one study (96 participants) suggests NAC (1200 mg) may not be better than placebo at reducing the severity of pain (MD 0.17, 95% CI -0.53 to 0.87). Compared to placebo, NAC (1200 mg) may not be better at improving physical QoL (MD -1.80, 95% CI -5.01 to 1.41) and mental QoL (MD 2.00, 95% CI -1.45 to 5.45; very low-certainty evidence), reducing the risk of adverse effects (gastrointestinal complaints, pruritus, or rash) (RR 0.92, 95% CI 0.75 to 1.14; low-certainty evidence), reducing the frequency of hospitalizations (rate ratio 0.98, 95% CI 0.41 to 2.38; low-certainty evidence), and sickle cell-related complications (RR 5.00, 95% CI 0.25 to 101.48; very low-certainty evidence), or increasing haemoglobin level (MD -0.18 g/dL, 95% CI -0.40 to 0.04; low-certainty evidence). L-arginine versus placebo L-arginine may not be better than placebo at reducing the frequency of crisis (monthly pain) (RR 0.71, 95% CI 0.26 to 1.95; 1 study, 50 participants; low-certainty evidence). However, L-arginine may be better than placebo at reducing the severity of pain (MD -1.41, 95% CI -1.65 to -1.18; 2 studies, 125 participants; low-certainty evidence). One participant allocated to L-arginine developed hives during infusion of L-arginine, another experienced acute clinical deterioration, and a participant in the placebo group had clinically relevant increases in liver function enzymes. The evidence is very uncertain whether L-arginine is better at reducing the mean number of days in hospital compared to placebo (MD -0.85 days, 95% CI -1.87 to 0.17; 2 studies, 125 participants; very low-certainty evidence). Also, L-arginine may not be better than placebo at increasing haemoglobin level (MD 0.4 g/dL, 95% CI -0.50 to 1.3; 2 studies, 106 participants; low-certainty evidence). No study in this comparison reported on QoL and sickle cell-related complications. Omega-3 versus placebo Very low-certainty evidence shows no evidence of a difference in the risk of adverse effects of omega-3 compared to placebo (RR 1.05, 95% CI 0.74 to 1.48; 1 study, 67 participants). Very low-certainty evidence suggests that omega-3 may not be better than placebo at increasing haemoglobin level (MD 0.36 g/L, 95% CI -0.21 to 0.93; 1 study, 67 participants). The study did not report on frequency of crisis, severity of pain, QoL, frequency of hospitalization, and sickle cell-related complications. AUTHORS' CONCLUSIONS There was inconsistent evidence on all outcomes to draw conclusions on the beneficial and harmful effects of antioxidants. However, L-arginine may be better than placebo at reducing the severity of pain at six months, and zinc may be better than placebo at increasing haemoglobin level. We are uncertain whether other antioxidants are beneficial for SCD. Larger studies conducted on each comparison would reduce the current uncertainties.
Collapse
Affiliation(s)
- Abiola B Bolarinwa
- Department of Haematology & Blood Transfusion Medicine, Lagos University Teaching Hospital, Lagos, Nigeria
| | - Olabisi Oduwole
- Department of Medical Laboratory Science, Achievers University, Owo, Nigeria
| | - Joseph Okebe
- Department of International Public Health, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Ann A Ogbenna
- Department of Haematology & Blood Transfusion Medicine, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Oluwakemi E Otokiti
- Department of Haematology & Blood Transfusion Medicine, Lagos University Teaching Hospital, Lagos, Nigeria
| | - Adejoke T Olatinwo
- Department of Haematology & Blood Transfusion Medicine, Lagos University Teaching Hospital, Lagos, Nigeria
| |
Collapse
|
11
|
Xi C, Palani C, Takezaki M, Shi H, Horuzsko A, Pace BS, Zhu X. Simvastatin-Mediated Nrf2 Activation Induces Fetal Hemoglobin and Antioxidant Enzyme Expression to Ameliorate the Phenotype of Sickle Cell Disease. Antioxidants (Basel) 2024; 13:337. [PMID: 38539870 PMCID: PMC10968127 DOI: 10.3390/antiox13030337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 06/04/2024] Open
Abstract
Sickle cell disease (SCD) is a pathophysiological condition of chronic hemolysis, oxidative stress, and elevated inflammation. The transcription factor Nrf2 is a master regulator of oxidative stress. Here, we report that the FDA-approved oral agent simvastatin, an inhibitor of hydroxymethyl-glutaryl coenzyme A reductase, significantly activates the expression of Nrf2 and antioxidant enzymes. Simvastatin also induces fetal hemoglobin expression in SCD patient primary erythroid progenitors and a transgenic mouse model. Simvastatin alleviates SCD symptoms by decreasing hemoglobin S sickling, oxidative stress, and inflammatory stress in erythroblasts. Particularly, simvastatin increases cellular levels of cystine, the precursor for the biosynthesis of the antioxidant reduced glutathione, and decreases the iron content in SCD mouse spleen and liver tissues. Mechanistic studies suggest that simvastatin suppresses the expression of the critical histone methyltransferase enhancer of zeste homolog 2 to reduce both global and gene-specific histone H3 lysine 27 trimethylation. These chromatin structural changes promote the assembly of transcription complexes to fetal γ-globin and antioxidant gene regulatory regions in an antioxidant response element-dependent manner. In summary, our findings suggest that simvastatin activates fetal hemoglobin and antioxidant protein expression, modulates iron and cystine/reduced glutathione levels to improve the phenotype of SCD, and represents a therapeutic strategy for further development.
Collapse
Affiliation(s)
- Caixia Xi
- Department of Pediatrics, Division of Hematology/Oncology, Augusta University, Augusta, GA 30912, USA; (C.X.); (C.P.)
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA (A.H.)
| | - Chithra Palani
- Department of Pediatrics, Division of Hematology/Oncology, Augusta University, Augusta, GA 30912, USA; (C.X.); (C.P.)
| | - Mayuko Takezaki
- Department of Pediatrics, Division of Hematology/Oncology, Augusta University, Augusta, GA 30912, USA; (C.X.); (C.P.)
| | - Huidong Shi
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA (A.H.)
| | - Anatolij Horuzsko
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA (A.H.)
| | - Betty S. Pace
- Department of Pediatrics, Division of Hematology/Oncology, Augusta University, Augusta, GA 30912, USA; (C.X.); (C.P.)
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA (A.H.)
| | - Xingguo Zhu
- Department of Pediatrics, Division of Hematology/Oncology, Augusta University, Augusta, GA 30912, USA; (C.X.); (C.P.)
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA (A.H.)
| |
Collapse
|
12
|
Gupta P, Kumar R. Nitric oxide: A potential etiological agent for vaso-occlusive crises in sickle cell disease. Nitric Oxide 2024; 144:40-46. [PMID: 38316197 DOI: 10.1016/j.niox.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/27/2023] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Nitric oxide (NO), a vasodilator contributes to the vaso-occlusive crisis associated with the sickle cell disease (SCD). Vascular nitric oxide helps in vasodilation, controlled platelet aggregation, and preventing adhesion of sickled red blood cells to the endothelium. It decreases the expression of pro-inflammatory genes responsible for atherogenesis associated with SCD. Haemolysis and activated endothelium in SCD patients reduce the bioavailability of NO which promotes the severity of sickle cell disease mainly causes vaso-occlusive crises. Additionally, NO depletion can also contribute to the formation of thrombus, which can cause serious complications such as stroke, pulmonary embolism etc. Understanding the multifaceted role of NO provides valuable insights into its therapeutic potential for managing SCD and preventing associated complications. Various clinical trials and studies suggested the importance of artificially induced nitric oxide and its supplements in the reduction of severity. Further research on the mechanisms of NO depletion in SCD is needed to develop more effective treatment strategies and improve the management of this debilitating disease.
Collapse
Affiliation(s)
- Parul Gupta
- ICMR-National Institute of Research in Tribal Health, India
| | - Ravindra Kumar
- ICMR-National Institute of Research in Tribal Health, India.
| |
Collapse
|
13
|
Gorur V, Kranc KR, Ganuza M, Telfer P. Haematopoietic stem cell health in sickle cell disease and its implications for stem cell therapies and secondary haematological disorders. Blood Rev 2024; 63:101137. [PMID: 37919142 DOI: 10.1016/j.blre.2023.101137] [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: 04/25/2023] [Revised: 10/04/2023] [Accepted: 10/04/2023] [Indexed: 11/04/2023]
Abstract
Gene modification of haematopoietic stem cells (HSCs) is a potentially curative approach to sickle cell disease (SCD) and offers hope for patients who are not eligible for allogeneic HSC transplantation. Current approaches require in vitro manipulation of healthy autologous HSC prior to their transplantation. However, the health and integrity of HSCs may be compromised by a variety of disease processes in SCD, and challenges have emerged in the clinical trials of gene therapy. There is also concern about increased susceptibility to haematological malignancies during long-term follow up of patients, and this raises questions about genomic stability in the stem cell compartment. In this review, we evaluate the evidence for HSC deficits in SCD and then discuss their potential causation. Finally, we suggest several questions which need to be addressed in order to progress with successful HSC manipulation for gene therapy in SCD.
Collapse
Affiliation(s)
- Vishaka Gorur
- William Harvey Research Institute, Queen Mary University of London, EC1M 6BQ, UK.
| | - Kamil R Kranc
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK.
| | - Miguel Ganuza
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK.
| | - Paul Telfer
- Blizard Institute, Queen Mary University of London, E1 2AT, UK.
| |
Collapse
|
14
|
Zavodszky G, Spieker C, Czaja B, van Rooij B. Cellular Blood Flow Modeling with HemoCell. Methods Mol Biol 2024; 2716:351-368. [PMID: 37702948 DOI: 10.1007/978-1-0716-3449-3_16] [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] [Indexed: 09/14/2023]
Abstract
Many of the intriguing properties of blood originate from its cellular nature. Bulk effects, such as viscosity, depend on the local shear rates and on the size of the vessels. While empirical descriptions of bulk rheology are available for decades, their validity is limited to the experimental conditions they were observed under. These are typically artificial scenarios (e.g., perfectly straight glass tube or in pure shear with no gradients). Such conditions make experimental measurements simpler; however, they do not exist in real systems (i.e., in a real human circulatory system). Therefore, as we strive to increase our understanding on the cardiovascular system and improve the accuracy of our computational predictions, we need to incorporate a more comprehensive description of the cellular nature of blood. This, however, presents several computational challenges that can only be addressed by high performance computing. In this chapter, we describe HemoCell ( https://www.hemocell.eu ), an open-source high-performance cellular blood flow simulation, which implements validated mechanical models for red blood cells and is capable of reproducing the emergent transport characteristics of such a complex cellular system. We discuss the accuracy and the range of validity, and demonstrate applications on a series of human diseases.
Collapse
|
15
|
Connes P. Blood rheology and vascular function in sickle cell trait and sickle cell disease: From pathophysiological mechanisms to clinical usefulness. Clin Hemorheol Microcirc 2024; 86:9-27. [PMID: 38073384 DOI: 10.3233/ch-238122] [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] [Indexed: 02/24/2024]
Abstract
Sickle cell disease (SCD) is an autosomal recessive disorder. Although the molecular mechanisms at the origin of SCD have been well characterized, its clinical expression is highly variable. SCD is characterized by blood rheological abnormalities, increased inflammation and oxidative stress, and vascular dysfunction. Individuals with only one copy of the mutated β-globin gene have sickle cell trait (SCT) and are usually asymptomatic. The first part of this review focuses on the biological responses of SCT carriers during exercise and on the effects of combined SCT and diabetes on vascular function, several biomarkers and clinical complications. The second part of the review focuses on SCD and shows that the magnitude of red blood cell (RBC) rheological alterations is highly variable from one patient to another, and this variability reflects the clinical and hematological variability: patients with the less deformable RBCs have high hemolytic rate and severe anemia, and are prone to develop leg ulcers, priapism, cerebral vasculopathy, glomerulopathy or pulmonary hypertension. In contrast, SCD patients characterized by the presence of more deformable RBCs (but still rigid) are less anemic and may exhibit increased blood viscosity, which increases the risk for vaso-occlusive events. Several genetic and cellular factors may modulate RBC deformability in SCD: co-existence of α-thalassemia, fetal hemoglobin level, oxidative stress, the presence of residual mitochondria into mature RBCs, the activity of various non-selective cationic ion channels, etc. The last part of this review presents the effects of hydroxyurea and exercise training on RBC rheology and other biomarkers in SCD.
Collapse
Affiliation(s)
- Philippe Connes
- Laboratory LIBM EA7424, University of Lyon 1, "Vascular Biology and Red Blood Cell" Team, Lyon, France
- Laboratory of Excellence Labex GR-Ex, Paris, France
| |
Collapse
|
16
|
Pandey H, Singh K, Ranjan R, Dass J, Tyagi S, Seth T, Saxena R, Mahapatra M. Prevalence and Impact of HMOX1 Polymorphism (rs2071746: A > T) in Indian Sickle Cell Disease Patients. J Lab Physicians 2023; 15:583-589. [PMID: 37780888 PMCID: PMC10539052 DOI: 10.1055/s-0043-1770068] [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: 06/27/2022] [Accepted: 04/17/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction Fetal hemoglobin (HbF) levels play significant role in lowering down the morbidity and mortality in sickle cell disease (SCD) patients. Coinheritance of heme oxygenase-1 (HMOX1) rs2071746:A > T polymorphism may contribute to variable HbF levels in Indian SCD patients. Objective This study was aimed to evaluate the role of HMOX1 polymorphism and its impact on HbF level in Indian SCD patients. Materials and Methods One-hundred twenty confirmed cases of SCD and 50 healthy controls were recruited. Their mean age was 11.5 ± 8.6 years (range: 3-23 years). Quantification of Hb, HbA2, HbF, and HbS was done by capillary zone electrophoresis. Allele-specific polymerase chain reaction was used to genotype HMOX1 (rs2071746:A > T) gene polymorphism. Results Out of the 120 cases of SCD, 65 were hemoglobin sickle-shaped (HbSS) and 55 were sickle-beta thalassemia (Sβ). Out of 65 HbSS patients, 29 (44.6%) were heterozygous (AT), 20 (30.76%) were homozygous (TT), and 16 (24.61%) were found wild-type (AA) genotype. Out of 55 Sβ, 22 (40%) were heterozygous, 18 (32%) were homozygous and 15 (28%) were wild-type. Patients carrying HMOX1 (rs2071746:A > T), AT, and TT genotypes had less anemia, painful crisis, splenomegaly, hepatomegaly, jaundice, and blood transfusion. HbF level was found higher in TT genotype (in HbSS the HbF levels was 25.1 ± 4.4; in sickle-beta thalassemia the HbF levels was 36.1 ± 4.7) than wild-type(AA) and was statistically significant ( p -value <0.001). Conclusion The TT genotype of the rs2071746:A > T polymorphism was associated with increased levels of Hb F ( p < 0.001). It can serve as a HbF modifier in Indian sickle cell diseases patients.
Collapse
Affiliation(s)
- Hareram Pandey
- Department of Hematology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Kanwaljeet Singh
- Lab Sciences & Molecular Medicine, Army Hospital Research and Referral, Delhi Cantt, Delhi, India
| | - Ravi Ranjan
- Department of Hematology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Jasmita Dass
- Department of Hematology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Seema Tyagi
- Department of Hematology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Tulika Seth
- Department of Hematology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Renu Saxena
- Department of Hematology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Manoranjan Mahapatra
- Department of Hematology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
17
|
Adam M, Musa MJ, Al-Qahtani SM, Alelyani M, Musa A, Elzaki M, Alzain AFH, Ali S, Medani A, Mukhtar EM, Gareeballah A. Ultrasound Imaging in Subjects with Sickle Cell Disease: The Saudi Arabia Experiences. Int J Gen Med 2023; 16:4931-4942. [PMID: 37928952 PMCID: PMC10625387 DOI: 10.2147/ijgm.s419013] [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: 06/04/2023] [Accepted: 10/17/2023] [Indexed: 11/07/2023] Open
Abstract
Background Abdominal organ sonography is a crucial part of the workup for treating sickle cell disease (SCD) patients. Objective The main objective of this study was to evaluate the abdominal organs in SCD patients using ultrasonography. Methodology A non-interventional descriptive cross-sectional study was carried out in Asir region Saudi Arabia from April 2019 to July 2020. The study was conducted in 78 patients with sickle cell disease (SCD). Data were gathered using a data collection sheet included demographic information, clinical information including medication types, and complications linked to SCD. Furthermore, the study evaluated abdominal ultrasound findings pertaining to the liver, gall bladder, spleen, and kidneys. The data were analyzed using Statistical Package for Social Sciences (SPSS). Results More than half of the study participants 43 (55.1%) were females. About 53.8% of the study participants received blood transfusions, and (11.5%) receive extra-vaccine. Concerning ultrasound findings, hepatomegaly was found in seventeen (21.8%), focal liver lesions in four (5.1%), gallstones in five (6.4%), splenomegaly in fifteen (19.3%), and the presence of splenic focal lesions was found in seven (9.0%). The most frequent complication associated with SCD was osteomyelitis sepsis in six cases (7.7%). The study revealed a significant correlation between the type of crisis and type of medication used and the size of the spleen (P-value <0.01), and no notable correlation was found between the types of crises and the size of the liver (P-value >0.05). Conclusion Abdominal sonography in SCD patients revealed a wide range of alterations in the liver, gallbladder, and spleen. The most frequently observed complications in SCD were hepatomegaly, splenomegaly, localized lesions in both organs, and the presence of gallstones.
Collapse
Affiliation(s)
- Mohamed Adam
- Department of Radiological Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Asir, Saudi Arabia
| | - Mustafa J Musa
- Department of Applied Radiologic Science, University of Jeddah, Kingdom of Saudi Arabia (KSA), Jeddah, Saudi Arabia
| | - Saleh M Al-Qahtani
- Department of Child Health, College of Medicine, King Khalid University, Abha, Asir, Saudi Arabia
| | - Magbool Alelyani
- Department of Radiological Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Asir, Saudi Arabia
| | - Alamin Musa
- Department of Radiological Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Asir, Saudi Arabia
| | - Maisa Elzaki
- Department of Diagnostic Radiology Technology, College of Applied Medical Sciences, Taibah University, Al-Madianah Al-Munawwarrah, Saudi Arabia
| | - Amel F H Alzain
- Department of Diagnostic Radiology Technology, College of Applied Medical Sciences, Taibah University, Al-Madianah Al-Munawwarrah, Saudi Arabia
| | - Sarra Ali
- Department of Diagnostic Radiography Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Afaf Medani
- Department of Radiological Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Asir, Saudi Arabia
| | - Emadeldedin Mohamed Mukhtar
- Department of Radiological Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Asir, Saudi Arabia
| | - Awadia Gareeballah
- Department of Diagnostic Radiology Technology, College of Applied Medical Sciences, Taibah University, Al-Madianah Al-Munawwarrah, Saudi Arabia
| |
Collapse
|
18
|
Chauhan W, Zennadi R. Keap1-Nrf2 Heterodimer: A Therapeutic Target to Ameliorate Sickle Cell Disease. Antioxidants (Basel) 2023; 12:antiox12030740. [PMID: 36978988 PMCID: PMC10045360 DOI: 10.3390/antiox12030740] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/04/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Sickle cell disease (SCD) is a monogenic inheritable disease characterized by severe anemia, increased hemolysis, and recurrent, painful vaso-occlusive crises due to the polymerization of hemoglobin S (HbS)-generated oxidative stress. Up until now, only four drugs are approved for SCD in the US. However, each of these drugs affects only a limited array of SCD pathologies. Importantly, curative therapies, such as gene therapy, or hematopoietic stem cell transplantation are not available for every patient because of their high costs, availability of donor matching, and their serious adverse effects. Therefore, there is an unmet medical need for novel therapeutic strategies that target broader SCD sequelae. SCD phenotypic severity can be alleviated by increasing fetal hemoglobin (HbF) expression. This results in the inhibition of HbS polymerization and thus sickling, and a reduction in oxidative stress. The efficacy of HbF is due to its ability to dilute HbS levels below the threshold required for polymerization and to influence HbS polymer stability in RBCs. Nuclear factor-E2-related factor 2 (Nrf2)/Kelch-like ECH-associated protein-1 (Keap1)-complex signaling is one of the most important cytoprotective signaling controlling oxidative stress. Nrf2 is present in most organs and, after dissociation from Keap1, it accumulates in the cytoplasm, then translocates to the nucleus where it binds to the antioxidant response element (ARE) sequences and increases the expression of various cytoprotective antioxidant genes. Keeping this in mind, various researchers have proposed a role of multiple agents, more importantly tert-Butylhydroquinone (tBHQ), curcumin, etc., (having electrophilic properties) in inhibiting keap1 activity, so that Nrf2 can translocate to the nucleus to activate the gamma globin gene, thus maintaining alpha-hemoglobin-stabilizing protein (AHSP) and HbF levels. This leads to reduced oxidative stress, consequently minimizing SCD-associated complications. In this review, we will discuss the role of the Keap-1–Nrf2 complex in hemoglobinopathies, especially in SCD, and how this complex might represent a better target for more effective treatment options.
Collapse
|
19
|
Allison RL, Burand A, Torres DN, Brandow AM, Stucky CL, Ebert AD. Sickle cell disease patient plasma sensitizes iPSC-derived sensory neurons from sickle cell disease patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.10.523446. [PMID: 36711992 PMCID: PMC9882050 DOI: 10.1101/2023.01.10.523446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Individuals living with sickle cell disease (SCD) experience severe recurrent acute and chronic pain. In order to develop novel therapies, it is necessary to better understand the neurobiological mechanisms underlying SCD pain. There are many barriers to gaining mechanistic insight into pathogenic SCD pain processes, such as differential gene expression and function of sensory neurons between humans and mice with SCD, as well as the limited availability of patient samples. These can be overcome by utilizing SCD patient-derived induced pluripotent stem cells (iPSCs) differentiated into sensory neurons (SCD iSNs). Here, we characterize the key gene expression and function of SCD iSNs to establish a model for higher-throughput investigation of intrinsic and extrinsic factors that may contribute to increased SCD patient pain. Importantly, identified roles for C-C Motif Chemokine Ligand 2 (CCL2) and endothelin 1 (ET1) in SCD pain can be recapitulated in SCD iSNs. Further, we find that plasma taken from SCD patients during acute pain increases SCD iSN calcium response to the nociceptive stimulus capsaicin compared to those treated with paired SCD patient plasma at baseline or healthy control plasma samples. Together, these data provide the framework necessary to utilize iSNs as a powerful tool to investigate the neurobiology of SCD and identify potential intrinsic mechanisms of SCD pain which may extend beyond a blood-based pathology.
Collapse
Affiliation(s)
- Reilly L. Allison
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Anthony Burand
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Damaris Nieves Torres
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - Amanda M. Brandow
- Department of Pediatrics, Section of Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI
| | - Cheryl L. Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
20
|
Gil GP, Ananina G, Maschietto M, Lima SCS, da Silva Costa SM, Baptista LDC, Ito MT, Costa FF, Costa ML, de Melo MB. Epigenetic analysis in placentas from sickle cell disease patients reveals a hypermethylation profile. PLoS One 2022; 17:e0274762. [PMID: 36129958 PMCID: PMC9491616 DOI: 10.1371/journal.pone.0274762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/05/2022] [Indexed: 11/25/2022] Open
Abstract
Pregnancy in Sickle Cell Disease (SCD) women is associated to increased risk of clinical and obstetrical complications. Placentas from SCD pregnancies can present increased abnormal findings, which may lead to placental insufficiency, favoring adverse perinatal outcome. These placental abnormalities are well known and reported, however little is known about the molecular mechanisms, such as epigenetics. Thus, our aim was to evaluate the DNA methylation profile in placentas from women with SCD (HbSS and HbSC genotypes), compared to uncomplicated controls (HbAA). We included in this study 11 pregnant women with HbSS, 11 with HbSC and 21 with HbAA genotypes. Illumina Methylation EPIC BeadChip was used to assess the whole placental DNA methylation. Pyrosequencing was used for array data validation and qRT-PCR was applied for gene expression analysis. Our results showed high frequency of hypermethylated CpGs sites in HbSS and HbSC groups with 73.5% and 76.2% respectively, when compared with the control group. Differentially methylated regions (DMRs) also showed an increased hypermethylation status for the HbSS (89%) and HbSC (86%) groups, when compared with the control group methylation data. DMRs were selected for methylation validation (4 DMRs-HbSS and 3 DMRs the HbSC groups) and after analyses three were validated in the HbSS group, and none in the HbSC group. The gene expression analysis showed differential expression for the PTGFR (-2.97-fold) and GPR56 (3.0-fold) genes in the HbSS group, and for the SPOCK1 (-2.40-fold) and ADCY4 (1.80-fold) genes in the HbSC group. Taken together, these data strongly suggest that SCD (HbSS and HbSC genotypes) can alter placental DNA methylation and lead to gene expression changes. These changes possibly contribute to abnormal placental development and could impact in the clinical course, especially for the fetus, possibly leading to increased risk of abortion, fetal growth restriction (FGR), stillbirth, small for gestational age newborns and prematurity.
Collapse
Affiliation(s)
- Gislene Pereira Gil
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Galina Ananina
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | | | | | - Sueli Matilde da Silva Costa
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Leticia de Carvalho Baptista
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Mirta Tomie Ito
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | | | - Maria Laura Costa
- Department of Obstetrics and Gynecology, University of Campinas, Campinas, São Paulo, Brazil
| | - Mônica Barbosa de Melo
- Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
21
|
Ibrahim A, Muhammad SA. Antioxidant-Rich Nutraceutical as a Therapeutic Strategy for Sickle Cell Disease. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2022:1-10. [PMID: 36069788 DOI: 10.1080/27697061.2022.2108930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 07/23/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Sickle cell disease (SCD) is a genetically inherited disease in which the "SS" individual possesses two copies of the abnormal beta-globin gene. This disease is one of the most dominant genetic diseases in the world. SCD is marked by the propensity of red cell hemoglobin to polymerize and distort the red cell from a biconcave disk shape into a sickle shape, resulting in a typical vaso-occlusive episode and accelerated hemolysis. Plants are rich sources of bioactive compounds that are promising anti-sickling agents to scavenge free radicals, thereby ensuring oxidative balance. The current review highlights the potential therapeutic benefits of antioxidant-rich nutraceutical in the treatment and management of sickle cell disease. The anti-sickling potential of nutraceutical is attributed to the presence of antioxidant bioactive chemicals such as alkaloids, polyphenols, vitamins, and minerals, which acts as scavengers of free radicals that prevent oxidative damage of the hemoglobin and prevent hemolysis, facilitating longer erythrocyte lifespan. The challenges of current therapies for SCD and future directions are also discussed.KEY TEACHING POINTSSickle cell disease is a genetically inherited disease in which SS individuals possess two copies of the abnormal beta-globin gene.Oxidative stress contributes to the pathophysiology of secondary dysfunction in sickle cell patients.Antioxidants can play a vital role in maintaining a balance between oxidant and antioxidant defense systems.Nutraceutical rich in antioxidants such as alkaloids, polyphenols, vitamins, and minerals is potential therapeutic agents for sickle cell disease.An antioxidant-rich nutraceutical may act to reduce vaso-occlusive crises.
Collapse
Affiliation(s)
- Abdulwasiu Ibrahim
- Department of Biochemistry and Molecular Biology, Usmanu Danfodiyo University, Sokoto, Nigeria
| | | |
Collapse
|
22
|
Lamarre Y, Nader E, Connes P, Romana M, Garnier Y. Extracellular Vesicles in Sickle Cell Disease: A Promising Tool. Bioengineering (Basel) 2022; 9:bioengineering9090439. [PMID: 36134985 PMCID: PMC9495982 DOI: 10.3390/bioengineering9090439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 12/12/2022] Open
Abstract
Sickle cell disease (SCD) is the most common hemoglobinopathy worldwide. It is characterized by an impairment of shear stress-mediated vasodilation, a pro-coagulant, and a pro-adhesive state orchestrated among others by the depletion of the vasodilator nitric oxide, by the increased phosphatidylserine exposure and tissue factor expression, and by the increased interactions of erythrocytes with endothelial cells that mediate the overexpression of adhesion molecules such as VCAM-1, respectively. Extracellular vesicles (EVs) have been shown to be novel actors involved in SCD pathophysiological processes. Medium-sized EVs, also called microparticles, which exhibit increased plasma levels in this pathology, were shown to induce the activation of endothelial cells, thereby increasing neutrophil adhesion, a key process potentially leading to the main complication associated with SCD, vaso-occlusive crises (VOCs). Small-sized EVs, also named exosomes, which have also been reported to be overrepresented in SCD, were shown to potentiate interactions between erythrocytes and platelets, and to trigger endothelial monolayer disruption, two processes also known to favor the occurrence of VOCs. In this review we provide an overview of the current knowledge about EVs concentration and role in SCD.
Collapse
Affiliation(s)
- Yann Lamarre
- Université Paris Cité and Université des Antilles, Inserm, BIGR, F-75015 Paris, France
| | - Elie Nader
- Laboratoire Inter-Universitaire de Biologie de la Motricité EA7424, Team “Vascular Biology and Red Blood Cell”, Université Claude Bernard Lyon 1, Université de Lyon, 69622 Lyon, France
| | - Philippe Connes
- Laboratoire Inter-Universitaire de Biologie de la Motricité EA7424, Team “Vascular Biology and Red Blood Cell”, Université Claude Bernard Lyon 1, Université de Lyon, 69622 Lyon, France
| | - Marc Romana
- Université Paris Cité and Université des Antilles, Inserm, BIGR, F-75015 Paris, France
| | - Yohann Garnier
- Université Paris Cité and Université des Antilles, Inserm, BIGR, F-75015 Paris, France
- Correspondence: ; Tel.: +590-590-891530
| |
Collapse
|
23
|
Elemo GN, Erukainure OL, Okafor JNC, Banerjee P, Preissner R, Nwachukwu Nicholas-Okpara VA, Atolani O, Omowunmi O, Ezeanyanaso CS, Awosika A, Shode F. Underutilized legumes, Cajanus cajan and Glycine max may bring about antisickling effect in sickle cell disease by modulation of redox homeostasis in sickled erythrocytes and alteration of its functional chemistry. J Food Biochem 2022; 46:e14322. [PMID: 35894096 DOI: 10.1111/jfbc.14322] [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: 01/09/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 11/28/2022]
Abstract
The antisickling and anti-oxidative effect of the Cajanus cajan, Glycine max, and their blends were investigated in sickled erythrocytes. The powdered samples were analyzed for their nutritional and anti-nutritional constituents. Their aqueous extracts were analyzed for in vitro antioxidant activities. The extracts were incubated with sickled erythrocytes at 37°C for 6 hours and the antisickling effect examined via microscopic analysis. The blend was the most active and its incubated cells were subjected to anti-oxidative analysis which covers for GSH, SOD, catalase, and lipid peroxidation (LPO). Chemical functional group of the treated cells was analyzed with FTIR spectroscopy. The in silico binding of the predominant amino acid to hemoglobin was also investigated. An increased concentration of leucine was observed in the blend compared to that of C. cajan and G. max, respectively. Vitamins C, B6, and B9 were the only vitamins observed in the blend. Phytate and oxalate were present in all samples. All extracts displayed significant (p < .05) scavenging activities. Treatment with the blend exacerbated SOD and catalase activities as well as the GSH level, while suppressing LPO. FTIR analysis of the treated cells showed the presence of hydrophobic functional groups. Leucine was the predominant amino acid, and it showed a potent molecular interaction with HIS-87 residue of the alpha chain of 1HCO. C. cajan and G. max blend inhibited sickling activities of sickle erythrocytes, while concomitantly exacerbating their endogenous antioxidant enzymes activity and modification of the functional chemistry. PRACTICAL APPLICATIONS: Cajanus cajan and Glycine max are among the common underutilized legumes in Nigeria. Aside their nutritional properties, these legumes have been used from time immemorial for the treatment and management of various ailments. Sickle cell anemia is a class of hemoglobinopathy common in Sub-Saharan Africa. There have been concerns about its treatment owing to the increasing scourge of the disease coupled to the financial burden of its management. This study reports the ability of the potentials of the legumes to prevent sickling activities of sickled erythrocytes and the possible biochemical mechanism involved.
Collapse
Affiliation(s)
- Gloria N Elemo
- Nutraceutical Laboratories, Nutrition and Toxicology Division, Federal Institute of Industrial Research, Lagos, Nigeria.,Department of Chemical Sciences, Ajayi Crowther University, Oyo, Nigeria
| | - Ochuko L Erukainure
- Department of Pharmacology, University of the Free State, Bloemfontein, South Africa
| | - Jane N C Okafor
- Nutraceutical Laboratories, Nutrition and Toxicology Division, Federal Institute of Industrial Research, Lagos, Nigeria
| | - Priyanka Banerjee
- Structural Bioinformatics Group, Institute for Physiology, Charité-University Medicine Berlin, Berlin, Germany
| | - Robert Preissner
- Structural Bioinformatics Group, Institute for Physiology, Charité-University Medicine Berlin, Berlin, Germany
| | | | | | - Olusola Omowunmi
- Laboratory Management & Services, Federal Institute of Industrial Research, Lagos, Nigeria
| | - Chika S Ezeanyanaso
- Polymer & Textile Division, Federal Institute of Industrial Research, Lagos, Nigeria
| | | | - Francis Shode
- Department of Biotechnology and Food Science, Durban University of Technology, Durban, South Africa.,Sholab Nutraceuticals (Pty) Ltd, Westville North, South Africa
| |
Collapse
|
24
|
Nemkov T, Skinner S, Diaw M, Diop S, Samb A, Connes P, D’Alessandro A. Plasma Levels of Acyl-Carnitines and Carboxylic Acids Correlate With Cardiovascular and Kidney Function in Subjects With Sickle Cell Trait. Front Physiol 2022; 13:916197. [PMID: 35910560 PMCID: PMC9326174 DOI: 10.3389/fphys.2022.916197] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Subjects with sickle cell trait (SCT) carry one copy of mutated β-globin gene at position E6V at the origin of the production of sickle hemoglobin (HbS). Indeed, individuals with SCT have both normal hemoglobin and HbS, in contrast to patients with sickle cell disease who inherited of two copies of the mutated gene. Although SCT is generally benign/asymptomatic, carriers may develop certain adverse outcomes such as renal complications, venous thromboembolism, exercise-induced rhabdomyolysis … However, little is known about whether similar metabolic pathways are affected in individuals with SCT and whether these metabolic derangements, if present, correlate to clinically relevant parameters. In this study, we performed metabolomics analysis of plasma from individuals with sickle cell trait (n = 34) compared to healthy controls (n = 30). Results indicated a significant increase in basal circulating levels of hemolysis markers, mono- (pyruvate, lactate), di- and tri-carboxylates (including all Krebs cycle intermediates), suggestive of systems-wide mitochondrial dysfunction in individuals with SCT. Elevated levels of kynurenines and indoles were observed in SCT samples, along with increases in the levels of oxidative stress markers (advanced glycation and protein-oxidation end-products, malondialdehyde, oxylipins, eicosanoids). Increases in circulating levels of acyl-carnitines and fatty acids were observed, consistent with increased membrane lipid damage in individuals with sickle cell trait. Finally, correlation analyses to clinical co-variates showed that alterations in the aforementioned pathways strongly correlated with clinical measurements of blood viscosity, renal (glomerular filtration rate, microalbuminuria, uremia) and cardiovascular function (carotid-femoral pulse wave velocity, blood pressure).
Collapse
Affiliation(s)
- Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| | - Sarah Skinner
- Inter-university Laboratory of Biology of Motor Function EA7424, Vascular Biology and the Red Blood Cell Team, Claude Bernard University Lyon 1, Lyon, France
| | - Mor Diaw
- Laboratory of Physiology and Functional Exploration, FMPO, UCAD, Dakar, Senegal
- IRL3189 Environnement, Santé, Sociétés CNRS/UCAD Dakar/ UGB Saint-Louis/ USTT Bamako/ CNRST Ouagadougou, Dakar, Senegal
| | - Saliou Diop
- Laboratory of Hemato-immunology, FMPO, UCAD, Dakar, Senegal
| | - Abdoulaye Samb
- Laboratory of Physiology and Functional Exploration, FMPO, UCAD, Dakar, Senegal
- IRL3189 Environnement, Santé, Sociétés CNRS/UCAD Dakar/ UGB Saint-Louis/ USTT Bamako/ CNRST Ouagadougou, Dakar, Senegal
| | - Philippe Connes
- Inter-university Laboratory of Biology of Motor Function EA7424, Vascular Biology and the Red Blood Cell Team, Claude Bernard University Lyon 1, Lyon, France
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| |
Collapse
|
25
|
Lopez NH, Li B, Palani C, Siddaramappa U, Takezaki M, Xu H, Zhi W, Pace BS. Salubrinal induces fetal hemoglobin expression via the stress-signaling pathway in human sickle erythroid progenitors and sickle cell disease mice. PLoS One 2022; 17:e0261799. [PMID: 35639781 PMCID: PMC9154101 DOI: 10.1371/journal.pone.0261799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/05/2022] [Indexed: 11/25/2022] Open
Abstract
Sickle cell disease (SCD) is an inherited blood disorder caused by a mutation in the HBB gene leading to hemoglobin S production and polymerization under hypoxia conditions leading to vaso-occlusion, chronic hemolysis, and progressive organ damage. This disease affects ~100,000 people in the United States and millions worldwide. An effective therapy for SCD is fetal hemoglobin (HbF) induction by pharmacologic agents such as hydroxyurea, the only Food and Drug Administration-approved drug for this purpose. Therefore, the goal of our study was to determine whether salubrinal (SAL), a selective protein phosphatase 1 inhibitor, induces HbF expression through the stress-signaling pathway by activation of p-eIF2α and ATF4 trans-activation in the γ-globin gene promoter. Sickle erythroid progenitors treated with 24μM SAL increased F-cells levels 1.4-fold (p = 0.021) and produced an 80% decrease in reactive oxygen species. Western blot analysis showed SAL enhanced HbF protein by 1.6-fold (p = 0.0441), along with dose-dependent increases of p-eIF2α and ATF4 levels. Subsequent treatment of SCD mice by a single intraperitoneal injection of SAL (5mg/kg) produced peak plasma concentrations at 6 hours. Chronic treatments of SCD mice with SAL mediated a 2.3-fold increase in F-cells (p = 0.0013) and decreased sickle erythrocytes supporting in vivo HbF induction.
Collapse
Affiliation(s)
- Nicole H. Lopez
- Department of Biochemistry and Cancer Biology, Augusta University, Augusta, GA, United States of America
| | - Biaoru Li
- Department of Pediatrics, Augusta University, Augusta, GA, United States of America
| | - Chithra Palani
- Department of Pediatrics, Augusta University, Augusta, GA, United States of America
| | - Umapathy Siddaramappa
- Department of Medicine, Division of Hematology/Oncology Augusta University, Augusta GA, United States of America
| | - Mayuko Takezaki
- Department of Pediatrics, Augusta University, Augusta, GA, United States of America
| | - Hongyan Xu
- Department of Biostatistics and Epidemiology, Augusta University, Augusta, GA, United States of America
| | - Wenbo Zhi
- Center for Biotechnology & Genomic Medicine, Augusta University, Augusta, GA, United States of America
| | - Betty S. Pace
- Department of Biochemistry and Cancer Biology, Augusta University, Augusta, GA, United States of America
- Department of Pediatrics, Augusta University, Augusta, GA, United States of America
| |
Collapse
|
26
|
Bou-Fakhredin R, De Franceschi L, Motta I, Eid AA, Taher AT, Cappellini MD. Redox Balance in β-Thalassemia and Sickle Cell Disease: A Love and Hate Relationship. Antioxidants (Basel) 2022; 11:antiox11050967. [PMID: 35624830 PMCID: PMC9138068 DOI: 10.3390/antiox11050967] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
β-thalassemia and sickle cell disease (SCD) are inherited hemoglobinopathies that result in both quantitative and qualitative variations in the β-globin chain. These in turn lead to instability in the generated hemoglobin (Hb) or to a globin chain imbalance that affects the oxidative environment both intracellularly and extracellularly. While oxidative stress is not among the primary etiologies of β-thalassemia and SCD, it plays a significant role in the pathogenesis of these diseases. Different mechanisms exist behind the development of oxidative stress; the result of which is cytotoxicity, causing the oxidation of cellular components that can eventually lead to cell death and organ damage. In this review, we summarize the mechanisms of oxidative stress development in β-thalassemia and SCD and describe the current and potential antioxidant therapeutic strategies. Finally, we discuss the role of targeted therapy in achieving an optimal redox balance.
Collapse
Affiliation(s)
- Rayan Bou-Fakhredin
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy; (R.B.-F.); (I.M.)
| | - Lucia De Franceschi
- Department of Medicine, University of Verona, and Azienda Ospedaliera Universitaria Verona, 37128 Verona, Italy;
| | - Irene Motta
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy; (R.B.-F.); (I.M.)
- UOC General Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Assaad A. Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon;
| | - Ali T. Taher
- Division of Hematology-Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon;
| | - Maria Domenica Cappellini
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy; (R.B.-F.); (I.M.)
- UOC General Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Correspondence:
| |
Collapse
|
27
|
Adeniyi O, Baptista R, Bhowmick S, Cookson A, Nash RJ, Winters A, Shen J, Mur LAJ. Isolation and Characterisation of Quercitrin as a Potent Anti-Sickle Cell Anaemia Agent from Alchornea cordifolia. J Clin Med 2022; 11:jcm11082177. [PMID: 35456270 PMCID: PMC9024604 DOI: 10.3390/jcm11082177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 02/02/2023] Open
Abstract
Alchornea cordifolia Müll. Arg. (commonly known as Christmas Bush) has been used traditionally in Africa to treat sickle cell anaemia (a recessive disease, arising from the S haemoglobin (Hb) allele), but the active compounds are yet to be identified. Herein, we describe the use of sequential fractionation coupled with in vitro anti-sickling assays to purify the active component. Sickling was induced in HbSS genotype blood samples using sodium metabisulphite (Na2S2O5) or through incubation in 100% N2. Methanol extracts of A. cordifolia leaves and its sub-fractions showed >70% suppression of HbSS erythrocyte sickling. The purified compound demonstrated a 87.2 ± 2.39% significant anti-sickling activity and 93.1 ± 2.69% erythrocyte sickling-inhibition at 0.4 mg/mL. Nuclear magnetic resonance (NMR) spectra and high-resolution mass spectroscopy identified it as quercitrin (quercetin 3-rhamnoside). Purified quercitrin also inhibited the polymerisation of isolated HbS and stabilized sickle erythrocytes membranes. Metabolomic comparisons of blood samples using flow-infusion electrospray-high resolution mass spectrometry indicated that quercitrin could convert HbSS erythrocyte metabolomes to be like HbAA. Sickling was associated with changes in antioxidants, anaerobic bioenergy, and arachidonic acid metabolism, all of which were reversed by quercitrin. The findings described could inform efforts directed to the development of an anti-sickling drug or quality control assessments of A. cordifolia preparations.
Collapse
Affiliation(s)
- Olayemi Adeniyi
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (O.A.); (R.B.); (S.B.); (A.C.); (A.W.)
- Biochemistry Unit, Department of Science Technology, The Federal Polytechnic, Ado-Ekiti 360231, Nigeria
| | - Rafael Baptista
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (O.A.); (R.B.); (S.B.); (A.C.); (A.W.)
| | - Sumana Bhowmick
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (O.A.); (R.B.); (S.B.); (A.C.); (A.W.)
| | - Alan Cookson
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (O.A.); (R.B.); (S.B.); (A.C.); (A.W.)
| | - Robert J. Nash
- PhytoQuest Ltd., Plas Gogerddan, Aberystwyth SY23 3EB, UK;
| | - Ana Winters
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (O.A.); (R.B.); (S.B.); (A.C.); (A.W.)
| | - Jianying Shen
- Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Correspondence: (J.S.); (L.A.J.M.)
| | - Luis A. J. Mur
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (O.A.); (R.B.); (S.B.); (A.C.); (A.W.)
- Correspondence: (J.S.); (L.A.J.M.)
| |
Collapse
|
28
|
Zinc, Magnesium, and Copper Levels in Patients with Sickle Cell Disease: A Systematic Review and Meta-analysis. Avicenna J Med 2022; 12:45-53. [PMID: 35833156 PMCID: PMC9272455 DOI: 10.1055/s-0042-1749612] [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] [Indexed: 11/18/2022] Open
Abstract
Background
Sickle cell disease (SCD) is associated with oxidative stress due to an imbalance between production and elimination of the reactive oxygen species. It has been reported that SCD patients are at risk of multiple micronutrients' deficiencies, including several trace elements involved in the antioxidation mechanisms. We aimed to assess the status of these micronutrients in SCD patients.
Methods
This study was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. The databases of MedLine, Embase, and PsycInfo were used for the systematic search from time the databases existed until April 2021. A total of 36 studies fulfilled the eligibility criteria. We calculated the pooled standardized mean difference (SMD) of serum zinc, magnesium, or copper levels among patients with SCD and their healthy controls.
Results
SCD patients had significantly lower zinc (SMD = −1.27 [95% CI: 1.67−0.87,
p
0.001]) and magnesium levels (SMD = −0.53 [95% CI: 1.0−0.06,
p
0.026] than their controls. Copper level was found to be significantly higher in SCD patients, with SMD = 0.68 (95% CI: 0.05−1.32,
p
0.004).
Conclusion
This review showed that SCD patients may potentially prompt to have lower zinc and magnesium levels and higher copper levels compared with those without the disease. Future research need to be directed to investigate clinical outcome of nutritional difficiencies in patients with SCD, as well as the possibility of implementing nutritional supplement programs which may help minimizing the harmful effects of the disease on human body.
Collapse
|
29
|
Reduced blood pressure in sickle cell disease is associated with decreased angiotensin converting enzyme (ACE) activity and is not modulated by ACE inhibition. PLoS One 2022; 17:e0263424. [PMID: 35113975 PMCID: PMC8812860 DOI: 10.1371/journal.pone.0263424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/18/2022] [Indexed: 12/11/2022] Open
Abstract
Background Sickle cell disease (SCD) incurs vaso-occlusive episodes and organ damage, including nephropathy. Despite displaying characteristics of vascular dysfunction, SCD patients tend to present relatively lower systemic blood pressure (BP), via an unknown mechanism. We investigated associations between BP and renin-angiotensin-system (RAS) components in SCD and determined whether an inhibitor of angiotensin converting enzyme (ACE; often used to slow SCD glomerulopathy) further modulates BP and RAS components in a murine model of SCD. Methods BP was compared in human subjects and mice with/without SCD. Plasma angiotensin II, ACE and renin were measured by immunoassay. BP was reevaluated after treating mice with enalapril (25 mg/kg, 5x/week) for 5 weeks; plasma and organs were stored for angiotensin II and ACE activity measurement, and quantitative real-time PCR. Results Diastolic BP and systolic BP were significantly lower in patients and mice with SCD, respectively, compared to controls. Reduced BP was associated with increased plasma renin and markers of kidney damage (mice) in SCD, as well as significantly decreased plasma ACE concentrations and ACE enzyme activity. As expected, enalapril administration lowered BP, plasma angiotensin II and organ ACE activity in control mice. In contrast, enalapril did not further reduce BP or organ ACE activity in SCD mice; however, plasma angiotensin II and renin levels were found to be significantly higher in enalapril-treated SCD mice than those of treated control mice. Conclusion Relative hypotension was confirmed in a murine model of SCD, in association with decreased ACE concentrations in both human and murine disease. Given that ACE inhibition has an accepted role in decreasing BP, further studies should investigate mechanisms by which ACE depletion, via both Ang II-dependent and alternative pathways, could contribute to reduce BP in SCD and understand how ACE inhibition confers Ang II-independent benefits on kidney function in SCD.
Collapse
|
30
|
Dosunmu-Ogunbi A, Yuan S, Shiwarski DJ, Tashman JW, Reynolds M, Feinberg A, Novelli EM, Shiva S, Straub AC. Endothelial superoxide dismutase 2 is decreased in sickle cell disease and regulates fibronectin processing. FUNCTION (OXFORD, ENGLAND) 2022; 3:zqac005. [PMID: 35274104 PMCID: PMC8900267 DOI: 10.1093/function/zqac005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/05/2022] [Accepted: 02/10/2022] [Indexed: 01/07/2023]
Abstract
Sickle cell disease (SCD) is a genetic red blood cell disorder characterized by increased reactive oxygen species (ROS) and a concordant reduction in antioxidant capacity in the endothelium. Superoxide dismutase 2 (SOD2) is a mitochondrial-localized enzyme that catalyzes the dismutation of superoxide to hydrogen peroxide. Decreased peripheral blood expression of SOD2 is correlated with increased hemolysis and cardiomyopathy in SCD. Here, we report for the first time that endothelial cells exhibit reduced SOD2 protein expression in the pulmonary endothelium of SCD patients. To investigate the impact of decreased SOD2 expression in the endothelium, SOD2 was knocked down in human pulmonary microvascular endothelial cells (hPMVECs). We found that SOD2 deficiency in hPMVECs results in endothelial cell dysfunction, including reduced cellular adhesion, diminished migration, integrin protein dysregulation, and disruption of permeability. Furthermore, we uncover that SOD2 mediates changes in endothelial cell function via processing of fibronectin through its inability to facilitate dimerization. These results demonstrate that endothelial cells are deficient in SOD2 expression in SCD patients and suggest a novel pathway for SOD2 in regulating fibronectin processing.
Collapse
Affiliation(s)
- Atinuke Dosunmu-Ogunbi
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, 15261, Pittsburgh, PA, USA,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 15261, Pittsburgh, PA, USA,Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | - Shuai Yuan
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | - Daniel J Shiwarski
- Department of Biomedical Engineering, Carnegie Mellon University, 15261, Pittsburgh, PA, USA
| | - Joshua W Tashman
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, 15261, Pittsburgh, PA, USA,Department of Biomedical Engineering, Carnegie Mellon University, 15261, Pittsburgh, PA, USA
| | - Michael Reynolds
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | - Adam Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, 15261, Pittsburgh, PA, USA,Department of Materials Science and Engineering, Carnegie Mellon University, 15261, Pittsburgh, PA, USA
| | - Enrico M Novelli
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 15261, Pittsburgh, PA, USA,Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 15261, Pittsburgh, PA, USA,Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, 15261, Pittsburgh, PA, USA
| | | |
Collapse
|
31
|
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
|
32
|
Investigation of the relationship between periodontal and systemic inflammation in children with Sickle Cell Disease: A case- control study. Cytokine 2021; 149:155724. [PMID: 34653827 DOI: 10.1016/j.cyto.2021.155724] [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: 02/01/2021] [Revised: 06/26/2021] [Accepted: 09/30/2021] [Indexed: 11/20/2022]
Abstract
Periodontal diseases are chronic inflammatory diseases and tissue destruction increases with oxidative stress in periodontal tissues. Periodontal diseases are associated with systemic diseases such as diabetes, cardio-vascular diseases and rheumatoid arthritis by means of systemic inflammation. Sickle cell disease (SCD) is a chronic inflammatory disease in which vaso-occlusive crisis and endothelial dysfunction are present. It is not known whether the chronic systemic inflammation seen in SCD affect periodontal tissues. The aim of this study was to investigate the relationship between periodontal and systemic inflammation in children with SCD. Forty-three children with SCD and 43 healthy children were included in the study. Physical, dental and periodontal statuses were examined, blood and saliva samples were taken. Levels of pro-inflammatory and oxidative stress mediators in serum and saliva were evaluated. The periodontal findings of the groups were similar. The majority of the subjects in both groups had gingival inflammation. In SCD group, significantly higher serum high sensitive C-reactive protein (Hs-CRP), interleukin (IL)-6, IL-8, tumor necrosis factor (TNF)-α, total oxidant status (TOS), nitric oxide (NO) and salivary IL-6 were observed (p < 0.05). There were positive correlations between salivary IL-6 levels and serum Hs-CRP levels (r = 0.303, p < 0.05). In addition; it was determined that salivary IL-6, TNF-α and NO levels were increased 3-6 times in children with a history of painful crisis or acute chest syndrome compared to children who had never had a painful crisis or acute chest syndrome. Although, observed oral health status was similar in both groups, salivary cytokine levels were increased in children with SCD. The higher salivary cytokine levels may be associated with chronic systemic inflammation and vaso-occlusion observed in children with SCD.
Collapse
|
33
|
Prasad KN, Bondy SC. Can a Micronutrient Mixture Delay the Onset and Progression of Symptoms of Single-Point Mutation Diseases? J Am Coll Nutr 2021; 41:489-498. [PMID: 34227926 DOI: 10.1080/07315724.2021.1910592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Single-point mutation diseases in which substitution of one nucleotide with another in a gene occurs include familial Alzheimer's disease (fAD), familial Parkinson's disease (fPD), and familial Creutzfeldt-Jacob disease (fCJD) as well as Huntington's disease (HD), sickle cell anemia, and hemophilia. Inevitability of occurrence of these diseases is certain. However, the time of appearance of symptoms could be influenced by the diet, environment, and possibly other genetic factors. There are no effective approaches to delay the onset or progression of symptoms of these diseases. The fact that increased oxidative stress and inflammation significantly contribute to the initiation and progression of these point mutation diseases shows that antioxidants could be useful. The major objectives are (a) to present evidence that increased oxidative stress and chronic inflammation are associated with selected single-point mutation diseases, such as fAD, fPD, and fCJD, HD, sickle cell anemia, and hemophilia; (b) to describe limited studies on the role of individual antioxidants in experimental models of some of these diseases; and (c) to discuss a rationale for utilizing a comprehensive mixture of micronutrients, which may delay the development and progression of symptoms of above diseases by simultaneously reducing oxidative and inflammatory damages.Key teaching pointsSelected single-point mutation diseases and their pattern of inheritanceCharacteristics of each selected single-point mutation diseaseEvidence for increased oxidative stress and inflammation in each diseasePotential reasons for failure of single antioxidants in human studiesRationale for using a comprehensive mixture of micronutrients in delaying the onset and progression of single-point mutation diseases.
Collapse
Affiliation(s)
| | - Stephen C Bondy
- Department of Occupational and Environmental Medicine and Department of Medicine, University of California Irvine, Irvine, California, USA
| |
Collapse
|
34
|
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: 11] [Impact Index Per Article: 2.8] [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
|
35
|
Cao H, Antonopoulos A, Henderson S, Wassall H, Brewin J, Masson A, Shepherd J, Konieczny G, Patel B, Williams ML, Davie A, Forrester MA, Hall L, Minter B, Tampakis D, Moss M, Lennon C, Pickford W, Erwig L, Robertson B, Dell A, Brown GD, Wilson HM, Rees DC, Haslam SM, Alexandra Rowe J, Barker RN, Vickers MA. Red blood cell mannoses as phagocytic ligands mediating both sickle cell anaemia and malaria resistance. Nat Commun 2021; 12:1792. [PMID: 33741926 PMCID: PMC7979802 DOI: 10.1038/s41467-021-21814-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
In both sickle cell disease and malaria, red blood cells (RBCs) are phagocytosed in the spleen, but receptor-ligand pairs mediating uptake have not been identified. Here, we report that patches of high mannose N-glycans (Man5-9GlcNAc2), expressed on diseased or oxidized RBC surfaces, bind the mannose receptor (CD206) on phagocytes to mediate clearance. We find that extravascular hemolysis in sickle cell disease correlates with high mannose glycan levels on RBCs. Furthermore, Plasmodium falciparum-infected RBCs expose surface mannose N-glycans, which occur at significantly higher levels on infected RBCs from sickle cell trait subjects compared to those lacking hemoglobin S. The glycans are associated with high molecular weight complexes and protease-resistant, lower molecular weight fragments containing spectrin. Recognition of surface N-linked high mannose glycans as a response to cellular stress is a molecular mechanism common to both the pathogenesis of sickle cell disease and resistance to severe malaria in sickle cell trait.
Collapse
Affiliation(s)
- Huan Cao
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | | | - Sadie Henderson
- grid.476695.f0000 0004 0495 4557Scottish National Blood Transfusion Service, Aberdeen, UK
| | - Heather Wassall
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - John Brewin
- grid.46699.340000 0004 0391 9020Department of Haematology, King’s College Hospital, London, UK
| | - Alanna Masson
- grid.417581.e0000 0000 8678 4766Department of Haematology, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Jenna Shepherd
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Gabriela Konieczny
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Bhinal Patel
- grid.7445.20000 0001 2113 8111Department of Life Sciences, Imperial College London, London, UK
| | - Maria-Louise Williams
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Adam Davie
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Megan A. Forrester
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Lindsay Hall
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Beverley Minter
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Dimitris Tampakis
- grid.13097.3c0000 0001 2322 6764Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University and Division of Cancer Studies, King’s College London, London, UK
| | - Michael Moss
- grid.476695.f0000 0004 0495 4557Scottish National Blood Transfusion Service, Aberdeen, UK
| | - Charlotte Lennon
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Wendy Pickford
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Lars Erwig
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Beverley Robertson
- grid.7445.20000 0001 2113 8111Department of Life Sciences, Imperial College London, London, UK
| | - Anne Dell
- grid.46699.340000 0004 0391 9020Department of Haematology, King’s College Hospital, London, UK
| | - Gordon D. Brown
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK ,grid.8391.30000 0004 1936 8024Medical Medical Research Council Centre for Medical Mycology at the University of Exeter, Exeter, UK
| | - Heather M. Wilson
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - David C. Rees
- grid.46699.340000 0004 0391 9020Department of Haematology, King’s College Hospital, London, UK
| | - Stuart M. Haslam
- grid.7445.20000 0001 2113 8111Department of Life Sciences, Imperial College London, London, UK
| | - J. Alexandra Rowe
- grid.4305.20000 0004 1936 7988Centre for Immunity, Infection and Evolution, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Robert N. Barker
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Mark A. Vickers
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK ,grid.476695.f0000 0004 0495 4557Scottish National Blood Transfusion Service, Aberdeen, UK ,grid.417581.e0000 0000 8678 4766Department of Haematology, Aberdeen Royal Infirmary, Aberdeen, UK
| |
Collapse
|
36
|
Engwa GA, Okolie A, Chidili JPC, Okore PA, Onu PC, Ugwu MO, Oko DE, Ferdinand PU. Relationship of oxidative stress and antioxidant response with vaso-occlusive crisis in sickle cell anaemia. Afr Health Sci 2021; 21:150-158. [PMID: 34394292 PMCID: PMC8356617 DOI: 10.4314/ahs.v21i1.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Though sickle cell anaemia (SCA) is known to promote oxidative stress, there is paucity of information on the relationship between oxidative stress and vaso-occlusive crisis (VOC). OBJECTIVE This study was undertaken to evaluate the relationship of oxidative stress and antioxidant response with VOC in SCA. METHODS A cross-sectional case-control study was carried out at University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla, Enugu Nigeria involving 116 individuals which included 36 SCA subject, 40 sickle cell carriers (AS) and 40 healthy individuals (AA). Baseline information as well as the frequency of VOC was obtained from the participants and anaemia as well as oxidative stress and antioxidant indices were assessed in blood. RESULTS Anaemia was prevalent (88.9 %) in SCA individuals compared to AS (52.5%) and AA (47.5 %) individuals. Nitric oxide scavenging (NOS) and superoxide dismutase (SOD) activities as well as glutathione level were significantly (p<0.005) lower while catalase activity was higher in SCA individuals compared to controls (AA and AS). Higher malondialdehyde (MDA) level was associated with very severe VOC while low level of NOS activity was associated with severe VOC in SCA individuals. CONCLUSION Sickle cell anaemia exhibited oxidative stress and alteration in the levels of antioxidant indices which was possibly associated with vaso-occlusive crisis.
Collapse
Affiliation(s)
- Godwill Azeh Engwa
- Biochemistry, Department of Chemical Sciences, Godfrey Okoye University, P.M.B 01014, Thinkers Corner, Enugu Nigeria
- Department of Biological and Environmental Sciences, Faculty of Sciences, Walter Sisulu University, Mthatha 5117, South Africa
| | - Amanda Okolie
- Biochemistry, Department of Chemical Sciences, Godfrey Okoye University, P.M.B 01014, Thinkers Corner, Enugu Nigeria
| | - John Paul Chinedu Chidili
- Biotechnology, Department of Biological Sciences, Godfrey Okoye University, P.M.B 01014, Thinkers Corner, Enugu Nigeria
| | - Precious Amara Okore
- Biochemistry, Department of Chemical Sciences, Godfrey Okoye University, P.M.B 01014, Thinkers Corner, Enugu Nigeria
| | - Paul Chigozie Onu
- Biochemistry, Department of Chemical Sciences, Godfrey Okoye University, P.M.B 01014, Thinkers Corner, Enugu Nigeria
| | - Maryrose Onyinye Ugwu
- Biotechnology, Department of Biological Sciences, Godfrey Okoye University, P.M.B 01014, Thinkers Corner, Enugu Nigeria
| | - Daniella Ebeshe Oko
- Biotechnology, Department of Biological Sciences, Godfrey Okoye University, P.M.B 01014, Thinkers Corner, Enugu Nigeria
| | - Paschaline U Ferdinand
- Bio-resources Development Center Arochukwu, National Biotechnology Development Agency (NABDA), Abuja Nigeria
| |
Collapse
|
37
|
Plasma microparticles of sickle patients during crisis or taking hydroxyurea modify endothelium inflammatory properties. Blood 2021; 136:247-256. [PMID: 32285120 DOI: 10.1182/blood.2020004853] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/05/2020] [Indexed: 12/29/2022] Open
Abstract
Microparticles (MPs) are submicron extracellular vesicles exposing phosphatidylserine (PS), detected at high concentration in the circulation of sickle cell anemia (SS) patients. Several groups studied the biological effects of MPs generated ex vivo. Here, we analyzed for the first time the impact of circulating MPs on endothelial cells (ECs) from 60 sickle cell disease (SCD) patients. MPs were collected from SCD patients and compared with MPs isolated from healthy individuals (AA). Other plasma MPs were purified from SS patients before and 2 years after the onset of hydroxyurea (HU) treatment or during a vaso-occlusive crisis and at steady-state. Compared with AA MPs, SS MPs increased EC ICAM-1 messenger RNA and protein levels, as well as neutrophil adhesion. We showed that ICAM-1 overexpression was primarily caused by MPs derived from erythrocytes, rather than from platelets, and that it was abolished by MP PS capping using annexin V. MPs from SS patients treated with HU were less efficient to induce a proinflammatory phenotype in ECs compared with MPs collected before therapy. In contrast, MPs released during crisis increased ICAM-1 and neutrophil adhesion levels, in a PS-dependent manner, compared with MPs collected at steady-state. Furthermore, neutrophil adhesion was abolished by a blocking anti-ICAM-1 antibody. Our study provides evidence that MPs play a key role in SCD pathophysiology by triggering a proinflammatory phenotype of ECs. We also uncover a new mode of action for HU and identify potential therapeutics: annexin V and anti-ICAM-1 antibodies.
Collapse
|
38
|
Vona R, Sposi NM, Mattia L, Gambardella L, Straface E, Pietraforte D. Sickle Cell Disease: Role of Oxidative Stress and Antioxidant Therapy. Antioxidants (Basel) 2021; 10:antiox10020296. [PMID: 33669171 PMCID: PMC7919654 DOI: 10.3390/antiox10020296] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
Sickle cell disease (SCD) is the most common hereditary disorder of hemoglobin (Hb), which affects approximately a million people worldwide. It is characterized by a single nucleotide substitution in the β-globin gene, leading to the production of abnormal sickle hemoglobin (HbS) with multi-system consequences. HbS polymerization is the primary event in SCD. Repeated polymerization and depolymerization of Hb causes oxidative stress that plays a key role in the pathophysiology of hemolysis, vessel occlusion and the following organ damage in sickle cell patients. For this reason, reactive oxidizing species and the (end)-products of their oxidative reactions have been proposed as markers of both tissue pro-oxidant status and disease severity. Although more studies are needed to clarify their role, antioxidant agents have been shown to be effective in reducing pathological consequences of the disease by preventing oxidative damage in SCD, i.e., by decreasing the oxidant formation or repairing the induced damage. An improved understanding of oxidative stress will lead to targeted antioxidant therapies that should prevent or delay the development of organ complications in this patient population.
Collapse
Affiliation(s)
- Rosa Vona
- Biomarkers Unit, Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.V.); (N.M.S.); (L.G.)
| | - Nadia Maria Sposi
- Biomarkers Unit, Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.V.); (N.M.S.); (L.G.)
| | - Lorenza Mattia
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00161 Rome, Italy;
- Endocrine-Metabolic Unit, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Lucrezia Gambardella
- Biomarkers Unit, Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.V.); (N.M.S.); (L.G.)
| | - Elisabetta Straface
- Biomarkers Unit, Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.V.); (N.M.S.); (L.G.)
- Correspondence: ; Tel.: +39-064-990-2443; Fax: +39-064-990-3690
| | - Donatella Pietraforte
- Core Facilities, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| |
Collapse
|
39
|
Messonnier LA, Bartolucci P, d'Humières T, Dalmais E, Lacour JR, Freund H, Galactéros F, Féasson L. Preventive measures for the critical postexercise period in sickle cell trait and disease. J Appl Physiol (1985) 2021; 130:485-490. [PMID: 33270510 DOI: 10.1152/japplphysiol.00855.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The immediate postexercise/physical activity period is critical for sickle cell trait (SCT) carriers and disease (SCD) patients. Exercise-related blood acidosis is known to trigger the cascade of HbS deoxygenation and polymerization, leading to red blood cell sickling and subsequent complications. Unfortunately, two facts worsen exercise-related blood acidosis during the initial postexercise period: First, blood lactate and H+ concentrations continue to increase for several minutes after exercise completion, exacerbating blood acidosis. Second, blood lactate concentration remains elevated and pH altered for 20-45 min during inactivity after intense exercise, keeping acid/base balance disturbed for a long period after exercise. Therefore, the risk of complications (including vasoocclusive crises and even sudden death) persists and even worsens several minutes after intense exercise completion in SCT carriers or SCD patients. Light physical activity following intense exercise (namely, active recovery) may, by accelerating lactate removal and acid/base balance restoration, reduce the risk of complications. Scientific evidence suggests that light exercise at or below the first lactate threshold is an appropriate strategy.
Collapse
Affiliation(s)
- Laurent A Messonnier
- Laboratoire Interuniversitaire de Biologie de la Motricité, EA 7424, Université Savoie Mont Blanc, Chambéry, France
| | - Pablo Bartolucci
- INSERM U955, Equipe 2 Transfusion et maladies du globule rouge, Laboratoire d'Excellence GRex, Créteil, France.,Institut Mondor de Recherche Biomédicale (IMRB), Université Paris Est-Créteil (UPEC) Faculté de Médecine, Créteil, France.,Reference Center for Sickle Cell Disease, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France
| | - Thomas d'Humières
- Service de Physiologie Cardiovasculaire, Hôpitaux Universitaires Henri Mondor, Assistance Publique-Hôpitaux de Paris, Créteil, France
| | - Etienne Dalmais
- Laboratoire Interuniversitaire de Biologie de la Motricité, EA 7424, Université Savoie Mont Blanc, Chambéry, France.,Centre d'Evaluation et de Prévention Articulaire, Challes-les-Eaux, France
| | - Jean-René Lacour
- Laboratoire de Physiologie de l'Exercice, Faculté de Médecine Lyon-Sud, Université de Lyon, Université Claude Bernard Lyon 1, Oullins, France
| | - Hubert Freund
- Laboratoire Interuniversitaire de Biologie de la Motricité, EA 7424, Université Savoie Mont Blanc, Chambéry, France
| | - Frédéric Galactéros
- Institut Mondor de Recherche Biomédicale (IMRB), Université Paris Est-Créteil (UPEC) Faculté de Médecine, Créteil, France.,Reference Center for Sickle Cell Disease, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France
| | - Léonard Féasson
- University Hospital of Saint-Etienne, Myology Unit, Department of Clinical and Exercise Physiology, Referent Center of Neuromuscular Diseases, Euro-NmD, Saint-Etienne, France.,Laboratoire Interuniversitaire de Biologie de la Motricité, EA 7424, Université de Lyon, Université Jean Monnet, Saint-Etienne, France
| |
Collapse
|
40
|
Fujii J, Homma T, Kobayashi S, Warang P, Madkaikar M, Mukherjee MB. Erythrocytes as a preferential target of oxidative stress in blood. Free Radic Res 2021; 55:562-580. [PMID: 33427524 DOI: 10.1080/10715762.2021.1873318] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Red blood cells (RBC) are specifically differentiated to transport oxygen and carbon dioxide in the blood and they lack most organelles, including mitochondria. The autoxidation of hemoglobin constitutes a major source of reactive oxygen species (ROS). Nitric oxide, which is produced by endothelial nitric oxide synthase (NOS3) or via the hemoglobin-mediated conversion of nitrite, interacts with ROS and results in the production of reactive nitrogen oxide species. Herein we present an overview of anemic diseases that are closely related to oxidative damage. Because the compensation of proteins by means of gene expression does not proceed in enucleated cells, antioxidative and redox systems play more important roles in maintaining the homeostasis of RBC against oxidative insult compared to ordinary cells. Defects in hemoglobin and enzymes that are involved in energy production and redox reactions largely trigger oxidative damage to RBC. The results of studies using genetically modified mice suggest that antioxidative enzymes, notably superoxide dismutase 1 and peroxiredoxin 2, play essential roles in coping with oxidative damage in erythroid cells, and their absence limits erythropoiesis, the life-span of RBC and consequently results in the development of anemia. The degeneration of the machinery involved in the proteolytic removal of damaged proteins appears to be associated with hemolytic events. The ubiquitin-proteasome system is the dominant machinery, not only for the proteolytic removal of damaged proteins in erythroid cells but also for the development of erythropoiesis. Hence, despite the fact that it is less abundant in RBC compared to ordinary cells, the aberrant ubiquitin-proteasome system may be associated with the development of anemic diseases via the accumulation of damaged proteins, as typified in sickle cell disease, and impaired erythropoiesis.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Japan
| | - Takujiro Homma
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Japan
| | - Sho Kobayashi
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Japan
| | - Prashant Warang
- ICMR - National Institute of Immunohaematology, Mumbai, India
| | | | | |
Collapse
|
41
|
Safwat NA, ELkhamisy MM, Abdel-Wahab SEA, Hamza MT, Boshnak NH, Kenny MA. Polymorphisms of the receptor for advanced glycation end products as vasculopathy predictor in sickle cell disease. Pediatr Res 2021; 89:185-190. [PMID: 32544923 DOI: 10.1038/s41390-020-1014-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND The genetic variants of the receptor for advanced glycation end products (RAGE) gene have been associated with vascular disease risk. The objective of this work was to explore the association of three single-nucleotide polymorphisms (SNPs) of RAGE gene (374T/A, 429T/C, and G82S) with vascular complications in SCD. METHODS The study was conducted on 40 children with SCD and 40 healthy children served as controls. All participants were genotyped for the three studied RAGE polymorphisms by polymerase chain reaction (PCR). RESULTS Regarding 374T/A polymorphism, the frequency of TA, TT genotypes and T allele were higher in patients (p < 0.001). T allele was associated with higher incidence of sickling crisis and stroke (p < 0.05). In the subgroup analyses of 429T/C polymorphism, an association between C allele and SCD vascular complications was observed (p < 0.05). Concerning the frequency of G82S genotypes of RAGE, GG variant was detected in 39 (97.5%) of the patients, as compared with 40 (100%) of controls (p = 0.3). A regression analysis proved that HbS%, serum ferritin, and the -374T and 429C alleles were significant independent predictors of frequent sickling episodes (p < 0.05). CONCLUSIONS The C allele of -429T/C and T allele of 374T/A RAGE polymorphisms may be considered as predictors for vascular dysfunction in SCD. IMPACT The C allele of -429T/C and T allele of 374T/A RAGE polymorphisms may be considered as predictors for vascular dysfunction in SCD patients. To our knowledge, our study is the first exploring the association of three single-nucleotide polymorphisms of RAGE gene (374T/A, 429T/C, and G82S) with vascular complications in SCD. Early identification of patients carrying these genetic variants might be of great importance not only to identify subjects at risk of vasculopathy but also to direct them to RAGE-targeted treatments.
Collapse
Affiliation(s)
- Nesma Ahmed Safwat
- Department of Clinical Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Mai Mohamed ELkhamisy
- Department of Clinical Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Mohamed Tarif Hamza
- Department of Clinical Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Noha Hussein Boshnak
- Department of Clinical Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mahmoud Adel Kenny
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
42
|
Verma HK, Swarnakar S, L.V.K.S. B. Genetic association of GSTM1, GSTT1, and GSTP1 polymorphisms with sickle cell disease complications: A systematic review and meta-analysis. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
43
|
Ali MA, Ahmad A, Chaudry H, Aiman W, Aamir S, Anwar MY, Khan A. Efficacy and safety of recently approved drugs for sickle cell disease: a review of clinical trials. Exp Hematol 2020; 92:11-18.e1. [PMID: 32841705 PMCID: PMC7442900 DOI: 10.1016/j.exphem.2020.08.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/04/2020] [Accepted: 08/19/2020] [Indexed: 02/08/2023]
Abstract
Sickle cell disease is prevalent in several parts of the world. Most hospitalizations of these patients are related to pain crisis episodes. Moreover, levels of hemoglobin are lower in sickle cell disease patients as compared with the general population. Complications related to sickle cell disease are managed with blood transfusions, hydroxyurea, and opioids. Despite these therapies, patients with sickle cell disease experience multiple pain crisis episodes leading to hospitalizations and end-organ damage. The US Food and Drug Administration has approved three new drugs-L-glutamine, voxelotor, and crizanlizumab-for the prophylaxis and treatment of complications related to sickle cell disease. This review was aimed at assessing the efficacy and safety of recently approved drugs for the treatment of sickle cell disease. A comprehensive search was made on PubMed and clinicaltrials.gov to look for clinical trials reporting the efficacy and safety of recently approved drugs for sickle cell disease. Based on the results of clinical trials, L-glutamine, voxelotor, and crizanlizumab were well tolerated by sickle cell disease patients. L-Glutamine and crizanlizumab reduced the number of sickle cell crisis episodes, while voxelotor improved the level of hemoglobin in sickle cell disease patients. These drugs were effective alone and in combination with hydroxyurea.
Collapse
Affiliation(s)
| | - Asrar Ahmad
- Internal Medicine, Abington Hospital-Jefferson Health, Abington, PA.
| | - Hafsa Chaudry
- Internal Medicine, King Edward Medical University, Lahore, Pakistan
| | - Wajeeha Aiman
- Internal Medicine, Nishtar Medical University, Multan, Pakistan
| | - Sobia Aamir
- Internal Medicine, The Children's Hospital and The Institute of Child Health, Lahore, Pakistan
| | | | - Anam Khan
- Internal Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
44
|
Hariharan P, Chavan V, Nadkarni A. Significance of heme oxygenase-1(HMOX1) gene on fetal hemoglobin induction in sickle cell anemia patients. Sci Rep 2020; 10:18506. [PMID: 33116199 PMCID: PMC7595119 DOI: 10.1038/s41598-020-75555-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/22/2020] [Indexed: 11/09/2022] Open
Abstract
Though the patients with sickle cell anemia (SCA) inherit same genetic mutation, they show considerable phenotypic heterogeneity. It has been observed that patients with elevated fetal hemoglobin (HbF) levels have a relatively mild clinical course. There is sparse literature on the association of higher HbF levels leading to reduction in the oxidative stress in SCA patients. Hence in this study, the significance between the HMOX1 gene polymorphisms and the HbF levels has been studied. Preliminary screening was carried out. Genotyping of 3 variants in the HMOX1 gene was performed in 90 SCA patients and 50 healthy controls by PCR–RFLP, GeneScan and direct DNA sequencing. It was observed that SCA patients with higher HbF levels, showed improved hematological indices with an inverse effect on HbS levels. The TT genotype of rs2071746 (A→T) polymorphism was found to be associated with elevated HbF levels (P: 0.012). Also, the long form (> 25 GT repeats) of rs3074372 (GT)n repeats was found to be linked with increased HbF levels. We could not find any association of rs2071749 (A→G) polymorphism with the HbF levels. As, the sickle cell anemia patients show significant oxidative stress due to hemolysis, the study of polymorphisms in the HMOX1 gene may act as a potential independent marker for elevated HbF levels.
Collapse
Affiliation(s)
- Priya Hariharan
- Department of Haematogenetics, ICMR-National Institute of Immunohaematology, 13th floor NMS Building, KEM Hospital Campus, Parel, Mumbai, 400012, Maharashtra, India
| | - Vrushali Chavan
- Department of Haematogenetics, ICMR-National Institute of Immunohaematology, 13th floor NMS Building, KEM Hospital Campus, Parel, Mumbai, 400012, Maharashtra, India
| | - Anita Nadkarni
- Department of Haematogenetics, ICMR-National Institute of Immunohaematology, 13th floor NMS Building, KEM Hospital Campus, Parel, Mumbai, 400012, Maharashtra, India.
| |
Collapse
|
45
|
Zeitoune R, Mogami R, Koifman ACB, Lopes AJ, Soares AR, Martins RAG, Maioli MCP. Diaphragm ultrasonography in adults with sickle cell anemia: evaluation of morphological and functional aspects. ACTA ACUST UNITED AC 2020; 25:372-382. [PMID: 33095119 DOI: 10.1080/16078454.2020.1833506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVES To assess morphological and functional aspects of the diaphragm by ultrasonography (US) in adults with sickle cell anemia (SCA) and evaluate if the diaphragmatic musculature can play a role in changes found in pulmonary function tests (PFTs) of these patients. METHODS This is a cross-sectional observational and single-center study involving 40 adults with SCA who underwent diaphragm US and PFTs with a maximum of 1 month between the two tests. Diaphragm US was performed in B and M modes, and echogenicity, thickness and movement of the muscle was assessed in different respiratory maneuvers. RESULTS Diaphragms had preserved echogenicity and the thickness was not significantly different between the groups. The SCA group exhibited significantly higher movement of the right hemidiaphragm during deep breathing (p = 0.004) and the sniff test (p = 0.0008) and lower movement of the left hemidiaphragm during quiet breathing (p = 0.009). There was a predominance of restrictive pattern (65%) and a global reduction in respiratory muscle strength (RMS) (70%). CONCLUSIONS This study shows that adults with SCA had normal morphostructural aspects and absence of diaphragm dysfunction. Otherwise, they presented greater movement of the right hemidiaphragm during deep breathing and sniff test maneuvers. Despite the restrictive pattern and the reduction in RMS found in PFTs, the diaphragm of young adults with SCA did not have weakness or paralysis.
Collapse
Affiliation(s)
- Rachel Zeitoune
- Marinha do Brasil, São Paulo, Brazil.,Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | - Roberto Mogami
- Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | - Ana Celia Baptista Koifman
- Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil.,Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Agnaldo Jose Lopes
- Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil.,Centro Universitário Augusto Motta (UNISUAM), Rio de Janeiro, Brazil
| | | | | | | |
Collapse
|
46
|
Pedrosa AM, Leal LKAM, Lemes RPG. Effects of hydroxyurea on cytotoxicity, inflammation and oxidative stress markers in neutrophils of patients with sickle cell anemia: dose-effect relationship. Hematol Transfus Cell Ther 2020; 43:468-475. [PMID: 33051133 PMCID: PMC8573033 DOI: 10.1016/j.htct.2020.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 06/18/2020] [Accepted: 07/28/2020] [Indexed: 02/02/2023] Open
Abstract
Introduction Although the efficacy of hydroxyurea (HU) in inhibiting erythrocyte sickling has been well demonstrated, the action of this drug on human neutrophils and the mechanism by which it improves the manifestations of the disease have not been studied thoroughly. We aimed to investigate the cell viability, along with inflammatory and oxidative markers in the neutrophils of sickle cell anemia (SCA) patients and the effects of HU therapy on these cells, by evaluating the dose-responsiveness. Methods In the present study, 101 patients (45 men and 56 women, aged 18–69 years) with SCA were divided into groups according to the use or not of HU: the SS group (without HU treatment, n = 47) and the SSHU group (under HU treatment, n = 54). The SSHU group was further stratified into subgroups according to the daily dose of the drug that patients already used: SSHU - 0.5 g (n = 19); SSHU - 1 g (n = 26) and SSHU - 1.5–2 g (n = 9). A control group (AA) comprised 50 healthy individuals. Neutrophils isolated from whole blood were analyzed using Trypan Blue, monoiodotyrosine (MTT) and lactate dehydrogenase (LDH) toxicity assays. Myeloperoxidase (MPO), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) activities and concentrations of interleukin 10 (IL-10), tumor necrosis factor alpha (TNF-α) and malonaldehyde (MDA) were also measured. Results Neutrophils from SCA patients showed membrane fragility and a significant decrease in cell viability when analyzed by Trypan Blue (p < 0.05), MTT (p < 0.001) and LDH (p = 0.011), compared to the AA group. Levels of inflammatory (MPO, TNF-α, and IL-10) and oxidative markers (SOD, GSH-Px, and MDA) were also altered (p < 0.05) in these cells, showing a significant difference in the SSHU-1g and SSHU - 1.5–2 g groups, compared to the SS group. Treatment with HU reverted the levels of all markers to concentrations similar to those in healthy individuals in a positive dose-effect relationship. Conclusion The HU did not generate a cytotoxic effect on neutrophils in SCA patients, but it modulated their oxidative and inflammatory mechanisms, promoting cytoprotection with a positive dose-effect.
Collapse
|
47
|
Rashidpour S, Zahedipour F, Karimi G, Jamialahmadi K. Protective Effects of Osthole against Free Radical-Induced Hemolysis of Erythrocytes. PHARMACEUTICAL SCIENCES 2020. [DOI: 10.34172/ps.2020.65] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background: Osthole, one of the most active components of Cnidium monnieri, has different pharmacological and biological effects such as boosting the immune system, reducing rheumatoid pain, hepatoprotective, and inhibitory effect on osteoporosis. Furthermore, it showed anti inflammatory, anti-cancer, and antioxidant properties. However, there is little information about the antioxidant effects of osthole using cell-based assays. In the current work, we used in vitro model of 2,2-azobis (2-amidinopropane) dihydrochloride (AAPH)-induced hemolysis of erythrocytes to investigate the protective effects of osthole against oxidative damage of biological membranes. Methods: Erythrocytes were challenged with 2, 2ꞌ-azobis (2-aminopropane) dihydrochloride (AAPH) as a model oxidant in the presence and absence of osthole. The protective effects of osthole on lipid peroxidation, protein carbonyl oxidation, glutathione (GSH) content of erythrocytes were evaluated and compared with control samples. Results: It was found that osthole has protective effects on erythrocyte hemolysis induced by AAPH at different concentrations in a time-dependent manner. Osthole also suppressed lipid and protein oxidation as well as reductions in GSH content in a concentration and timedependent manner. Conclusion: Osthole showed protective effects against free radical-induced hemolysis in rat erythrocytes. Therefore, it can be considered as a supplement for the prevention or treatment of a variety of human health problems associated with oxidative stress. However, further investigations are required to illustrate other possible impacts of osthole on cells.
Collapse
Affiliation(s)
- Soroush Rashidpour
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Zahedipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
48
|
Potential causal role of l-glutamine in sickle cell disease painful crises: A Mendelian randomization analysis. Blood Cells Mol Dis 2020; 86:102504. [PMID: 32949984 DOI: 10.1016/j.bcmd.2020.102504] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/09/2020] [Accepted: 09/09/2020] [Indexed: 01/12/2023]
Abstract
In a recent clinical trial, the metabolite l-glutamine was shown to reduce painful crises in sickle cell disease (SCD) patients. To support this observation and identify other metabolites implicated in SCD clinical heterogeneity, we profiled 129 metabolites in the plasma of 705 SCD patients. We tested correlations between metabolite levels and six SCD-related complications (painful crises, cholecystectomy, retinopathy, leg ulcer, priapism, aseptic necrosis) or estimated glomerular filtration rate (eGFR), and used Mendelian randomization (MR) to assess causality. We found a potential causal relationship between l-glutamine levels and painful crises (N = 1278, odds ratio (OR) [95% confidence interval] = 0.68 [0.52-0.89], P = 0.0048). In two smaller SCD cohorts (N = 299 and 406), the protective effect of l-glutamine was observed (OR = 0.82 [0.50-1.34]), although the MR result was not significant (P = 0.44). We identified 66 significant correlations between the levels of other metabolites and SCD-related complications or eGFR. We tested these correlations for causality using MR analyses and found no significant causal relationship. The baseline levels of quinolinic acid were associated with prospectively ascertained survival in SCD patients, and this effect was dependent on eGFR. Metabolomics provide a promising approach to prioritize small molecules that may serve as biomarkers or drug targets in SCD.
Collapse
|
49
|
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
|
50
|
Zhu X, Xi C, Ward A, Takezaki M, Shi H, Peterson KR, Pace BS. NRF2 mediates γ-globin gene regulation through epigenetic modifications in a β-YAC transgenic mouse model. Exp Biol Med (Maywood) 2020; 245:1308-1318. [PMID: 32715783 DOI: 10.1177/1535370220945305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
IMPACT STATEMENT Sickle cell disease is an inherited hemoglobin disorder that affects over 100,000 people in the United States causing high morbidity and early mortality. Although new treatments were recently approved by the FDA, only one drug Hydroxyurea induces fetal hemoglobin expression to inhibit sickle hemoglobin polymerization in red blood cells. Our laboratory previously demonstrated the ability of the NRF2 activator, dimethyl fumarate to induce fetal hemoglobin in the sickle cell mouse model. In this study, we investigated molecular mechanisms of γ-globin gene activation by NRF2. We observed the ability of NRF2 to modulate chromatin structure in the human β-like globin gene locus of β-YAC transgenic mice during development. Furthermore, an NRF2/TET3 interaction regulates γ-globin gene DNA methylation. These findings provide potential new molecular targets for small molecule drug developed for treating sickle cell disease.
Collapse
Affiliation(s)
- Xingguo Zhu
- Division of Hematology/Oncology, Department of Pediatrics, Augusta University, Augusta, GA 30912, USA.,Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Caixia Xi
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Alexander Ward
- Division of Hematology/Oncology, Department of Pediatrics, Augusta University, Augusta, GA 30912, USA
| | - Mayuko Takezaki
- Division of Hematology/Oncology, Department of Pediatrics, Augusta University, Augusta, GA 30912, USA
| | - Huidong Shi
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Kenneth R Peterson
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Betty S Pace
- Division of Hematology/Oncology, Department of Pediatrics, Augusta University, Augusta, GA 30912, USA.,Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA.,Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|