1
|
Zadeh FJ, Fateh A, Saffari H, Khodadadi M, Eslami Samarin M, Nikoubakht N, Dadgar F, Goodarzi V. The vaso-occlusive pain crisis in sickle cell patients: A focus on pathogenesis. Curr Res Transl Med 2025; 73:103512. [PMID: 40220659 DOI: 10.1016/j.retram.2025.103512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025]
Abstract
Vaso-occlusive pain crisis (VOC) is recognized as a prominent complication of sickle cell disease, accompanied by debilitating pain and serious consequences for patients, making it the primary cause of visits to hospital emergency departments. In the etiology of VOC, the intricate interaction of endothelial cells, hypoxia, inflammation, and the coagulation system is pivotal. Hemoglobin S polymerization under hypoxic conditions leads to the formation of rigid and adhesive red blood cells that interact with vascular endothelial cells and other blood cells, causing occlusion and subsequent inflammation. Hemolysis of red blood cells results in anemia and heightened inflammation, whereas oxidative stress and involvement of the coagulation system further complicate matters. In this review, we strive to examine the pathophysiology of VOC from these mentioned aspects by consolidating findings from various studies, as a comprehensive understanding of the causes of VOC is essential for the development of targeted therapeutic interventions and the prevention and management of pain, ultimately improving the quality of life for patients.
Collapse
Affiliation(s)
| | - Azadeh Fateh
- Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hamed Saffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammadamin Eslami Samarin
- Student Research Committee, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Universal Scientific Education and Research Network(USERN),Tehran,Iran
| | - Nasim Nikoubakht
- Department of Anesthesiology, Hazrat-e Rasool General Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Dadgar
- Department of Internal Medicine, Lorestan University of Medical Science, Khorramabad, Iran; Student Research Committe, Lorestan University of Medical Science, Khorramabad, Iran
| | - Vahid Goodarzi
- Department of Anesthesiology, Rasoul-Akram Medical Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
2
|
Bou-Fakhredin R, Cappellini MD, Taher AT, De Franceschi L. Hypercoagulability in hemoglobinopathies: Decoding the thrombotic threat. Am J Hematol 2025; 100:103-115. [PMID: 39400943 DOI: 10.1002/ajh.27500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 10/15/2024]
Abstract
Beta (β)-thalassemia and sickle cell disease (SCD) are characterized by a hypercoagulable state, which can significantly influence organ complication and disease severity. While red blood cells (RBCs) and erythroblasts continue to play a central role in the pathogenesis of thrombosis in β-thalassemia and SCD, additional factors such as free heme, inflammatory vasculopathy, splenectomy, among other factors further contribute to the complexity of thrombotic risk. Thus, understanding the role of the numerous factors driving this hypercoagulable state will enable healthcare practitioners to enhance preventive and treatment strategies and develop novel therapies for the future. We herein describe the pathogenesis of thrombosis in patients with β-thalassemia and SCD. We also identify common mechanisms underlying the procoagulant profile of hemoglobinopathies translating into thrombotic events. Finally, we review the currently available prevention and clinical management of thrombosis in these patient populations.
Collapse
Affiliation(s)
- Rayan Bou-Fakhredin
- Department of Clinical Sciences and Community, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, SC Medicina ad Indirizzo Metabolico, Milan, Italy
| | - Maria Domenica Cappellini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, SC Medicina ad Indirizzo Metabolico, Milan, Italy
| | - Ali T Taher
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Lucia De Franceschi
- Department of Medicine, University of Verona, and Azienda Ospedaliera Universitaria Verona, Verona, Italy
| |
Collapse
|
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
|
Anorue EC, Joshua PE. Evaluation of anti-sickling effects of two varieties of Cajanus cajan (L.) Huth on sickle cell beta thalassemia. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118280. [PMID: 38714239 DOI: 10.1016/j.jep.2024.118280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/14/2024] [Accepted: 04/29/2024] [Indexed: 05/09/2024]
Abstract
ETHNO-PHARMACOLOGICAL RELEVANCE Globally, the prevalence of sickle cell disease is on the rise, with developing countries experiencing particularly alarming mortality rate compared to developed nations. The World Health Organization (WHO) and United Nations (UN) have acknowledged sickle cell disease as a significant global public health concern. Unfortunately, a cure for this condition is yet to be discovered, and existing allopathic treatments, while offering relief, come with serious side effects. In recent times, there has been a growing interest in exploring the potential of medicinal plants for treating sickle cell disease due to their content of secondary metabolites that may impact the disease's mechanisms. Cajanus cajan, a crucial grain legume in rain-fed agriculture in semi-arid tropics, has been traditionally used in folk medicine to manage various illnesses and is suggested to possess anti-sickling properties. AIM OF THE STUDY The present study investigated two varieties of C. cajan for their effectiveness in treating sickle cell beta thalassemia, a variant of sickle cell disease. MATERIALS AND METHODS The study was divided into four groups consisting of the untreated group (group 1), group treated with standard drug (group 2), group treated with white C. cajan (group 3) and group treated with brown C. cajan (group 4). The effects of the two variety of C. cajan were measured by polymerization test, reversibility test, osmotic fragility test, deoxygenation and beta globin synthesis test. RESULT The results revealed that both varieties of C. cajan demonstrated a reduction in polymerization rates, reversed sickled red blood cells, increased the oxygen affinity of Hb-S/β, elevated the Fe2+/Fe3+ ratio, and maintained the membrane stability of red blood cells. Notably, the white variety exhibited superior anti-sickling properties compared to the brown variety. CONCLUSION This suggests that this significant leguminous crop could be utilized for the treatment and management of sickling disorders, particularly in low-income countries where conventional treatments may be financially inaccessible to patients.
Collapse
Affiliation(s)
- Eleazar Chukwuemeka Anorue
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, 410001, Nsukka, Enugu State, Nigeria; Department of Chemistry, School of Sciences, Shalom Science and Technology Academy, Enugu State, Nigeria.
| | - Parker Elijah Joshua
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, 410001, Nsukka, Enugu State, Nigeria
| |
Collapse
|
5
|
Maman D, Fournier L, Steinfeld Y, Berkovich Y. Etiology, Outcomes, and Complications of Total Hip Arthroplasty in Younger Patients: A Nationwide Big Data Analysis. J Clin Med 2024; 13:4535. [PMID: 39124801 PMCID: PMC11313375 DOI: 10.3390/jcm13154535] [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: 06/28/2024] [Revised: 07/11/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Background: This study investigates the rising trend of total hip arthroplasty (THA) in patients under 55 years old, commonly referred to as "younger" THA patients. Traditionally a procedure for older adults with osteoarthritis, THA is increasingly performed on younger patients. Methods: Using data from the Nationwide Inpatient Sample (NIS) from 2016 to 2019, we analyze the factors driving this trend, including the causes of hip issues, patient characteristics, and coexisting medical conditions. The study examines in-hospital mortality, length of stay, post-surgical complications, and hospitalization costs for 231,630 THA patients aged 18-54.9, identified using ICD-10 codes. Results: Statistical analysis revealed that younger patients (aged 18-34.9) had higher rates of chronic anemia, inflammatory bowel disease, sickle cell disorders, connective tissue disorders, and coagulation defects compared to patients aged 35-44.9 and 45-54.9. They also experienced the longest hospital stays (2.08 days) and highest costs ($70,540). Significant odds ratios were found for sickle cell disorders (36.078), coagulation defects (1.566), inflammatory bowel disease (2.582), connective tissue disorders (11.727), hip dislocation (3.447), and blood transfusion (1.488) in younger patients compared to other THA patients. Conclusions: Comprehensive analysis of these unique needs is crucial for optimizing care, tailoring treatment, managing co-existing conditions, and personalizing recovery strategies to improve outcomes and quality of life for younger THA patients.
Collapse
Affiliation(s)
- David Maman
- Carmel Medical Center, Technion—Israel Institute of Technology, Haifa 2611001, Israel; (L.F.); (Y.B.)
| | - Linor Fournier
- Carmel Medical Center, Technion—Israel Institute of Technology, Haifa 2611001, Israel; (L.F.); (Y.B.)
| | | | - Yaron Berkovich
- Carmel Medical Center, Technion—Israel Institute of Technology, Haifa 2611001, Israel; (L.F.); (Y.B.)
| |
Collapse
|
6
|
Carpintieri S, Uyar E, Anand C, Buryk Y. Navigating the Interplay of Sickle Cell Vasculopathy and Moyamoya Cerebrovascular Changes: A Case Report. Cureus 2024; 16:e67302. [PMID: 39301408 PMCID: PMC11412159 DOI: 10.7759/cureus.67302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2024] [Indexed: 09/22/2024] Open
Abstract
Sickle cell disease (SCD) is a hereditary hemoglobinopathy that can lead to progressive vasculopathy, increasing the risk of cerebrovascular complications. Moyamoya syndrome (MMS), a rare disorder characterized by stenosis of the internal carotid arteries, can occur in SCD patients due to chronic endothelial damage and inflammation. The coexistence of these conditions can result in severe cerebrovascular complications, presenting unique diagnostic and therapeutic challenges. We present a 35-year-old African American male with a complex interplay of advanced SCD and MMS, manifesting as extensive cerebrovascular disease and recurrent ischemic strokes. A CT angiogram (CTA) of the head showed diffusely decreased caliber of the right M1 segment, appearing worse compared to prior studies. CTA of the head and neck demonstrated a new cut-off of the distal right M3 segment with an asymmetric paucity of arborizing vessels within the right middle cerebral artery (MCA) distribution, consistent with progressive sickle cell vasculopathy and also demonstrated abnormal dilated collateral vessels. Further imaging with MRI exhibited multiple prior ischemic strokes in various vascular territories despite previous revascularization surgery with a left superficial temporal artery to MCA bypass. The patient's progressive cerebrovascular disease was attributed to sickle cell vasculopathy exacerbated by MMS, resulting in compromised cerebral perfusion through distinct pathological mechanisms. Management involved a multidisciplinary treatment approach, including chronic transfusions, antiplatelet therapy, surgical revascularization with extracranial-intracranial bypass, seizure management, and neuropsychiatric support. Despite maximal therapy, the patient experienced recurrent cerebrovascular events and progressive neurological deficits, highlighting the challenges in controlling these intertwined disease processes. It signifies the importance of early recognition of this rare co-occurrence and implementation of prompt multidisciplinary treatment to improve outcomes.
Collapse
Affiliation(s)
| | - Elias Uyar
- Medical School, Ross University School of Medicine, Miramar, USA
| | - Christian Anand
- Medical School, St. George's University School of Medicine, True Blue, GRD
| | - Yaroslav Buryk
- Pulmonary and Critical Care, Jackson Memorial Hospital, Miami, USA
| |
Collapse
|
7
|
Zhu K, Lv F, Hou X, Wang F, Pang L, Zhong M. Thrombosis in vasculitis: An updated review of etiology, pathophysiology, and treatment. Heliyon 2024; 10:e30615. [PMID: 38975109 PMCID: PMC11225688 DOI: 10.1016/j.heliyon.2024.e30615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 07/09/2024] Open
Abstract
Introduction Thromboembolic disease is a complication of many vasculitides. A common observation is that thromboembolic events coincide with the period of vasculitic disease, but the mechanism by which this occurs remains unclear. Inflammatory thrombosis is now recognized as a symptom of arteritis rheumatic, and vasculitides such as Behçet's syndrome (BS), and anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitis (AAV) or giant cell arteritis (GCA). This systematic review aimed to explain recent findings related to etiology, pathophysiology, and treatment methods for BS, AAV, and medium/large-vessel vasculitis. Methods A comprehensive literature search on English sources from PubMed, Scopus, MEDLINE, Science Direct, ProQuest, AIM, CINAHIL, and ELDIS databases was used to find the relevant articles and reports. The relevant papers (having full text) were obtained until June 2023. Two independent reviewers screened the titles and abstracts of the obtained articles, and a third arbitrator resolved disputes between the reviewers. Results and conclusion It is becoming increasingly clear that certain systemic inflammatory diseases, like vasculitis, are linked to a higher risk of both venous and arterial thrombosis. An increased incidence of thromboembolic disease in AAV has been noted, particularly during times of active disease. Growing evidence supports the use of immunosuppression in the management of venous thrombosis in vasculitis. These patients also have a higher risk of developing ischemic disease.
Collapse
Affiliation(s)
- Kai Zhu
- Department of Vascular Surgery, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou City, 2530000, China
| | - Feng Lv
- Department of Vascular Surgery, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou City, 2530000, China
| | - Xiangqian Hou
- Department of Vascular Surgery, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou City, 2530000, China
| | - Feng Wang
- Department of Vascular Surgery, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou City, 2530000, China
| | - Linbin Pang
- Department of Vascular Surgery, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou City, 2530000, China
| | - Miqian Zhong
- Department of Hematopathology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 2530000, China
| |
Collapse
|
8
|
Hamdy M, Shaheen IA, Khallaf M, Selim YMM. Thrombin activatable fibrinolysis inhibitor plasma levels and TAFI Thr325Ile genetic polymorphism in a cohort of Egyptian sickle cell disease patients and impact on disease severity. Pediatr Blood Cancer 2024; 71:e30959. [PMID: 38520679 DOI: 10.1002/pbc.30959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/07/2024] [Accepted: 02/28/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Thrombin is a critical protease modulating thrombosis as well as inflammation, which are one of the main pathophysiological mechanisms in sickle vasculopathy, and its levels were reported to be high in sickle cell disease (SCD). The thrombin-thrombomodulin complex activates the TAFI inhibitor of fibrinolysis, which acts by reducing plasmin affinity for its substrate thus hindering fibrinolysis. OBJECTIVE We aimed to determine the influence of the Thr325Ile single nucleotide polymorphism (SNP) on TAFI antigen levels and potential effects on the severity of SCD in a cohort of Egyptian patients. METHODS Genotyping of Thr325lle polymorphism using Taq-Man SNP genotyping assay and TAFI level measurement using an enzyme-linked immunosorbent assay were performed for 80 SCD patients (45 homozygous HbSS, 16 S/β0 and 19 Sβ+) as well as 80 age- and gender-matched healthy control subjects. RESULTS Plasma TAFI levels were higher in SCD patients with Thr325Ile polymorphism, yet the difference was not statistically significant (p = .204). SCD patients with polymorphic genotypes had a greater number of hospital admissions (p = .03). Ten patients with acute chest syndrome had the homozygous polymorphic genotype (GG), and all patients with pulmonary hypertension had the polymorphic genotype (six were homozygous [GG] and five were heterozygous [GA]). Patients with SCD complicated with pulmonary hypertension showed significantly higher plasma TAFI levels (p = .044). CONCLUSION The analysis of Thr325Ile polymorphisms combined with plasma TAFI levels suggests that the analyzed SNP could influence plasma TAFL levels and SCD disease severity and hospitalization rates, which could be predictors for complex disease.
Collapse
Affiliation(s)
- Mona Hamdy
- Department of Pediatrics, Pediatric Hematology and BMT Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Iman A Shaheen
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed Khallaf
- Department of Pediatrics, Pediatric Hematology and BMT Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Yasmeen M M Selim
- Department of Pediatrics, Pediatric Hematology and BMT Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
9
|
Žigon P, Shao B. Editorial: Inflammation, the link between venous and arterial thrombosis. Front Cardiovasc Med 2024; 11:1433858. [PMID: 38873263 PMCID: PMC11171132 DOI: 10.3389/fcvm.2024.1433858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Affiliation(s)
- Polona Žigon
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- FAMNIT, University of Primorska, Koper, Slovenia
| | - Bojing Shao
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| |
Collapse
|
10
|
Chatzidavid S, Flevari P, Tombrou I, Anastasiadis G, Dimopoulou M. Pulmonary Hypertension in Sickle Cell Disease: Novel Findings of Gene Polymorphisms Related to Pathophysiology. Int J Mol Sci 2024; 25:4792. [PMID: 38732015 PMCID: PMC11084253 DOI: 10.3390/ijms25094792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Pulmonary hypertension (PH) is a progressive and potentially fatal complication of sickle cell disease (SCD), affecting 6-10% of adult SCD patients. Various mechanisms and theories have been evaluated to explain the pathophysiology of this disease. However, questions remain, particularly regarding the clinical heterogeneity of the disease in terms of symptoms, complications, and survival. Beyond the classical mechanisms that have been thoroughly investigated and include hemolysis, nitric oxide availability, endothelial disorders, thrombosis, and left heart failure, attention is currently focused on the potential role of genes involved in such processes. Potential candidate genes are investigated through next-generation sequencing, with the transforming growth factor-beta (TGF-β) pathway being the initial target. This field of research may also provide novel targets for pharmacologic agents in the future, as is already the case with idiopathic PH. The collection and processing of data and samples from multiple centers can yield reliable results that will allow a better understanding of SCD-related PH as a part of the disease's clinical spectrum. This review attempts to capture the most recent findings of studies on gene polymorphisms that have been associated with PH in SCD patients.
Collapse
Affiliation(s)
| | | | | | | | - Maria Dimopoulou
- Thalassemia and Sickle Cell Disease Unit, Center of Expertise in Rare Hematological Diseases (Hemoglobinopathies), Laikon General Hospital Member of EuroBlood NET, 16 Sevastoupoleos Str., 11526 Athens, Greece; (S.C.); (P.F.); (I.T.); (G.A.)
| |
Collapse
|
11
|
Khurana K, Mahajan S, Acharya S, Kumar S, Toshniwal S. Clinical Biomarkers of Acute Vaso-Occlusive Sickle Cell Crisis. Cureus 2024; 16:e56389. [PMID: 38633967 PMCID: PMC11022002 DOI: 10.7759/cureus.56389] [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: 01/16/2024] [Accepted: 03/17/2024] [Indexed: 04/19/2024] Open
Abstract
It is known that an inherited blood condition called sickle cell disease (SCD) is a result of one gene. A number of blood and urine biomarkers have been determined in association with lab and clinical history for SCD patients. SCD has numerous interacting pathways associated with it, which have been identified by biomarkers. These mechanisms consist of some examples, such as endothelial vasodilation response, hypercoagulability, hemolysis, inflammation, oxidative stress, vascular dysfunction, and reperfusion injury among others. To effectively manage SCD, a comprehensive panel of validated blood and urine biomarkers must be established. Despite its monogenic inheritance, the complex nature of the SCD phenotype has impeded progress in its treatment. However, significant strides have been made in clinical biotechnology, paving the way for potential breakthroughs. In SCD, a panel of verified blood and urine biomarkers must be established, however. Despite monogenic inheritance, the great complexity of the SCD phenotype has hindered progress in its management. With few exceptions, clinical biomarkers of illness severity have been found through epidemiological investigations; nevertheless, systematic integration of these biomarkers into clinical treatment algorithms has not occurred. Furthermore, sickle cell crisis, the primary acute consequence of SCD, has been difficult to diagnose with the biomarkers now in use. Inadequate care and a lack of appropriate outcome measures for clinical research are the consequences of these diagnostic constraints. A new chapter in SCD customized treatment has begun with recent advancements in molecular and imaging diagnostics. Strategies in precision medicine are especially relevant now that molecular therapies are within reach. The significance of biochemical indicators linked to clinical manifestation and sub-phenotype identification in SCD is reviewed in this research.
Collapse
Affiliation(s)
- Kashish Khurana
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Satish Mahajan
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sourya Acharya
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sunil Kumar
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Saket Toshniwal
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
12
|
Packialakshmi B, Limerick E, Ackerman HC, Lin X, Nekhai S, Oliver JD, Stewart IJ, Knepper MA, Fitzhugh C, Zhou X. Proteomic analyses of urinary exosomes identify novel potential biomarkers for early diagnosis of sickle cell nephropathy, a sex-based study. Front Physiol 2024; 15:1300667. [PMID: 38426210 PMCID: PMC10901968 DOI: 10.3389/fphys.2024.1300667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 01/26/2024] [Indexed: 03/02/2024] Open
Abstract
Sickle cell nephropathy (SCN) is a leading cause of morbidity and mortality in sickle cell disease (SCD). Early intervention is crucial for mitigating its effects. However, current diagnostic methods rely on generic tests and may not detect SCN until irreversible renal damage occurs. Therefore, specific biomarkers for early diagnosis of SCN are needed. Urinary exosomes, membrane-bound vesicles secreted by renal podocytes and epithelial cells, contain both common and cell type-specific membrane and cytosolic proteins, reflecting the physiologic and pathophysiologic states of the kidney. Using proteomics, we analyzed the proteomes of urinary exosomes from humanized SCD mice at 2 months (without albuminuria) and 4 months (with albuminuria) of age. Excretion of 164 proteins were significantly increased and 176 proteins was significantly decreased in the exosomes when mice developed albuminuria. Based on the relevance to SCD, chronic kidney disease and Western blot confirmation in mice, we analyzed protein abundance of heparanase, cathepsin C, α2-macroglobulin and sarcoplasmic endoplasmic Ca2+ ATPase-3 (SERCA3) in the urinary exosomes and urine of 18 SCD subjects without albuminuria and 12 subjects with albuminuria using Western blot analyses. Both male and female subjects increased or tended to increase the excretion of these proteins in their urinary exosomes upon developing albuminuria, but female subjects demonstrated stronger correlations between the excretion of these proteins and urine albumin creatinine ratio (UACR) compared to male subjects. In contrast, exosomal excretion of Tamm-Horsfall protein, β-actin and SHP-1 was independent of albuminuria. These findings provide a foundation for a time-course study to determine whether increases in the levels of these proteins precede the onset of albuminuria in patients, which will help determine the potential of these proteins as biomarkers for early detection of SCN.
Collapse
Affiliation(s)
- Balamurugan Packialakshmi
- Department of Medicine, Uniformed Services University of Health Sciences, Bethesda, MD, United States
| | - Emily Limerick
- Cellular and Molecular Therapeutic Branch, National Heart Lung and Blood Institute, Bethesda, MD, United States
| | - Hans C. Ackerman
- Physiology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, United States
| | - Xionghao Lin
- Department of Medicine, Howard University, Washington, DC, United States
| | - Sergei Nekhai
- Department of Medicine, Howard University, Washington, DC, United States
| | - James D. Oliver
- Department of Medicine, Uniformed Services University of Health Sciences, Bethesda, MD, United States
- Nephrology Service, Walter Reed National Military Medical Center, Bethesda, MD, United States
| | - Ian J. Stewart
- Department of Medicine, Uniformed Services University of Health Sciences, Bethesda, MD, United States
| | - Mark A. Knepper
- System Biology Center, National Heart Lung and Blood Institute, Bethesda, MD, United States
| | - Courtney Fitzhugh
- Cellular and Molecular Therapeutic Branch, National Heart Lung and Blood Institute, Bethesda, MD, United States
| | - Xiaoming Zhou
- Department of Medicine, Uniformed Services University of Health Sciences, Bethesda, MD, United States
| |
Collapse
|
13
|
LaCroix IS, Cralley A, Moore EE, Cendali FI, Dzieciatkowska M, Hom P, Mitra S, Cohen M, Silliman C, Sauaia A, Hansen KC, D’Alessandro A. Omics Signatures of Tissue Injury and Hemorrhagic Shock in Swine. Ann Surg 2023; 278:e1299-e1312. [PMID: 37334680 PMCID: PMC10728352 DOI: 10.1097/sla.0000000000005944] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
OBJECTIVE Advanced mass spectrometry methods were leveraged to analyze both proteomics and metabolomics signatures in plasma upon controlled tissue injury (TI) and hemorrhagic shock (HS)-isolated or combined-in a swine model, followed by correlation to viscoelastic measurements of coagulopathy via thrombelastography. BACKGROUND TI and HS cause distinct molecular changes in plasma in both animal models and trauma patients. However, the contribution to coagulopathy of trauma, the leading cause of preventable mortality in this patient population remains unclear. The recent development of a swine model for isolated or combined TI+HS facilitated the current study. METHODS Male swine (n=17) were randomized to either isolated or combined TI and HS. Coagulation status was analyzed by thrombelastography during the monitored time course. The plasma fractions of the blood draws (at baseline; end of shock; and at 30 minutes, 1, 2, and 4 hours after shock) were analyzed by mass spectrometry-based proteomics and metabolomics workflows. RESULTS HS-isolated or combined with TI-caused the most severe omic alterations during the monitored time course. While isolated TI delayed the activation of coagulation cascades. Correlation to thrombelastography parameters of clot strength (maximum amplitude) and breakdown (LY30) revealed signatures of coagulopathy which were supported by analysis of gene ontology-enriched biological pathways. CONCLUSION The current study provides a comprehensive characterization of proteomic and metabolomic alterations to combined or isolated TI and HS in a swine model and identifies early and late omics correlates to viscoelastic measurements in this system.
Collapse
Affiliation(s)
- Ian S. LaCroix
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - Alexis Cralley
- Department of Surgery, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Ernest E. Moore
- Department of Surgery, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
- Ernest E Moore Shock Trauma Center at Denver Health, Denver, CO, USA
| | - Francesca I Cendali
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - Patrick Hom
- Department of Surgery, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Sanchayita Mitra
- Department of Surgery, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | | | - Christopher Silliman
- Vitalant Research Institute, Denver, CO, USA
- Department of Pediatrics, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Angela Sauaia
- Ernest E Moore Shock Trauma Center at Denver Health, Denver, CO, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
14
|
Aboderin FI, Oduola T, Davison GM, Oguntibeju OO. A Review of the Relationship between the Immune Response, Inflammation, Oxidative Stress, and the Pathogenesis of Sickle Cell Anaemia. Biomedicines 2023; 11:2413. [PMID: 37760854 PMCID: PMC10525295 DOI: 10.3390/biomedicines11092413] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/09/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
Sickle cell anaemia (SCD) is a life-threatening haematological disorder which is predominant in sub-Saharan Africa and is triggered by a genetic mutation of the β-chain haemoglobin gene resulting in the substitution of glutamic acid with valine. This mutation leads to the production of an abnormal haemoglobin molecule called haemoglobin S (HbS). When deoxygenated, haemoglobin S (HbS) polymerises and results in a sickle-shaped red blood cell which is rigid and has a significantly shortened life span. Various reports have shown a strong link between oxidative stress, inflammation, the immune response, and the pathogenesis of sickle cell disease. The consequence of these processes leads to the development of vasculopathy (disease of the blood vessels) and several other complications. The role of the immune system, particularly the innate immune system, in the pathogenesis of SCD has become increasingly clear in recent years of research; however, little is known about the roles of the adaptive immune system in this disease. This review examines the interaction between the immune system, inflammation, oxidative stress, blood transfusion, and their effects on the pathogenesis of sickle cell anaemia.
Collapse
Affiliation(s)
- Florence Ifechukwude Aboderin
- Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville 7535, South Africa;
| | - Taofeeq Oduola
- Department of Chemical Pathology, Usmanu Danfodiyo University, Sokoto 840004, Nigeria;
| | - Glenda Mary Davison
- SAMRC/CPUT Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville 7535, South Africa;
| | - Oluwafemi Omoniyi Oguntibeju
- Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville 7535, South Africa;
| |
Collapse
|
15
|
Naramreddy S, Varma A, Taksande A, Meshram RJ. The Role of Antiplatelet in the Management of Sickle Cell Disease Patients. Cureus 2023; 15:e42058. [PMID: 37602132 PMCID: PMC10434724 DOI: 10.7759/cureus.42058] [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: 07/05/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Sickle cell disease (SCD) is a genetic disorder characterized by abnormal hemoglobin, leading to red blood cell deformities and subsequent vaso-occlusive events. Platelet activation and adhesion play a significant role in the pathophysiology of SCD, contributing to the development of complications such as vaso-occlusive events, stroke, acute chest syndrome, and other manifestations. Antiplatelet therapy has emerged as a potential strategy to mitigate these complications by modulating the platelet function and reducing thrombotic events. This review article provides an overview of antiplatelet therapy's role in managing SCD patients. It discusses the pathophysiological abnormalities in the platelet function in SCD, the rationale for antiplatelet therapy, and the evidence supporting its use in various clinical scenarios. The article explores aspirin as the primary antiplatelet agent in SCD, including its mechanism of action, dosing considerations, and efficacy and safety data. Additionally, it highlights other antiplatelet agents, such as clopidogrel, prasugrel, ticagrelor, and emerging therapies under investigation. Clinical applications of antiplatelet therapy in primary and secondary prevention and the management of acute chest syndrome and other SCD complications are also discussed. Safety considerations are emphasized, including bleeding risk assessment, monitoring, and patient selection for antiplatelet therapy. Finally, the review highlights future research and clinical practice directions, including the development of novel antiplatelet agents, combination therapies, and the integration of antiplatelet therapy with other SCD treatments. Overall, this review provides a comprehensive understanding of the current role of antiplatelet therapy in SCD management, the challenges faced, and future directions for improving patient outcomes.
Collapse
Affiliation(s)
- Sudheeshreddy Naramreddy
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Ashish Varma
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Amar Taksande
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Revat J Meshram
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
16
|
Bhasin N, Sarode R. Acute Chest Syndrome in Sickle Cell Disease. Transfus Med Rev 2023; 37:150755. [PMID: 37741793 DOI: 10.1016/j.tmrv.2023.150755] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/25/2023]
Abstract
Acute chest syndrome (ACS) is the leading cause of mortality among individuals with sickle cell disease (SCD) accounting for 25% of all deaths. The etiologies and clinical manifestations of ACS are variable among children and adults, with a lack of clear risk stratification guidelines for the practicing clinician. In addition, the management of ACS is based on limited evidence and is currently guided primarily by expert opinion. This manuscript reviews the pathophysiology, risk factors, and current management strategies for ACS through a review of published data on this subject between 1988 and 2022. Blood transfusion is often used as a therapeutic intervention for ACS to increase blood's oxygen-carrying capacity and reduce complications by reducing hemoglobin S (HbS) percentage, based on the very low quality of the evidence about its efficacy. The benefit of RBC transfusion for ACS has been described in case series and observational studies, but randomized studies comparing simple transfusion vs. exchange transfusions for ACS are lacking. In this review, we conclude that the development of clinical and laboratory risk stratification is necessary to further study an optimal management strategy for individuals with ACS to avoid transfusion-related complications while minimizing mortality.
Collapse
Affiliation(s)
- Neha Bhasin
- Division of Hematology, Department of Pediatrics, UCSF Benioff Children's Hospital Oakland, USA.
| | - Ravi Sarode
- Department of Pathology and Internal Medicine (Hematology/Oncology), UT Southwestern Medical Center, USA
| |
Collapse
|
17
|
Giriraja KV, Bhatnagar SK, Tomlinson L, Sancilio F. An open-label, multicenter, phase 2 study of a food enriched with docosahexaenoic acid in adults with sickle cell disease. Prostaglandins Leukot Essent Fatty Acids 2023; 193:102574. [PMID: 37121179 DOI: 10.1016/j.plefa.2023.102574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 05/02/2023]
Abstract
Sickle cell disease (SCD) induces red blood cell sickling, which causes debilitating symptoms including vaso-occlusion and inflammation. We investigated a food enriched with omega-3 fatty acids to determine its effect on certain factors: blood cell membrane fatty acid composition (including anti-inflammatory elements-docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA)-and the pro-inflammatory, arachidonic acid (AA)); the inflammation biomarker, C-reactive protein (CRP); and vaso-occlusive crisis (VOC) pain. Ten adults with SCD ingested the food, daily, for 28 days. Evaluated measures included blood cell membrane fatty acid ratios (AA vs omega-3 (DHA+EPA)), CRP (mg/L) levels, and Visual Analogue Scale (VAS) scores (a VOC assessment). The food was well tolerated and led to a statistically significant CRP reduction (39%). However, changes in omega-3 fatty acid ratios and VAS scores were not significant. Overall, while the omega-3-enriched food reduced inflammation, larger, blinded studies are needed to assess its effectiveness on other measures.
Collapse
Affiliation(s)
- K V Giriraja
- Rajalakshmi Hospital & Research Centre, Bangalore, Karnataka, 560097, India
| | - S K Bhatnagar
- Abhinav Multi-specialty Hospital, Nagpur, Maharashtra, 440017, India
| | - L Tomlinson
- Sancilio & Company, Inc. Stuart, Florida, 34997, United States of America
| | - F Sancilio
- Sancilio & Company, Inc. Stuart, Florida, 34997, United States of America; Florida Atlantic University, Boca Raton, Florida, 33431, United States of America.
| |
Collapse
|
18
|
Olasupo MO, Fagbenro DA, Oluwagbamila B, Olasupo MG. Emotional reaction to pain as predictor of depression among selected Nigerians living with sickle cell disease in Ile-Ife. PSYCHOL HEALTH MED 2023; 28:1030-1038. [PMID: 35607734 DOI: 10.1080/13548506.2022.2078496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This study examined the predictive role of emotional reaction to pain on depression and investigated if there are significant age differences in depression among people living with sickle cell disease. A cross-sectional design carried out at Obafemi Awolowo University Health Centre (OAUHC) in Osun State, Nigeria, was conveniently used to select 71 respondents (females = 70.4%), with a median age of 19 years (SD = 5.94). Beck Depression Inventory (BDI) and Short-Form McGill Pain Questionnaire (SF-MPQ) were used to collect data from the respondents from 11 January to 15 February 2019. Simple linear regression analysis revealed that emotional reaction to pain significantly predicts depression among individuals living with sickle cell disease (R2 = 0.16, F(1, 69) = 16.70, p < .05)). One-way ANOVA results also showed a significant influence of age on depression (F(2, 68) = 4.439; p <.05)). The study concluded that emotional reaction to pain and age play significant roles in depression among people living with sickle cell disease in the study setting.
Collapse
Affiliation(s)
- Matthew O Olasupo
- Department of Psychology, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Dare A Fagbenro
- Department of Psychology, Obafemi Awolowo University, Ile-Ife, Nigeria
| | | | | |
Collapse
|
19
|
Patel S, Chandrakar D, Wasnik PN, Nayak S, Shah S, Nanda R, Mohapatra E. Altered T-cell profile in sickle cell disease. Biomark Med 2023; 17:241-252. [PMID: 37204241 DOI: 10.2217/bmm-2023-0086] [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: 05/20/2023] Open
Abstract
Background: Impaired immune status due to altered T-cell response in sickle cell disease (SCD) might provide substantial insight into immune activity in SCD patients. Materials & methods: A total of 30 healthy control, 20 SCD patients in a crisis state and 38 SCD patients in a steady state were evaluated for T-cell subsets. Results: A significant decrease in CD8+ (p = 0.012) and CD8+45RA-197+ (p = 0.015) T-cells were observed among SCD patients. Naive T-cells (45RA+197+; p < 0.01) were elevated and effector (RA-197-) and central memory (RA-197+) T-cells were grossly reduced in the crisis state. Negative regression of naive T-cells with CD8+57+ affirmed immune inactivation. The predictor score reflected 100% sensitivity for predicting the crisis state (area under the curve = 0.851; p < 0.001). Conclusion: Monitoring naive T-cells with predictive scores can help assess the early shift from a steady state to a crisis state.
Collapse
Affiliation(s)
- Suprava Patel
- Department of Biochemistry, AIIMS, Raipur, Chhattisgarh, 492099, India
| | - Diksha Chandrakar
- Department of Biochemistry, AIIMS, Raipur, Chhattisgarh, 492099, India
| | - Preetam N Wasnik
- Department of Medicine, AIIMS, Raipur, Chhattisgarh, 492099, India
| | - Saurav Nayak
- Department of Biochemistry, AIIMS, Bhubaneswar, 751019, Odisha, India
| | - Seema Shah
- Department of Biochemistry, AIIMS, Raipur, Chhattisgarh, 492099, India
| | - Rachita Nanda
- Department of Biochemistry, AIIMS, Raipur, Chhattisgarh, 492099, India
| | - Eli Mohapatra
- Department of Biochemistry, AIIMS, Raipur, Chhattisgarh, 492099, India
| |
Collapse
|
20
|
Man Y, Kucukal E, Liu S, An R, Goreke U, Wulftange WJ, Sekyonda Z, Bode A, Little JA, Manwani D, Stavrou EX, Gurkan UA. A microfluidic device for assessment of E-selectin-mediated neutrophil recruitment to inflamed endothelium and prediction of therapeutic response in sickle cell disease. Biosens Bioelectron 2023; 222:114921. [PMID: 36521205 PMCID: PMC9850363 DOI: 10.1016/j.bios.2022.114921] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022]
Abstract
Neutrophil recruitment to the inflamed endothelium is a multistep process and is of utmost importance in the development of the hallmark vaso-occlusive crisis in sickle cell disease (SCD). However, there lacks a standardized, clinically feasible approach for assessing neutrophil recruitment to the inflamed endothelium for individualized risk stratification and therapeutic response prediction in SCD. Here, we describe a microfluidic device functionalized with E-selectin, a critical endothelial receptor for the neutrophil recruitment process, as a strategy to assess neutrophil binding under physiologic flow in normoxia and clinically relevant hypoxia in SCD. We show that hypoxia significantly enhances neutrophil binding to E-selectin and promotes the formation of neutrophil-platelet aggregates. Moreover, we identified two distinct patient populations: a more severe clinical phenotype with elevated lactate dehydrogenase levels and absolute reticulocyte counts but lowered fetal hemoglobin levels associated with constitutively less neutrophil binding to E-selectin. Mechanistically, we demonstrate that the extent of neutrophil activation correlates with membrane L-selectin shedding, resulting in the loss of ligand interaction sites with E-selectin. We also show that inhibition of E-selectin significantly reduces leukocyte recruitment to activated endothelial cells. Our findings add mechanistic insight into neutrophil-endothelial interactions under hypoxia and provide a clinically feasible means for assessing neutrophil binding to E-selectin using clinical whole blood samples, which can help guide therapeutic decisions for SCD patients.
Collapse
Affiliation(s)
- Yuncheng Man
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Erdem Kucukal
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Shichen Liu
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Ran An
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Utku Goreke
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - William J Wulftange
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Zoe Sekyonda
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Allison Bode
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH, USA; Department of Medicine, Hematology and Oncology Division, CWRU School of Medicine, Cleveland, OH, USA
| | - Jane A Little
- Department of Hematology, UNC Blood Research Center, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Deepa Manwani
- Department of Pediatrics, Albert Einstein College of Medicine/Children's Hospital at Montefiore, Bronx, NY, USA
| | - Evi X Stavrou
- Department of Medicine, Hematology and Oncology Division, CWRU School of Medicine, Cleveland, OH, USA; Medicine Service, Section of Hematology-Oncology, Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, OH, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Umut A Gurkan
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
21
|
Bhalla N, Bhargav A, Yadav SK, Singh AK. Allogeneic hematopoietic stem cell transplantation to cure sickle cell disease: A review. Front Med (Lausanne) 2023; 10:1036939. [PMID: 36910492 PMCID: PMC9995916 DOI: 10.3389/fmed.2023.1036939] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/24/2023] [Indexed: 02/25/2023] Open
Abstract
Sickle cell disease (SCD) had first been mentioned in the literature a century ago. Advancement in the molecular basis of the pathophysiology of the disease opens the door for various therapeutic options. Though life-extending treatments are available for treating patients with SCD, allogeneic hematopoietic stem cell transplantation (HSCT) is the only option as of yet. A major obstacle before HSCT to cure patients with SCD is the availability of donors. Matched sibling donors are available only for a small percentage of patients. To expand the donor pool, different contrasting approaches of allogeneic HSCT like T-cell replete and deplete have been tested. None of those tested approaches have been without the risk of GvHD and graft rejection. Other limitations such as transplantation-related infections and organ dysfunction caused by the harsh conditioning regimen need to be addressed on a priority basis. In this review, we will discuss available allogeneic HSCT approaches to cure SCD, as well as recent advancements to make the approach safer. The center of interest is using megadose T-cell-depleted bone marrow in conjugation with donor-derived CD8 veto T cells to achieve engraftment and tolerance across MHC barriers, under reduced intensity conditioning (RIC). This approach is in phase I/II clinical trial at the MD Anderson Cancer Centre and is open to patients with hemoglobinopathies.
Collapse
Affiliation(s)
- Nishka Bhalla
- Centre for Stem Cell Research, Christian Medical College, Vellore, Tamilnadu, India
| | - Anjali Bhargav
- Centre for Stem Cell Research, Christian Medical College, Vellore, Tamilnadu, India
| | | | - Aloukick Kumar Singh
- Centre for Stem Cell Research, Christian Medical College, Vellore, Tamilnadu, India
| |
Collapse
|
22
|
COVID-19 and Sickle Cell Disease in the Province of Quebec, Canada: Outcomes after Two Years of the Pandemic. J Clin Med 2022; 11:jcm11247361. [PMID: 36555978 PMCID: PMC9781039 DOI: 10.3390/jcm11247361] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Patients with sickle cell disease (SCD) are considered at higher risk of severe COVID-19 infection. However, morbidity and mortality rates are variable among countries. To date, there are no published reports that document outcomes of SCD patients with COVID-19 in Canada. METHODS A web-based registry was implemented in June 2020 capturing outcomes of SCD patients with COVID-19 from March 2020 to April 2022 and comparing them to the general population of Quebec, Canada. RESULTS After 24 months of the pandemic, 185 SCD patients with confirmed SARS-CoV-2 infection were included in the registry. Overall, the population was young (median age 12 years old) and had few comorbidities. No deaths were reported. Risk of hospitalization and admission to intensive care unit (ICU) because of COVID-19 was higher in patients with SCD than in the general population (relative risks (RR) 5.15 (95% confidence interval (95% CI) 3.84-6.91), p ˂ 0.001 and 4.56 (95% CI 2.09-9.93) p ˂ 0.001). A history of arterial hypertension or acute chest syndrome in the past 12 months was associated with a higher risk of severe disease (RR = 3.06 (95% CI 1.85-5.06) p = 0.008 and 2.27 (95% CI 1.35-3.83) p = 0.01). Hospitalized patients had lower hemoglobin F than non-hospitalized patients (12% vs. 17%, p = 0.02). For those who had access to vaccination at the time of infection, 25 out of 26 patients were adequately vaccinated and had mild disease. CONCLUSIONS The SCD population is at higher risk of severe disease than the general population. However, we report favorable outcomes as no deaths occurred. Registries will continue to be critical to document the impact of novel COVID-19 specific therapy and vaccines for the SCD population.
Collapse
|
23
|
An R, Gurkan UA. Emerging functional microfluidic assays for the study of thromboinflammation in sickle cell disease. Curr Opin Hematol 2022; 29:327-334. [PMID: 35916533 PMCID: PMC10440906 DOI: 10.1097/moh.0000000000000731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW This review briefly summarizes the significant impact of thromboinflammation in sickle cell disease in relation to recent advances in biomarkers that are used in functional microfluidic assays. RECENT FINDINGS Sickle cell disease (SCD) is an inherited hemoglobinopathy that affects 100 000 Americans and millions worldwide. Patients with SCD exhibit chronic haemolysis, chronic inflammation and thrombosis, and vaso-occlusion, triggering various clinical complications, including organ damage and increased mortality and morbidity. Recent advances in functional microfluidic assays provide direct biomarkers of disease, including abnormal white blood cell and red blood cell adhesion, cell aggregation, endothelial degradation and contraction, and thrombus formation. SUMMARY Novel and emerging functional microfluidic assays are a promising and feasible strategy to comprehensively characterize thromboinflammatory reactions in SCD, which can be used for personalized risk assessment and tailored therapeutic decisions.
Collapse
Affiliation(s)
- Ran An
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Umut A. Gurkan
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
24
|
Diallo L, Guindo A, Kéita I, Baraïka MA, Dembélé AK, Touré BA, Diallo DA. [Platelet count in the steady state phase and clinical severity of sickle cell disease in a reference centre for sickle cell disease in Mali]. Pan Afr Med J 2022; 43:52. [PMID: 36578815 PMCID: PMC9755550 DOI: 10.11604/pamj.2022.43.52.32674] [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: 12/02/2021] [Accepted: 09/11/2022] [Indexed: 12/30/2022] Open
Abstract
Risk factors associated with complications occurring in sickle cell disease are not fully elucidated. The purpose of this study was to evaluate the existence of an association between the clinical severity of sickle cell disease and platelet count in the steady state phase in patients with sickle cell disease followed up at the Center for Research and Control of Sickle Cell Disease in Bamako, Mali. We conducted a retrospective review of 40 medical records of patients aged 5 to 42 years with sickle cell disease at the Center for Research and Control of Sickle Cell Disease in Bamako, Mali. Clinical severity of sickle cell disease was assessed according to the criteria of VOC and/or hospitalizations < 2 or ≥ 2 per year. Data entry was carried out using the Excel 2013 version. The statistical tests used were the Chi2, Student and Mac Nemar tests. Of the 40 patients, 82.5% had haemolytic phenotype and 17.5% hyperviscous phenotype; complications of sickle cell disease were more frequent in the haemolytic phenotype group (p < 0.05). There was a significant association between mean platelet count ≥ 450 G/L in the steady state phase and the annual number of CVOs ≥ 2 (p = 0.002). This study shows that mean platelet count ≥ 450 G/L in sickle cell patients in the steady state phase could be a risk factor for the frequent occurrence of CVO. It underlines the importance of conducting prospective studies focusing on both hyperplateletosis and platelet activation markers in larger sample sizes, as well as therapeutic trials involving platelet activation inhibitors, such as Crizanlizumab, a humanised anti-P-selectin monoclonal antibodies.
Collapse
Affiliation(s)
- Lamine Diallo
- Centre de Recherche et de Lutte contre la Drépanocytose, Bamako, Mali
| | - Aldiouma Guindo
- Centre de Recherche et de Lutte contre la Drépanocytose, Bamako, Mali
| | - Ibrahima Kéita
- Centre de Recherche et de Lutte contre la Drépanocytose, Bamako, Mali
| | | | | | | | - Dapa Aly Diallo
- Centre de Recherche et de Lutte contre la Drépanocytose, Bamako, Mali,Corresponding author: Dapa Aly Diallo, Centre de Recherche et de Lutte contre la Drépanocytose, Bamako, Mali.
| |
Collapse
|
25
|
Lee MT, Ogu UO. Sickle Cell Disease in the New Era: Advances in Drug Treatment. Transfus Apher Sci 2022; 61:103555. [DOI: 10.1016/j.transci.2022.103555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
26
|
Direct Oral Anticoagulants in Sickle Cell Disease: A Systematic Review and Meta-Analysis. Blood Adv 2022; 6:5061-5066. [PMID: 35728061 PMCID: PMC9631619 DOI: 10.1182/bloodadvances.2022007308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/02/2022] [Indexed: 11/20/2022] Open
|
27
|
Richter RP, Payne GA, Ambalavanan N, Gaggar A, Richter JR. The endothelial glycocalyx in critical illness: A pediatric perspective. Matrix Biol Plus 2022; 14:100106. [PMID: 35392182 PMCID: PMC8981764 DOI: 10.1016/j.mbplus.2022.100106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 12/18/2022] Open
Abstract
The vascular endothelium is the interface between circulating blood and end organs and thus has a critical role in preserving organ function. The endothelium is lined by a glycan-rich glycocalyx that uniquely contributes to endothelial function through its regulation of leukocyte and platelet interactions with the vessel wall, vascular permeability, coagulation, and vasoreactivity. Degradation of the endothelial glycocalyx can thus promote vascular dysfunction, inflammation propagation, and organ injury. The endothelial glycocalyx and its role in vascular pathophysiology has gained increasing attention over the last decade. While studies characterizing vascular glycocalyx injury and its downstream consequences in a host of adult human diseases and in animal models has burgeoned, studies evaluating glycocalyx damage in pediatric diseases are relatively few. As children have unique physiology that differs from adults, significant knowledge gaps remain in our understanding of the causes and effects of endothelial glycocalyx disintegrity in pediatric critical illness. In this narrative literature overview, we offer a unique perspective on the role of the endothelial glycocalyx in pediatric critical illness, drawing from adult and preclinical data in addition to pediatric clinical experience to elucidate how marked derangement of the endothelial surface layer may contribute to aberrant vascular biology in children. By calling attention to this nascent field, we hope to increase research efforts to address important knowledge gaps in pediatric vascular biology that may inform the development of novel therapeutic strategies.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme 2
- CD, cell differentiation marker
- COVID-19, coronavirus disease 2019
- CPB, cardiopulmonary bypass
- CT, component therapy
- Children
- Critical illness
- DENV NS1, dengue virus nonstructural protein 1
- DM, diabetes mellitus
- ECLS, extracorporeal life support
- ECMO, extracorporeal membrane oxygenation
- EG, endothelial glycocalyx
- Endothelial glycocalyx
- FFP, fresh frozen plasma
- GAG, glycosaminoglycan
- GPC, glypican
- HPSE, heparanase
- HSV, herpes simplex virus
- IV, intravenous
- MIS-C, multisystem inflammatory syndrome in children
- MMP, matrix metalloproteinase
- Pragmatic, Randomized Optimal Platelet and Plasma Ratios
- RHAMM, receptor for hyaluronan-mediated motility
- S protein, spike protein
- SAFE, Saline versus Albumin Fluid Evaluation
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- SDC, syndecan
- SDF, sidestream darkfield
- SIRT1, sirtuin 1
- TBI, traumatic brain injury
- TBSA, total body surface area
- TMPRSS2, transmembrane protease serine 2
- Th2, type 2 helper T cell
- VSMC, vascular smooth muscle cell
- Vascular biology
- WB+CT, whole blood and component therapy
- eNOS, endothelial nitric oxide synthase
Collapse
Affiliation(s)
- Robert P. Richter
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Injury Science, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory A. Payne
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Namasivayam Ambalavanan
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Translational Research in Normal and Disordered Development Program, University of Alabama, Birmingham, AL, USA
| | - Amit Gaggar
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jillian R. Richter
- Center for Injury Science, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
28
|
Scarpato B, Strykowski R, Lawrence R, Khan SL, Newman J, Spring MR, Gupta VK, Patel J, Cohen RT, Sloan JM, Nouraie SM, Klings ES. Risk factors for Venous Thromboembolism and clinical outcomes in adults with sickle cell disease. THROMBOSIS UPDATE 2022. [DOI: 10.1016/j.tru.2022.100101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
29
|
Abdel-Bakky MS, Amin E, Ewees MG, Mahmoud NI, Mohammed HA, Altowayan WM, Abdellatif AAH. Coagulation System Activation for Targeting of COVID-19: Insights into Anticoagulants, Vaccine-Loaded Nanoparticles, and Hypercoagulability in COVID-19 Vaccines. Viruses 2022; 14:228. [PMID: 35215822 PMCID: PMC8876839 DOI: 10.3390/v14020228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/06/2022] [Accepted: 01/21/2022] [Indexed: 01/08/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), also known as COVID-19, is currently developing into a rapidly disseminating and an overwhelming worldwide pandemic. In severe COVID-19 cases, hypercoagulability and inflammation are two crucial complications responsible for poor prognosis and mortality. In addition, coagulation system activation and inflammation overlap and produce life-threatening complications, including coagulopathy and cytokine storm, which are associated with overproduction of cytokines and activation of the immune system; they might be a lead cause of organ damage. However, patients with severe COVID-19 who received anticoagulant therapy had lower mortality, especially with elevated D-dimer or fibrin degradation products (FDP). In this regard, the discovery of natural products with anticoagulant potential may help mitigate the numerous side effects of the available synthetic drugs. This review sheds light on blood coagulation and its impact on the complication associated with COVID-19. Furthermore, the sources of natural anticoagulants, the role of nanoparticle formulation in this outbreak, and the prevalence of thrombosis with thrombocytopenia syndrome (TTS) after COVID-19 vaccines are also reviewed. These combined data provide many research ideas related to the possibility of using these anticoagulant agents as a treatment to relieve acute symptoms of COVID-19 infection.
Collapse
Affiliation(s)
- Mohamed S. Abdel-Bakky
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 52471, Saudi Arabia;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
| | - Elham Amin
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt;
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 52471, Saudi Arabia;
| | - Mohamed G. Ewees
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 11787, Egypt; (M.G.E.); (N.I.M.)
| | - Nesreen I. Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 11787, Egypt; (M.G.E.); (N.I.M.)
| | - Hamdoon A. Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 52471, Saudi Arabia;
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
| | - Waleed M. Altowayan
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Qassim 52471, Saudi Arabia;
| | - Ahmed A. H. Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Qasssim 52471, Saudi Arabia
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| |
Collapse
|
30
|
Narciso MG, Hoeting B, James JM, VandenHeuvel K, Nasimuzzaman M. A mutant fibrinogen that is unable to form fibrin can improve renal phenotype in mice with sickle cell anemia. EJHAEM 2021; 2:462-465. [PMID: 35844706 PMCID: PMC9176143 DOI: 10.1002/jha2.204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/10/2021] [Accepted: 04/11/2021] [Indexed: 11/15/2022]
Abstract
Sickle cell anemia (SCA) causes nephropathy which may progress to kidney failure. To determine if soluble fibrinogen (FibAEK) can prevent kidney damage in mice with SCA, we performed bone marrow transplantation (BMT) of Berkeley sickle mice into wild-type fibrinogen (FibWT), and FibAEK mice that bear a germ-line mutation in fibrinogen Aα chain at thrombin cleavage site which prevents fibrin formation. We found improved albuminuria in SS FibAEK mice compared with SS FibWT mice at 12 months post-BMT due to the reduced kidney fibrosis, ischemic lesions, and increased survival of podocytes in the glomeruli, but did not improve urine concentrating defect. Therefore, our study clarifies the distinct role of fibrinogen and fibrin in the renal pathology of SCA.
Collapse
Affiliation(s)
- Marilou G. Narciso
- Division of Experimental Hematology and Cancer BiologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Blair Hoeting
- Division of Experimental Hematology and Cancer BiologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Jeanne M. James
- Division Pediatric CardiologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Katherine VandenHeuvel
- Pathology CoreCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- University of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Md. Nasimuzzaman
- Division of Experimental Hematology and Cancer BiologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- University of Cincinnati College of MedicineCincinnatiOhioUSA
| |
Collapse
|
31
|
Alpha thalassemia, but not β S-globin haplotypes, influence sickle cell anemia clinical outcome in a large, single-center Brazilian cohort. Ann Hematol 2021; 100:921-931. [PMID: 33586016 DOI: 10.1007/s00277-021-04450-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 02/03/2021] [Indexed: 12/13/2022]
Abstract
Alpha thalassemia and beta-globin haplotype are considered classical genetic disease modifiers in sickle cell anemia (SCA) causing clinical heterogeneity. Nevertheless, their functional impact on SCA disease emergence and progression remains elusive. To better understand the role of alpha thalassemia and beta-globin haplotype in SCA, we performed a retrospective study evaluating the clinical manifestations of 614 patients. The univariate analysis showed that the presence of alpha-thalassemia -3.7-kb mutation (αα/-α and -α/-α) decreased the risk of stroke development (p = 0.046), priapism (p = 0.033), and cholelithiasis (p = 0.021). Furthermore, the cumulative incidence of stroke (p = 0.023) and cholelithiasis (p = 0.006) was also significantly lower for patients carrying the alpha thalassemia -3.7-kb mutation. No clinical effects were associated with the beta-globin haplotype analysis, which could be explained by the relatively homogeneous haplotype composition in our cohort. Our results reinforce that alpha thalassemia can provide protective functions against hemolysis-related symptoms in SCA. Although, several genetic modifiers can impact the inflammatory state of SCA patients, the alpha thalassemia mutation remains one of the most recurrent genetic aberration and should therefore always be considered first.
Collapse
|
32
|
Hopp MT, Imhof D. Linking Labile Heme with Thrombosis. J Clin Med 2021; 10:427. [PMID: 33499296 PMCID: PMC7865584 DOI: 10.3390/jcm10030427] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
Thrombosis is one of the leading causes of death worldwide. As such, it also occurs as one of the major complications in hemolytic diseases, like hemolytic uremic syndrome, hemorrhage and sickle cell disease. Under these conditions, red blood cell lysis finally leads to the release of large amounts of labile heme into the vascular compartment. This, in turn, can trigger oxidative stress and proinflammatory reactions. Moreover, the heme-induced activation of the blood coagulation system was suggested as a mechanism for the initiation of thrombotic events under hemolytic conditions. Studies of heme infusion and subsequent thrombotic reactions support this assumption. Furthermore, several direct effects of heme on different cellular and protein components of the blood coagulation system were reported. However, these effects are controversially discussed or not yet fully understood. This review summarizes the existing reports on heme and its interference in coagulation processes, emphasizing the relevance of considering heme in the context of the treatment of thrombosis in patients with hemolytic disorders.
Collapse
Affiliation(s)
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany;
| |
Collapse
|
33
|
Demagny J, Driss A, Stepanian A, Anguel N, Affo L, Roux D, Habibi A, Benghezal S, Capdenat S, Coppo P, Driss F, Veyradier A. ADAMTS13 and von Willebrand factor assessment in steady state and acute vaso-occlusive crisis of sickle cell disease. Res Pract Thromb Haemost 2021; 5:197-203. [PMID: 33537544 PMCID: PMC7845082 DOI: 10.1002/rth2.12460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/29/2020] [Accepted: 11/05/2020] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Sickle cell disease (SCD) is characterized by vaso-occlusive crisis (VOC), acute chest syndrome (ACS) and multiorgan failure (MOF) complicated by thrombosis. Von Willebrand factor (VWF) is a strong marker of SCD-related endothelial injury. OBJECTIVES To decipher the role of VWF and its specific-cleaving metalloprotease, ADAMTS13, in the vaso-occlusive and thrombotic process of SCD. PATIENTS/METHODS We investigated the VWF antigen (Ag), ADAMTS13 activity, ADAMTS13 Ag and ADAMTS13 IgGs in a cohort of 65 patients with SCD prospectively enrolled in a 20-month period from three centers. Patients were divided into two groups: an asymptomatic group (n = 30) with treated or untreated SCD at steady state, and a VOC/ACS group (n = 35) with SCD with VOC/ACS requiring either medical management or intensive care management for MOF. RESULTS AND CONCLUSIONS VWF:Ag levels were increased (median, 167 IU/dL; interquartile range [IQR], 124 - 279), especially in patients with VOC SCD (227 IU/dL; IQR, 134-305; P = .04), and positively correlated with inflammatory markers (P < .02). Median ADAMTS13 activity was normal (70 IU/dL; IQR, 60-80), but 7 patients exhibited a partial deficiency between 25 and 45 IU/dL. ADAMTS13 activity/VWF:Ag ratio, however, did not change during VOC. Median ADAMTS13:Ag was slightly decreased (611 ng/mL; IQR, 504-703) with no significant difference between groups. Surprisingly, ADAMTS13 IgGs were detected in 33 (51%) of our patients. We conclude that, in SCD, VWF:Ag and nonrelevant ADAMTS13 IgGs may reflect the severity of the inflammatory vasculopathy enhancing vaso-occlusive and thrombotic complications.
Collapse
Affiliation(s)
- Julien Demagny
- Service d’Hématologie BiologiqueHôpital LariboisièreAP‐HP.NordUniversité de ParisParisFrance
| | - Aurélie Driss
- Service d’HémaphérèseHôpital de BicêtreAP‐HP.SudUniversité Paris SaclayLe Kremlin BicêtreFrance
| | - Alain Stepanian
- Service d’Hématologie BiologiqueHôpital LariboisièreAP‐HP.NordUniversité de ParisParisFrance
| | - Nadia Anguel
- Service de Réanimation MédicaleHôpital de BicêtreAP‐HP.SudUniversité Paris SaclayLe Kremlin BicêtreFrance
| | - Louis Affo
- Service de Médecine InterneHôpital Louis MourierAP‐HP.NordUniversité de ParisParisFrance
| | - Damien Roux
- Service de Réanimation Médico‐ChirurgicaleHôpital Louis MourierAP‐HP.NordUniversité de ParisParisFrance
| | - Anoosha Habibi
- Unité des maladies du globule rougeHôpital Henri MondorAP‐HPUniversité Paris Est Créteil‐Val de marneParisFrance
| | - Sandrine Benghezal
- Service d’Hématologie BiologiqueHôpital LariboisièreAP‐HP.NordUniversité de ParisParisFrance
| | - Sophie Capdenat
- Service d’Hématologie BiologiqueHôpital LariboisièreAP‐HP.NordUniversité de ParisParisFrance
| | - Paul Coppo
- Centre National de Référence des Microangiopathies Thrombotiques (CNR‐MAT)Département D’hématologie cliniqueHôpital Saint AntoineAPHP.SUUniversité Pierre et Marie CurieParisFrance
| | - Françoise Driss
- Service d’HémaphérèseHôpital de BicêtreAP‐HP.SudUniversité Paris SaclayLe Kremlin BicêtreFrance
| | - Agnès Veyradier
- Service d’Hématologie BiologiqueHôpital LariboisièreAP‐HP.NordUniversité de ParisParisFrance
| |
Collapse
|
34
|
Association between inflammatory molecules, nitric oxide metabolites and leg ulcers in individuals with sickle cell anemia. Hematol Transfus Cell Ther 2020; 44:169-176. [PMID: 33371972 PMCID: PMC9123559 DOI: 10.1016/j.htct.2020.09.152] [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: 05/25/2020] [Revised: 07/13/2020] [Accepted: 09/19/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction Leg ulcers (LUs) are relatively common in patients with sickle cell anemia (SCA). The role of inflammation and nitric oxide (NO) pathways in the pathophysiology of the LU is not understood. Objective The aim of this study was to verify the association between inflammatory molecules and nitric oxide metabolites (NOx) and the occurrence of the LU in patients with SCA. Method It was a cross-sectional study on adult participants with SCA followed at Fundação Hemominas, a public blood center in Brazil. Eligible participants were recruited and included in one of two groups: Group 1, comprised of cases with SCA (Hb SS) and at least one LU at the time of inclusion in the study and Group 2, comprised of controls with SCA without a history of LU, matched by sex and age to cases. Participants were interviewed to obtain sociodemographic data and blood samples were collected. Clinical and laboratory data were abstracted from medical records. Nitric oxide metabolites (NOx) and inflammatory molecules were quantified using an immunoassay and Multiplex xMAP® technology, respectively. Eighty-seven individuals were included, ranging in age from 17 to 61 years (mean 40 ± 10.7 years); 30 had LU and 57 were controls without LU. Results Participants with LU had significantly higher levels of interleukin 8 (IL-8), IL-10, IL-15, NOx and platelet and white blood cell (WBC) counts, when compared to those without LU. Participants with LU had a significantly higher risk of having a history of osteomyelitis and a higher use of antiseptic soap in bathing, when compared to those without LU. Conclusion In conclusion, our results showed that NOx, inflammatory molecules and hematological features were associated with LU in Brazilian adults with SCA.
Collapse
|
35
|
Della-Moretta S, Delatore L, Purcell M, Huang Y, Heinlein M, Adkins E, Desai P. The Effect of Use of Individualized Pain Plans in Sickle Cell Patients Presenting to the Emergency Department. Ann Emerg Med 2020; 76:S21-S27. [PMID: 32928458 DOI: 10.1016/j.annemergmed.2020.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
STUDY OBJECTIVE Sickle cell disease (SCD) is an inherited hematologic disorder that affects approximately 100,000 US individuals and results in greater than 200,000 emergency department (ED) visits annually in the United States, with pain being the most common complaint. The objective of this retrospective study is to determine the effect of implementing individualized pain plans in the treatment of patients with SCD in the ED on time to first opioid, length of stay, and disposition. METHODS At The Ohio State University Wexner Medical Center, a multidisciplinary group including hematologists and ED physicians was formed and enacted a protocol for using individualized pain plans, with the goal of decreasing time to treatment for patients with SCD who presented to the ED with chief complaint of pain. In this retrospective study, data from the year before through the year of implementation were gathered. Generalized linear models were fit to compare time to first opioid, length of stay, and disposition before and after protocol implementation. RESULTS Data showed a 48% decrease in time to first opioid and a 22% decrease in length of ED stay after protocol implementation. No significant change was found in disposition or length of inpatient admission before and after protocol initiation. CONCLUSION The use of individualized pain plans in the treatment of patients with SCD in the ED is a useful method of not only ensuring rapid and adequate treatment but also decreasing use of health care resources.
Collapse
Affiliation(s)
| | | | | | - Ying Huang
- Department of Internal Medicine, Columbus, OH
| | - Melanie Heinlein
- Wexner Medical Center at the Ohio State University, Columbus, OH
| | - Eric Adkins
- Department of Emergency Medicine, Columbus, OH
| | - Payal Desai
- Department of Internal Medicine, Columbus, OH; Division of Hematolgy, Columbus, OH.
| |
Collapse
|
36
|
Ladeira VS, de Oliveira Toledo SL, Ferreira LGR, Oliveira MM, Silva APF, de Oliveira WV, Duarte RCF, Renó CDO, Dusse LMS, Dos Santos HL, Carvalho MDG, Pinheiro MDB, Rios DRA. Thrombin generation in vivo and ex vivo in sickle cell disease patients. Thromb Res 2020; 197:165-171. [PMID: 33221576 DOI: 10.1016/j.thromres.2020.10.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/26/2020] [Accepted: 10/29/2020] [Indexed: 12/18/2022]
Abstract
Activation of coagulation is an important hallmark of sickle cell disease (SCD) and it is believed that hypercoagulability plays a role to the disease pathophysiology. Studies have sought to identify how hemostatic biomarkers are expressed in SCD, however, the results are inconclusive. In this context, our objective was to evaluate the thrombin generation in vivo and ex vivo in SCD patients and the association between these biomarkers and the use of HU. This cross-sectional study was carried out with patients diagnosed with SCD, users or not of Hydroxyurea (HU), and healthy individuals as controls. D dimer (D-Di) was evaluated by ELISA and (TGT) thrombin generation test by CAT method. D-Di plasma levels were significantly higher in SCD patients when compared to the controls. TGT parameters such as peak, ETP and normalized ETP at low TF concentration and time-to-peak, peak, ETP and normalized ETP values at high TF concentration were lower in SCD patients than in controls. In contrast, the normalized activated protein C sensitivity ratio (nAPCsr) was higher in patients compared to controls, indicating resistance to the action of this natural anticoagulant. Regarding the use of HU, comparing users and non-users of this drug, no difference was observed in D-Di levels and in most TGT parameters. Our data analyzed together allow us to conclude that patients with SCD present a state of hypercoagulability in vivo due to the higher levels of D-Di and resistance to APC assessed ex vivo which is consistent with the coagulation imbalance described in SCD patients.
Collapse
Affiliation(s)
- Valéria Sutana Ladeira
- Universidade Federal de São João del-Rei, Campus Centro Oeste Dona Lindu, Brazil; Fundação Hemominas, Minas Gerais, Brazil
| | | | | | - Marina Mendes Oliveira
- Universidade Federal de São João del-Rei, Campus Centro Oeste Dona Lindu, Brazil; Fundação Hemominas, Minas Gerais, Brazil
| | | | | | | | | | | | | | - Maria das Graças Carvalho
- Universidade Federal de São João del-Rei, Campus Centro Oeste Dona Lindu, Brazil; Faculdade de Farmácia, Universidade Federal de Minas Gerais, Brazil
| | | | | |
Collapse
|
37
|
Shet AS, Lizarralde-Iragorri MA, Naik RP. The molecular basis for the prothrombotic state in sickle cell disease. Haematologica 2020; 105:2368-2379. [PMID: 33054077 PMCID: PMC7556662 DOI: 10.3324/haematol.2019.239350] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/22/2020] [Indexed: 12/15/2022] Open
Abstract
The genetic and molecular basis of sickle cell disease (SCD) has long since been characterized but the pathophysiological basis is not entirely defined. How a red cell hemolytic disorder initiates inflammation, endothelial dysfunction, coagulation activation and eventually leads to vascular thrombosis, is yet to be elucidated. Recent evidence has demonstrated a high frequency of unprovoked/recurrent venous thromboembolism (VTE) in SCD, with an increased risk of mortality among patients with a history of VTE. Here, we thoroughly review the molecular basis for the prothrombotic state in SCD, specifically highlighting emerging evidence for activation of overlapping inflammation and coagulation pathways, that predispose to venous thromboembolism. We share perspectives in managing venous thrombosis in SCD, highlighting innovative therapies with the potential to influence the clinical course of disease and reduce thrombotic risk, while maintaining an acceptable safety profile.
Collapse
Affiliation(s)
- Arun S. Shet
- Laboratory of Sickle Thrombosis and Vascular Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda
| | | | - Rakhi P. Naik
- Division of Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
38
|
Conran N, De Paula EV. Thromboinflammatory mechanisms in sickle cell disease - challenging the hemostatic balance. Haematologica 2020; 105:2380-2390. [PMID: 33054078 PMCID: PMC7556678 DOI: 10.3324/haematol.2019.239343] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/19/2020] [Indexed: 11/11/2022] Open
Abstract
Sickle cell disease (SCD) is an inherited hemoglobinopathy that is caused by the presence of abnormal hemoglobin S (HbS) in red blood cells, leading to alterations in red cell properties and shape, as the result of HbS dexoygenation and subsequent polymerization. SCD pathophysiology is characterized by chronic inflammatory processes, triggered by hemolytic and vaso-occlusive events, which lead to the varied complications, organ damage and elevated mortality seen in individuals with the disease. In association with activation of the endothelium and leukocytes, hemostatic alterations and thrombotic events are well-documented in SCD. Here we discuss the role for inflammatory pathways in modulating coagulation and inducing platelet activation in SCD, due to tissue factor activation, adhesion molecule expression, inflammatory mediator production and the induction of innate immune responses, amongst other mechanisms. Thromboinflammatory pathways may play a significant role in some of the major complications of SCD, such as stroke, venous thromboembolism and possibly acute chest syndrome, besides exacerbating the chronic inflammation and cellular interactions that trigger vaso-occlusion, ischemia-reperfusion processes, and eventually organ damage.
Collapse
Affiliation(s)
- Nicola Conran
- Hematology Center, University of Campinas, UNICAMP, Cidade Universitária, Campinas-SP, Brazil
| | - Erich V. De Paula
- Hematology Center, University of Campinas, UNICAMP, Cidade Universitária, Campinas-SP, Brazil
| |
Collapse
|
39
|
Dib PRB, Quirino-Teixeira AC, Merij LB, Pinheiro MBM, Rozini SV, Andrade FB, Hottz ED. Innate immune receptors in platelets and platelet-leukocyte interactions. J Leukoc Biol 2020; 108:1157-1182. [PMID: 32779243 DOI: 10.1002/jlb.4mr0620-701r] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/11/2020] [Accepted: 06/28/2020] [Indexed: 12/14/2022] Open
Abstract
Platelets are chief cells in hemostasis. Apart from their hemostatic roles, platelets are major inflammatory effector cells that can influence both innate and adaptive immune responses. Activated platelets have thromboinflammatory functions linking hemostatic and immune responses in several physiological and pathological conditions. Among many ways in which platelets exert these functions, platelet expression of pattern recognition receptors (PRRs), including TLR, Nod-like receptor, and C-type lectin receptor families, plays major roles in sensing and responding to pathogen-associated or damage-associated molecular patterns (PAMPs and DAMPs, respectively). In this review, an increasing body of evidence is compiled showing the participation of platelet innate immune receptors, including PRRs, in infectious diseases, sterile inflammation, and cancer. How platelet recognition of endogenous DAMPs participates in sterile inflammatory diseases and thrombosis is discussed. In addition, platelet recognition of both PAMPs and DAMPs initiates platelet-mediated inflammation and vascular thrombosis in infectious diseases, including viral, bacterial, and parasite infections. The study also focuses on the involvement of innate immune receptors in platelet activation during cancer, and their contribution to tumor microenvironment development and metastasis. Finally, how innate immune receptors participate in platelet communication with leukocytes, modulating leukocyte-mediated inflammation and immune functions, is highlighted. These cell communication processes, including platelet-induced release of neutrophil extracellular traps, platelet Ag presentation to T-cells and platelet modulation of monocyte cytokine secretion are discussed in the context of infectious and sterile diseases of major concern in human health, including cardiovascular diseases, dengue, HIV infection, sepsis, and cancer.
Collapse
Affiliation(s)
- Paula Ribeiro Braga Dib
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil.,Laboratory of Immunology, Infectious Diseases and Obesity, Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Anna Cecíllia Quirino-Teixeira
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Laura Botelho Merij
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Mariana Brandi Mendonça Pinheiro
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Stephane Vicente Rozini
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Fernanda Brandi Andrade
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eugenio Damaceno Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| |
Collapse
|
40
|
Lizarralde-Iragorri MA, Shet AS. Sickle Cell Disease: A Paradigm for Venous Thrombosis Pathophysiology. Int J Mol Sci 2020; 21:ijms21155279. [PMID: 32722421 PMCID: PMC7432404 DOI: 10.3390/ijms21155279] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023] Open
Abstract
Venous thromboembolism (VTE) is an important cause of vascular morbidity and mortality. Many risk factors have been identified for venous thrombosis that lead to alterations in blood flow, activate the vascular endothelium, and increase the propensity for blood coagulation. However, the precise molecular and cellular mechanisms that cause blood clots in the venous vasculature have not been fully elucidated. Patients with sickle cell disease (SCD) demonstrate all the risk factors for venous stasis, activated endothelium, and blood hypercoagulability, making them particularly vulnerable to VTE. In this review, we will discuss how mouse models have elucidated the complex vascular pathobiology of SCD. We review the dysregulated pathways of inflammation and coagulation in SCD and how the resultant hypercoagulable state can potentiate thrombosis through down-regulation of vascular anticoagulants. Studies of VTE pathogenesis using SCD mouse models may provide insight into the intersection between the cellular and molecular processes involving inflammation and coagulation and help to identify novel mechanistic pathways.
Collapse
|
41
|
Biochemical and therapeutic effects of Omega-3 fatty acids in sickle cell disease. Complement Ther Med 2020; 52:102482. [PMID: 32951732 DOI: 10.1016/j.ctim.2020.102482] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/02/2020] [Accepted: 06/08/2020] [Indexed: 01/29/2023] Open
Abstract
Sickle cell disease (SCD) is a hematologic disorder with complex pathophysiology that includes chronic hemolysis, vaso-occlusion and inflammation. Increased leukocyte-erythrocyte-endothelial interactions, due to upregulated expression of adhesion molecules and activated endothelium, are thought to play a primary role in initiation and progression of SCD vaso-occlusive crisis and end-organ damage. Several new pathophysiology-based therapeutic options for SCD are being developed, chiefly targeting the inflammatory pathways. Omega-3 fatty acids are polyunsaturated fatty acids that are known to have effects on diverse physiological processes. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are the principal biologically active omega-3 fatty acids. The therapeutic effects of DHA and EPA on chronic inflammatory disorders and cardiovascular diseases are well recognized. The therapeutic effects of omega-3 fatty acids are attributed to their anti-inflammatory and anti-thrombotic eicosanoids, and the novel class of EPA and DHA derived lipid mediators: resolvins, protectins and maresins. Blood cell membranes of patients with SCD have abnormal fatty acids composition characterized by high ratio of pro-inflammatory arachidonic acid (AA) to anti-inflammatory DHA and EPA (high omega-6/omega-3 ratio). In addition, experimental and clinical studies provide evidence that treatment with DHA does confer improvement in rheological properties of sickle RBC, inflammation and hemolysis. The clinical studies have shown improvements in VOC rate, markers of inflammation, adhesion, and hemolysis. In toto, the results of studies on the therapeutic effects of omega-3 fatty acids in SCD provide good body of evidence that omega-3 fatty acids could be a safe and effective treatment for SCD.
Collapse
|
42
|
Salinas Cisneros G, Thein SL. Recent Advances in the Treatment of Sickle Cell Disease. Front Physiol 2020; 11:435. [PMID: 32508672 PMCID: PMC7252227 DOI: 10.3389/fphys.2020.00435] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/08/2020] [Indexed: 12/31/2022] Open
Abstract
Sickle cell anemia (SCA) was first described in the Western literature more than 100 years ago. Elucidation of its molecular basis prompted numerous biochemical and genetic studies that have contributed to a better understanding of its pathophysiology. Unfortunately, the translation of such knowledge into developing treatments has been disproportionately slow and elusive. In the last 10 years, discovery of BCL11A, a major γ-globin gene repressor, has led to a better understanding of the switch from fetal to adult hemoglobin and a resurgence of efforts on exploring pharmacological and genetic/genomic approaches for reactivating fetal hemoglobin as possible therapeutic options. Alongside therapeutic reactivation of fetal hemoglobin, further understanding of stem cell transplantation and mixed chimerism as well as gene editing, and genomics have yielded very encouraging outcomes. Other advances have contributed to the FDA approval of three new medications in 2017 and 2019 for management of sickle cell disease, with several other drugs currently under development. In this review, we will focus on the most important advances in the last decade.
Collapse
Affiliation(s)
- Gabriel Salinas Cisneros
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States.,Division of Hematology and Oncology, Children's National Medical Center, Washington, DC, United States
| | - Swee L Thein
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
43
|
Kazak A, Ozkaraman A. The Effect of Progressive Muscle Relaxation Exercises on Pain on Patients with Sickle Cell Disease: Randomized Controlled Study. Pain Manag Nurs 2020; 22:177-183. [PMID: 32224022 DOI: 10.1016/j.pmn.2020.02.069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 02/04/2020] [Accepted: 02/23/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE The study aimed to evaluate the effect of progressive muscle relaxation exercises on pain in patients with sickle cell disease. MATERIALS-METHOD This randomized controlled interventional study was conducted in a hospital in Mersin, Turkey between October 2017 and July 2018. The study sample comprised 58 patients who were aged >18 years, conscious, had sickle cell disease, reported pain, and were treated with non-opioid or weak opioid analgesic based on the physician's recommendation (treatment group = 29, control group = 29). Data were collected by the individual presentation form and visual analog scale. During the study, both groups were treated with analgesics prescribed by the physician for three days. In addition to the analgesics, the treatment group was performed progressive muscle relaxation exercises for 30 minutes whereas the control group was rested. Pain level of both groups was evaluated at three time points every day for three days. In the study, frequency distributions and descriptive statistics were presented for categorical and numerical variables, respectively. Chi-square analysis, independent samples t-test, Mann-Whitney U test, three-way ANOVA, and Sidak test were used to analyze the difference between the variables. RESULTS The mean age of the patients was 29.59 ± 6.94 years, and 53.4% of the patients were female and 69% were single. The mean pain score of the treatment group at the third time point on days 1, 2, and 3 was significantly lower than the control group (p < .05). There was no statistically significant difference between the two groups in terms of coping methods for pain, pain location, and complaints accompanying pain (p > .05). CONCLUSION Progressive muscle relaxation exercises were found to be effective in the pain management of patients with sickle cell anemia.
Collapse
Affiliation(s)
- Aysun Kazak
- Vocational School of Health Services, Gumushane University, Gumushane, Turkey
| | - Ayse Ozkaraman
- Department of Nursing, Eskisehir Osmangazi University, Eskisehir, Turkey.
| |
Collapse
|