1
|
Rehill AM, McCluskey S, Ledwith AE, Ryan TAJ, Ünlü B, Leon G, Charles-Messance H, Gilbert EH, Klavina P, Day EA, Coppinger J, O’Sullivan JM, McMahon C, O’Donnell JS, Curtis AM, O’Neill LAJ, Sheedy FJ, Preston RJS. Trained immunity causes myeloid cell hypercoagulability. SCIENCE ADVANCES 2025; 11:eads0105. [PMID: 40053582 PMCID: PMC11887800 DOI: 10.1126/sciadv.ads0105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025]
Abstract
The pathogenic basis for increased thrombotic risk in individuals with inflammatory diseases is poorly understood. Myeloid cell "trained immunity" describes persistent innate immune cell memory arising from prior exposure to an inflammatory stimulus, leading to an enhanced immune response to subsequent unrelated stimuli. We identify enhanced myeloid cell prothrombotic activity as a maladaptive consequence of trained immunity. Lipopolysaccharide (LPS) stimulation of macrophages trained previously with β-glucan or heme exhibited significantly enhanced procoagulant activity compared to macrophages stimulated with LPS alone, which was mediated by enhanced acid sphingomyelinase-mediated tissue factor decryption. Furthermore, splenic monocytes isolated from β-glucan-trained mice revealed enhanced procoagulant activity up to 4 weeks after β-glucan administration compared to monocytes from control mice over the same time period. Moreover, hematopoietic progenitor cells and bone marrow interstitial fluid from β-glucan-trained mice had enhanced procoagulant activity compared to control mice. Trained immunity and associated metabolic perturbations may therefore represent an opportunity for targeted intervention in immunothrombotic disease development.
Collapse
Affiliation(s)
- Aisling M. Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children’s Research Centre, Our Lady’s Children’s Hospital Crumlin, Dublin, Ireland
| | - Seán McCluskey
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children’s Research Centre, Our Lady’s Children’s Hospital Crumlin, Dublin, Ireland
| | - Anna E. Ledwith
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Tristram A. J. Ryan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Betül Ünlü
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Gemma Leon
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children’s Research Centre, Our Lady’s Children’s Hospital Crumlin, Dublin, Ireland
| | | | - Edmund H. Gilbert
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Paula Klavina
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children’s Research Centre, Our Lady’s Children’s Hospital Crumlin, Dublin, Ireland
| | - Emily A. Day
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Judith Coppinger
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Jamie M. O’Sullivan
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Corrina McMahon
- National Children’s Research Centre, Our Lady’s Children’s Hospital Crumlin, Dublin, Ireland
| | - James S. O’Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Annie M. Curtis
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Luke A. J. O’Neill
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Frederick J. Sheedy
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Roger J. S. Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children’s Research Centre, Our Lady’s Children’s Hospital Crumlin, Dublin, Ireland
| |
Collapse
|
2
|
Yurtsever N, Tong N, Geetha S, Nandi V, Shi PA. Post-exchange neutrophil count, but not post-hematocrit, predicts endogenous erythropoiesis in patients with sickle cell disease undergoing chronic red cell exchange. Transfusion 2024; 64:2270-2278. [PMID: 39404130 DOI: 10.1111/trf.18044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND With chronic transfusion in sickle cell disease (SCD), equipoise exists regarding whether increasing the post-procedure hematocrit (Hct) suppresses endogenous erythropoiesis. Reticulocytosis predicts SCD morbidity and mortality, so this study's objective was to clarify the role of the post-procedure Hct in suppressing reticulocytosis and to identify other potential red cell exchange (RCE) parameters predictive of reticulocytosis. STUDY DESIGN AND METHODS This retrospective analysis of 17 patients who underwent chronic RCE at a single institution between 2014 and 2022 examined both standard red cell exchanges (SRCE) and exchanges preceded by isovolemic hemodilution (IVH-RCE). Post-procedure parameters with biologic plausibility to influence the subsequent procedure's absolute reticulocyte count (sPre-ARC) were examined using regression modeling. RESULTS Neither post-hematocrit, nor post-hemoglobin (Hb), nor ΔHb/day was associated with sPre-ARC or the change in HbS% per day (ΔHbS%/day). Concurrent Hb was predictive for SRCE but not IVH-RCE, where ARC trended lower than with SRCE. Male gender and post-procedure neutrophil and white cell counts were predictors of sPre-ARC, consistent with their associations with SCD morbidity and mortality. IVH-RCE had a stronger correlation than standard RCE between pre-Hct and neutrophil or white cell depletion. DISCUSSION Although targeting a post-procedure Hct maintains a higher subsequent pre-procedure Hb and a lower sPre-HbS%, it does not lead to sustained suppression of reticulocytosis as measured by the sPre-ARC or the ΔHbS%/day. IVH-RCE or the addition of hydroxyurea could be considered in those patients with high reticulocyte, white blood cell, or neutrophil counts.
Collapse
Affiliation(s)
- Nalan Yurtsever
- Department of Pathology and Laboratory Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Nicholas Tong
- Department of Pathology and Laboratory Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Saroja Geetha
- Department of Pathology and Laboratory Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Vijay Nandi
- Clinical Services, New York Blood Center, New York, New York, USA
| | - Patricia A Shi
- Clinical Services, New York Blood Center, New York, New York, USA
- Division of Medical Oncology and Hematology, Monter Cancer Center Northwell Health Cancer Institute, New Hyde Park, New York, USA
| |
Collapse
|
3
|
Tozatto‐Maio K, Rós FA, Weinlich R, Rocha V. Inflammatory pathways and anti-inflammatory therapies in sickle cell disease. Hemasphere 2024; 8:e70032. [PMID: 39698332 PMCID: PMC11655128 DOI: 10.1002/hem3.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 07/12/2024] [Accepted: 09/17/2024] [Indexed: 12/20/2024] Open
Abstract
Sickle cell disease (SCD) is a monogenic disease, resulting from a single-point mutation, that presents a complex pathophysiology and high clinical heterogeneity. Inflammation stands as a prominent characteristic of SCD. Over the past few decades, the role of different cells and molecules in the regulation of the inflammatory process has been elucidated. In conjunction with the polymerization of hemoglobin S (HbS), intravascular hemolysis, which releases free heme, HbS, and hemoglobin-related damage-associated molecular patterns, initiates multiple inflammatory pathways that are not yet fully comprehended. These complex phenomena lead to a vicious cycle that perpetuates vaso-occlusion, hemolysis, and inflammation. To date, few inflammatory biomarkers can predict disease complications; conversely, there is a plethora of therapies that reduce inflammation in SCD, although clinical outcomes vary widely. Importantly, whether the clinical heterogeneity and complications are related to the degree of inflammation is not known. This review aims to further our understanding of the roles of main immune cells, and other inflammatory factors, as potential prognostic biomarkers for predicting clinical outcomes or identifying novel treatments for SCD.
Collapse
Affiliation(s)
- Karina Tozatto‐Maio
- Centro de Ensino e PesquisaHospital Israelita Albert EinsteinSão PauloBrazil
- Divisão de Hematologia, Hemoterapia e Terapia CelularHospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão PauloBrazil
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco‐Immuno‐Hematology (LIM‐31), Department of Hematology and Cell TherapyHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
| | - Felipe A. Rós
- Divisão de Hematologia, Hemoterapia e Terapia CelularHospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão PauloBrazil
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco‐Immuno‐Hematology (LIM‐31), Department of Hematology and Cell TherapyHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
| | - Ricardo Weinlich
- Centro de Ensino e PesquisaHospital Israelita Albert EinsteinSão PauloBrazil
| | - Vanderson Rocha
- Divisão de Hematologia, Hemoterapia e Terapia CelularHospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão PauloBrazil
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco‐Immuno‐Hematology (LIM‐31), Department of Hematology and Cell TherapyHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
- Instituto D'Or de Ensino e Pesquisa, Rede D'OrSao PauloBrazil
- Department of Hematology, Churchill HospitalUniversity of OxfordOxfordUK
| |
Collapse
|
4
|
Shahin WA, Aldeeb H, Alsulami M, Tammas A, Albatniji F, Almanea A, Zayed AM, Alabbas F, Alzahrani A, Bin Ali T, Elyamany G, Almaghrabi RH, Elfaraidi H. Children with sickle cell disease: are they protected from serious COVID-19? Front Pediatr 2024; 12:1337377. [PMID: 39435386 PMCID: PMC11491405 DOI: 10.3389/fped.2024.1337377] [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: 01/11/2024] [Accepted: 09/05/2024] [Indexed: 10/23/2024] Open
Abstract
Background COVID-19, the pandemic that hit the world in 2020, resulted in millions of deaths, with the elderly and adults succumbing to the disease more often than children. However, the presence of underlying morbidities increased the risk of death. Sickle cell disease (SCD) was previously classified as a major risk factor for severe COVID-19 disease. However, presently, there are only a limited number of studies that identify the clinical course of children with SCD and COVID-19. Methods We conducted a retrospective observational study on children with SCD admitted due to COVID-19 at three different institutions in Saudi Arabia between March 2020 and March 2022. We studied the demographic and clinical characteristics of patients admitted to the hospital. Results Seventy-six patients with SCD had PCR-confirmed SARS-CoV-2 during the study period; 50.0% of our patient population were children (6-12 years old). Gender was evenly distributed, with 53.9% girls and 46.1% boys. Symptoms more commonly related to the COVID-19 infection included fever, cough, malaise, and vomiting. Chest x-ray findings revealed mild and non-specific symptoms only in approximately one-third (28) of the included children. The most common symptoms associated with SCD were vaso-occlusive crisis (47.4%) and abdominal pain (11.8%). The overall general appearance of most of the patients was reassuring. The median length of hospital stay was 4.2 ± 2.7 days. The mean white blood cell count was 11.4 ± 5.2 × 109/L, and the mean hemoglobin level was 8.3 ± 1.5 g/dl. Despite the fact that higher levels of mean D-dimer, lactate dehydrogenase, and ferritin were reported in these patients, the clinical outcome was not affected. All recruited patients received hydroxyurea as maintenance therapy. The outcome of our study was reassuring, with no significant morbidity or mortality observed among the recruited patients. Conclusion Despite SCD being a chronic disease with known specific complications, there has been a claim that COVID-19 infection adds further risk. The results of this study suggest that the overall outcome of COVID-19 was favorable, with no reported mortality. Further research is needed to understand the factors that contributed to this favorable outcome. In children with SCD, it is still questionable whether hydroxyurea is one of the protective factors against severe COVID-19. Validation through large-scale research is recommended.
Collapse
Affiliation(s)
- Walaa Aboulkasem Shahin
- Department of Pediatrics, Specialized Children's Hospital, Cairo University, Cairo, Egypt
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Hayam Aldeeb
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Majed Alsulami
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Abdullah Tammas
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Fatma Albatniji
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Aljawhara Almanea
- Hematology Division, Department of Pediatrics, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | | | - Fahad Alabbas
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Azzah Alzahrani
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Tahani Bin Ali
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Ghaleb Elyamany
- Department of Central Military Laboratory and Blood Bank, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | | | - Huda Elfaraidi
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Lewis J, Guilcher GMT, Greenway SC. Reviewing the impact of hydroxyurea on DNA methylation and its potential clinical implications in sickle cell disease. Eur J Haematol 2024; 113:264-272. [PMID: 38831675 DOI: 10.1111/ejh.14247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 06/05/2024]
Abstract
Hydroxyurea (HU) is the most common drug therapy for sickle cell disease (SCD). The clinical benefits of HU derive from its upregulation of fetal hemoglobin (HbF), which reduces aggregation of the mutated sickle hemoglobin protein (HbS) and reduces SCD symptoms and complications. However, some individuals do not respond to HU, or stop responding over time. Unfortunately, current understanding of the mechanism of action of HU is limited, hindering the ability of clinicians to identify those patients who will respond to HU and to optimize treatment for those receiving HU. Given that epigenetic modifications are essential to erythropoiesis and HbF expression, we hypothesize that some effects of HU may be mediated by epigenetic modifications, specifically DNA methylation. However, few studies have investigated this possibility and the effects of HU on DNA methylation remain relatively understudied. In this review, we discuss the evidence linking HU treatment to DNA methylation changes and associated gene expression changes, with an emphasis on studies that were performed in individuals with SCD. Overall, although HU can affect DNA methylation, research on these changes and their clinical effects remains limited. Further study is likely to contribute to our understanding of hematopoiesis and benefit patients suffering from SCD.
Collapse
Affiliation(s)
- Jasmine Lewis
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gregory M T Guilcher
- Department of Pediatrics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Steven C Greenway
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
6
|
Lizarralde-Iragorri MA, Parachalil Gopalan B, Merriweather B, Brooks J, Hill M, Lovins D, Pierre-Charles R, Cullinane A, Dulau-Florea A, Lee DY, Villasmil R, Jeffries N, Shet AS. Isoquercetin for thromboinflammation in sickle cell disease: a randomized double-blind placebo-controlled trial. Blood Adv 2024; 8:172-182. [PMID: 38157227 PMCID: PMC10787266 DOI: 10.1182/bloodadvances.2023011542] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/07/2023] [Indexed: 01/03/2024] Open
Abstract
ABSTRACT Data from a small trial in patients with cancer suggest that isoquercetin (IQ) treatment lowered thrombosis biomarkers and prevented clinical thrombosis, but, to our knowledge, no studies of IQ have been conducted to target thromboinflammation in adults with sickle cell disease (SCD). We conducted a randomized, double-blind, placebo-controlled trial in adults with steady-state SCD (hemoglobin SS [HbSS], HbSβ0thal, HbSβ+thal, or HbSC). The primary outcome was the change in plasma soluble P-selectin (sP-selectin) after treatment compared with baseline, analyzed in the intention-to-treat population. Between November 2019 and July 2022, 46 patients (aged 40 ± 11 years, 56% female, 75% under hydroxyurea treatment) were randomized to receive IQ (n = 23) or placebo (n = 23). IQ was well tolerated and all the adverse events (AEs; n = 21) or serious AEs (n = 14) recorded were not attributable to the study drug. The mean posttreatment change for sP-selectin showed no significant difference between the treatment groups (IQ, 0.10 ± 6.53 vs placebo, 0.74 ± 4.54; P = .64). In patients treated with IQ, whole-blood coagulation (P = .03) and collagen-induced platelet aggregation (P = .03) were significantly reduced from the baseline. Inducible mononuclear cell tissue factor gene expression and plasma protein disulfide isomerase reductase activity were also significantly inhibited (P = .003 and P = .02, respectively). Short-term fixed-dose IQ in patients with SCD was safe with no off-target bleeding and was associated with changes from the baseline in the appropriate direction for several biomarkers of thromboinflammation. The trial was registered at www.clinicaltrials.gov as #NCT04514510.
Collapse
Affiliation(s)
- Maria A Lizarralde-Iragorri
- Sickle Thrombosis and Vascular Biology Lab, Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Bindu Parachalil Gopalan
- Sickle Thrombosis and Vascular Biology Lab, Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Brenda Merriweather
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Jennifer Brooks
- Office of the Clinical Director, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Mai Hill
- Office of the Clinical Director, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Dianna Lovins
- Office of the Clinical Director, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Ruth Pierre-Charles
- Office of the Clinical Director, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Ann Cullinane
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Alina Dulau-Florea
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Duck-Yeon Lee
- Biochemistry Core Facility, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Rafael Villasmil
- Flow Cytometry Core Facility, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Neal Jeffries
- Office of Biostatistics Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Arun S Shet
- Sickle Thrombosis and Vascular Biology Lab, Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
7
|
Liu Y, Su S, Shayo S, Bao W, Pal M, Dou K, Shi PA, Aygun B, Campbell-Lee S, Lobo CA, Mendelson A, An X, Manwani D, Zhong H, Yazdanbakhsh K. Hemolysis dictates monocyte differentiation via two distinct pathways in sickle cell disease vaso-occlusion. J Clin Invest 2023; 133:e172087. [PMID: 37490346 PMCID: PMC10503794 DOI: 10.1172/jci172087] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023] Open
Abstract
Sickle cell disease (SCD) is a hereditary hemoglobinopathy characterized by painful vaso-occlusive crises (VOC) and chronic hemolysis. The mononuclear phagocyte system is pivotal to SCD pathophysiology, but the mechanisms governing monocyte/macrophage differentiation remain unknown. This study examined the influence of hemolysis on circulating monocyte trajectories in SCD. We discovered that hemolysis stimulated CSF-1 production, partly by endothelial cells via Nrf2, promoting classical monocyte (CMo) differentiation into blood patrolling monocytes (PMo) in SCD mice. However, hemolysis also upregulated CCL-2 through IFN-I, inducing CMo transmigration and differentiation into tissue monocyte-derived macrophages. Blocking CMo transmigration by anti-P selectin antibody in SCD mice increased circulating PMo, corroborating that CMo-to-tissue macrophage differentiation occurs at the expense of CMo-to-blood PMo differentiation. We observed a positive correlation between plasma CSF-1/CCL-2 ratios and blood PMo levels in patients with SCD, underscoring the clinical significance of these two opposing factors in monocyte differentiation. Combined treatment with CSF-1 and anti-P selectin antibody more effectively increased PMo numbers and reduced stasis compared with single-agent therapies in SCD mice. Altogether, these data indicate that monocyte fates are regulated by the balance between two heme pathways, Nrf2/CSF-1 and IFN-I/CCL-2, and suggest that the CSF-1/CCL-2 ratio may present a diagnostic and therapeutic target in SCD.
Collapse
Affiliation(s)
| | - Shan Su
- Laboratory of Complement Biology
| | | | | | | | - Kai Dou
- Laboratory of Immune Regulation, and
| | - Patricia A. Shi
- Clinical Research in Sickle Cell Disease, New York Blood Center, New York, New York, USA
| | - Banu Aygun
- Cohen Children’s Medical Center, New Hyde Park, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Sally Campbell-Lee
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | | | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, New York, USA
| | - Deepa Manwani
- Department of Pediatrics, Montefiore Medical Center, Albert Einstein College of Medicine, Children’s Hospital at Montefiore, New York, New York, USA
| | - Hui Zhong
- Laboratory of Immune Regulation, and
| | | |
Collapse
|
8
|
Zheng Y, Gossett JM, Chen PL, Barton M, Ryan M, Yu J, Kang G, Hankins JS, Chou ST. Proinflammatory state promotes red blood cell alloimmunization in pediatric patients with sickle cell disease. Blood Adv 2023; 7:4799-4808. [PMID: 37023228 PMCID: PMC10469551 DOI: 10.1182/bloodadvances.2022008647] [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] [Received: 07/29/2022] [Revised: 02/09/2023] [Accepted: 02/27/2023] [Indexed: 04/08/2023] Open
Abstract
We examined risk factors for red blood cell (RBC) alloimmunization in pediatric patients with sickle cell disease, focusing on the recipients' inflammatory state at the time of transfusion and anti-inflammatory role of hydroxyurea (HU). Among 471 participants, 55 (11.70%) participants were alloimmunized and formed 59 alloantibodies and 17 autoantibodies with an alloimmunization rate of 0.36 alloantibodies per 100 units. Analysis of 27 participants in whom alloantibodies were formed with specificities showed 23.8% (30/126) of units transfused during a proinflammatory event resulting in alloantibody formation compared with 2.8% (27/952) of units transfused at steady state. Therefore, transfusion during proinflammatory events increased the risk for alloimmunization (odds ratio [OR], 4.22; 95% confidence interval [CI], 1.64-10.85; P = .003). Further analysis of all the 471 participants showed that alloimmunization of patients who received episodic transfusion, mostly during proinflammatory events, was not reduced with HU therapy (OR, 6.52; 95% CI, 0.85-49.77; P = .071), HU therapy duration (OR, 1.13; 95% CI, 0.997-1.28; P = .056), or HU dose (OR, 1.06; 95% CI, 0.96-1.16; P = .242). The analysis also identified high transfusion burden (OR, 1.02; 95% CI, 1.003-1.04; P = .020) and hemoglobin S (HbSS) and HbSβ0-thalassemia genotypes (OR, 11.22, 95% CI, 1.51-83.38; P = .018) as additional risk factors for alloimmunization. In conclusion, the inflammatory state of transfusion recipients affects the risk of RBC alloimmunization, which is not modified by HU therapy. Judicious use of transfusion during proinflammatory events is critical for preventing alloimmunization.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jeffrey M. Gossett
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Pei-Lin Chen
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Martha Barton
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Missy Ryan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jing Yu
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Guolian Kang
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jane S. Hankins
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Stella T. Chou
- Departments of Pediatrics and Pathology, The Children’s Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA
| |
Collapse
|
9
|
Hydroxyurea as a promising ADAM17 inhibitor. Med Hypotheses 2023. [DOI: 10.1016/j.mehy.2023.111021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
10
|
In-Depth Immunological Typization of Children with Sickle Cell Disease: A Preliminary Insight into Its Plausible Correlation with Clinical Course and Hydroxyurea Therapy. J Clin Med 2022; 11:jcm11113037. [PMID: 35683425 PMCID: PMC9181704 DOI: 10.3390/jcm11113037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/22/2022] [Accepted: 05/26/2022] [Indexed: 02/04/2023] Open
Abstract
Sickle cell disease (SCD) is a condition of functional hypo-/a-splenism in which predisposition to bacterial infections is only a facet of a wide spectrum of immune-dysregulation disorders forming the clinical expression of a peculiar immunophenotype. The objective of this study was to perform an in-depth immunophenotypical characterization of SCD pediatric patients, looking for plausible correlations between immunological biomarkers, the impact of hydroxyurea (HU) treatment and clinical course. This was an observational case−control study including 43 patients. The cohort was divided into two main groups, SCD subjects (19/43) and controls (24/43), differing in the presence/absence of an SCD diagnosis. The SCD group was split up into HU+ (12/19) and HU− (7/19) subgroups, respectively receiving or not a concomitant HU treatment. The principal outcomes measured were differences in the immunophenotyping between SCD patients and controls through chi-squared tests, t-tests, and Pearson’s correlation analysis between clinical and immunological parameters. Leukocyte and neutrophil increase, T-cell depletion with prevalence of memory T-cell compartment, NK and B-naïve subset elevation with memory and CD21low B subset reduction, and IgG expansion, significantly distinguished the SCD HU− subgroup from controls, with naïve T cells, switched-memory B cells and IgG maintaining differences between the SCD HU+ group and controls (p-value of <0.05). The mean CD4+ central-memory T-cell% count was the single independent variable showing a positive correlation with vaso-occlusive crisis score in the SCD group (Pearson’s R = 0.039). We report preliminary data assessing plausible clinical implications of baseline and HU-related SCD immunophenotypical alterations, which need to be validated in larger samples, but potentially affecting hypo-/a-splenism immuno-chemoprophylactic recommendations.
Collapse
|
11
|
Alagbe AE, Domingos IF, Adekile AD, Blotta MHSL, Santos MNN. Anti-inflammatory cytokines in sickle cell disease. Mol Biol Rep 2022; 49:2433-2442. [PMID: 35000064 DOI: 10.1007/s11033-021-07009-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022]
Abstract
Sickle cell disease (SCD) is a well-studied monogenetic disease with an established chronic inflammatory component. The paradigm shift towards inflammation has made the pathophysiology of SCD even more complex. Studies have shown that an imbalance between the pro-inflammatory and anti-inflammatory cytokines in SCD exists; however, the reports are skewed toward the pro-inflammatory mediators. We enumerate recent in vitro and in vivo studies on anti-inflammatory cytokines in SCD patients, and discuss the biology of anti-inflammatory cytokines including the already reported IL-2, TGF-β, and IL-10 as well as the recently discovered IL-27, IL-35 and IL-37. This review will improve the understanding of the pathophysiology of SCD and aid in the search of new therapeutic options for patients with SCD.
Collapse
Affiliation(s)
- Adekunle E Alagbe
- Department of Clinical Pathology, School of Medical Sciences, State University of Campinas - UNICAMP, Campinas, São Paulo, Brazil
| | - Igor F Domingos
- Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Adekunle D Adekile
- Department of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Maria H S L Blotta
- Department of Clinical Pathology, School of Medical Sciences, State University of Campinas - UNICAMP, Campinas, São Paulo, Brazil
| | - Magnun N N Santos
- Department of Clinical Pathology, School of Medical Sciences, State University of Campinas - UNICAMP, Campinas, São Paulo, Brazil.
| |
Collapse
|
12
|
Liisborg C, Skov V, Kjær L, Hasselbalch HC, Sørensen TL. Patients with MPNs and retinal drusen show signs of complement system dysregulation and a high degree of chronic low-grade inflammation. EClinicalMedicine 2022; 43:101248. [PMID: 35128362 PMCID: PMC8808164 DOI: 10.1016/j.eclinm.2021.101248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/16/2021] [Accepted: 12/08/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND The hematopoietic stem cell disorders, myeloproliferative neoplasms (MPNs), are characterised by chronic low-grade inflammation (CLI). Recently, we showed that patients with MPNs have an increased prevalence of drusen and age-related macular degeneration (AMD), and drusen prevalence seemed associated with higher CLI. Studying MPNs may reveal more about drusen pathophysiology. This study investigated CLI further by measuring cytokine levels and complement system markers, comparing these between patients with MPNs and AMD. METHODS This cross-sectional study, between July 2018 and November 2020 conducted at Zealand University Hospital (ZUH) - Roskilde, Denmark, included 29 patients with neovascular AMD (nAMD), 28 with intermediate-stage AMD (iAMD), 62 with MPNs (35 with drusen - MPNd and 27 with healthy retinas - MPNn). With flow cytometry, we measured complement-regulatory-proteins (Cregs). With immunoassays, we investigated cytokine levels combined into a summary-inflammation-score (SIS). FINDINGS The MPNd and nAMD groups had similar SIS, significantly higher than the MPNn and iAMD groups. Additionally, we found SIS to increase over the MPN biological continuum from early cancer stage, essential thrombocytaemia (ET), over polycythaemia vera (PV) to the late-stage primary myelofibrosis (PMF). MPNs showed signs of complement dysregulation, with Cregs expression lower in PV than ET and PMF and even lower in PV patients with drusen. INTERPRETATION This study suggests that MPNd have a higher CLI than MPNn and may indicate systemic CLI to play a greater part in, and even initiate drusen formation. We suggest using MPNs as a "Human Inflammation Model" of drusen development. The CLI in MPNs elicits drusen formation, triggering more CLI creating a vicious cycle, increasing the risk of developing AMD. FUNDING Fight for Sight, Denmark, and Region Zealand's research promotion fund.
Collapse
Affiliation(s)
- Charlotte Liisborg
- Department of Ophthalmology, Zealand University Hospital, Vestermarksvej 23, Roskilde DK-4000, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
- Corresponding author.
| | - Vibe Skov
- Department of Haematology, Zealand University Hospital, Vestermarksvej 15-17, Roskilde 4000, Denmark
| | - Lasse Kjær
- Department of Haematology, Zealand University Hospital, Vestermarksvej 15-17, Roskilde 4000, Denmark
| | - Hans Carl Hasselbalch
- Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
- Department of Haematology, Zealand University Hospital, Vestermarksvej 15-17, Roskilde 4000, Denmark
| | - Torben Lykke Sørensen
- Department of Ophthalmology, Zealand University Hospital, Vestermarksvej 23, Roskilde DK-4000, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| |
Collapse
|
13
|
Ben Moftah M, Eswayah A. Repurposing of Hydroxyurea Against COVID-19: A Promising Immunomodulatory Role. Assay Drug Dev Technol 2022; 20:55-62. [PMID: 34990284 DOI: 10.1089/adt.2021.090] [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] [Indexed: 12/27/2022] Open
Abstract
Cytokine release syndrome, a prominent mechanism of morbidity and mortality in patients with coronavirus disease 2019 (COVID-19), can cause multiple bodily reactions, including excessive release of proinflammatory mediators, with tumor necrosis factor-α (TNF-α) being the most prevalent cytokine combined with persistently elevated D-dimer levels that are indicative of potential thrombotic events, low levels of endogenous nitric oxide (NO) generation, and progressive decrease in hemoglobin production. In our argument, the conceptual repurposing of hydroxyurea (HU) for managing COVID-19 can provide a promising therapeutic option originating from a rich history of investigational antiviral activity. HU as a proposed supportive therapeutic agent for treating COVID-19 can exemplify a successful remedial choice through its anti-inflammatory activity along with an intrinsic propensity to control the circulatory levels of key cytokines including TNF-α. HU has the ability to undergo in vivo NO conversion acting as NO donor together with being a prominent inducer of fetal hemoglobin (HbF) production. The combination of the mentioned two properties allows HU to possess evident capability of protecting against thrombotic events by controlling D-dimer levels. The implication of our hypothetical argument sheds light on the curative potential of HU, which can be strategically harnessed against COVID-19.
Collapse
Affiliation(s)
- Moayed Ben Moftah
- Department of Medicinal and Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tripoli, Tripoli, Libya
| | - Asma Eswayah
- Department of Medicinal and Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tripoli, Tripoli, Libya
| |
Collapse
|
14
|
Muniz Carvalho E, Silva Sousa EH, Bernardes‐Génisson V, Gonzaga de França Lopes L. When NO
.
Is not Enough: Chemical Systems, Advances and Challenges in the Development of NO
.
and HNO Donors for Old and Current Medical Issues. Eur J Inorg Chem 2021. [DOI: 10.1002/ejic.202100527] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Edinilton Muniz Carvalho
- Bioinorganic Group Department of Organic and Inorganic Chemistry Center of Sciences Federal University of Ceará Pici Campus Fortaleza 60455-760 Brazil
- CNRS Laboratoire de Chimie de Coordination LCC UPR 8241 205 Route de Narbonne, 44099 31077 Toulouse, Cedex 4 France
- Université de Toulouse Université Paul Sabatier UPS 118 Route de Narbonne 31062 Toulouse, Cedex 9 France
| | - Eduardo Henrique Silva Sousa
- Bioinorganic Group Department of Organic and Inorganic Chemistry Center of Sciences Federal University of Ceará Pici Campus Fortaleza 60455-760 Brazil
| | - Vania Bernardes‐Génisson
- CNRS Laboratoire de Chimie de Coordination LCC UPR 8241 205 Route de Narbonne, 44099 31077 Toulouse, Cedex 4 France
- Université de Toulouse Université Paul Sabatier UPS 118 Route de Narbonne 31062 Toulouse, Cedex 9 France
| | - Luiz Gonzaga de França Lopes
- Bioinorganic Group Department of Organic and Inorganic Chemistry Center of Sciences Federal University of Ceará Pici Campus Fortaleza 60455-760 Brazil
| |
Collapse
|
15
|
de Freitas Dutra V, Leal VNC, Fernandes FP, Souza CRL, Figueiredo MS, Pontillo A. Genetic contribution and functional impairment of inflammasome in sickle cell disease. Cytokine 2021; 149:155717. [PMID: 34627079 DOI: 10.1016/j.cyto.2021.155717] [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: 11/28/2020] [Revised: 04/16/2021] [Accepted: 09/20/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Sickle cell disease (SCD), one of the most common single-gene disorders, is caused by mutations in the hemoglobin ß-chain gene. Clinical presentation is heterogeneous, and inflammation is a common condition. Thereby, we hypothesized that inflammasome and related cytokine IL-1ß could represent significant SCD pathogenesis contributors. MATERIAL AND METHODS 161 SCD (SS/Sβ) patients were enrolled for the study. Seven single nucleotide polymorphisms (SNPs) in 5 inflammasome genes (NLRP1, NLRP3, NLRC4, CARD8, IL1B) were selected based on minor allele frequency. Total peripheral blood mononuclear cells (PBMC) and monocytes were isolated from 10 out of 161 SCD patients (HbSS) and 10 healthy donors (control group, Ctrl) for inflammasome analysis. RESULTS SCD patients presented a functional impairment of inflammasome, with monocytes and peripheral blood mononuclear cells (PBMC) exhibiting a different NLRP3 inflammasome activation rate. Gain-of-function variants in NLRP1 and IL1B genes resulted associated with a mild SCD clinical presentation. DISCUSSION Our results can contribute to the understanding of SCD inflammation. SCD patients showed possible exhaustion of monocytes due to chronic inflammation, moreover others cells in PBMC can contribute to the NLRP3 inflammasome activation. NLRP1 gain-of-function was associated with mild clinical presentation, suggesting that other inflammasome receptors can be involved in SCD. This is the first study reporting a significant contribution of inflammasome SNPs in SCD.
Collapse
Affiliation(s)
- Valéria de Freitas Dutra
- Hematology and Blood Transfusion Division, Clinical and Experimetnal Oncology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/UNIFESP), R Dr. Diogo de Farias, 824, 04037-002 Vila Clementino, São Paulo, SP, Brazil.
| | - Vinícius Nunes Cordeiro Leal
- Laboratory of Immunogenetics, Department of Immunology, Institute of Biomedical Sciences/ICB, University of São Paulo/USP, Av. Prof. Lineu Prestes, 1730, 05508-000 Butantã, São Paulo, SP, Brazil.
| | - Fernanda Pereira Fernandes
- Laboratory of Immunogenetics, Department of Immunology, Institute of Biomedical Sciences/ICB, University of São Paulo/USP, Av. Prof. Lineu Prestes, 1730, 05508-000 Butantã, São Paulo, SP, Brazil.
| | - Cláudia Regina Lustosa Souza
- Hematology and Blood Transfusion Division, Clinical and Experimetnal Oncology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/UNIFESP), R Dr. Diogo de Farias, 824, 04037-002 Vila Clementino, São Paulo, SP, Brazil
| | - Maria Stella Figueiredo
- Hematology and Blood Transfusion Division, Clinical and Experimetnal Oncology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/UNIFESP), R Dr. Diogo de Farias, 824, 04037-002 Vila Clementino, São Paulo, SP, Brazil.
| | - Alessandra Pontillo
- Laboratory of Immunogenetics, Department of Immunology, Institute of Biomedical Sciences/ICB, University of São Paulo/USP, Av. Prof. Lineu Prestes, 1730, 05508-000 Butantã, São Paulo, SP, Brazil
| |
Collapse
|
16
|
Philadelphia-Negative Chronic Myeloproliferative Neoplasms during the COVID-19 Pandemic: Challenges and Future Scenarios. Cancers (Basel) 2021; 13:cancers13194750. [PMID: 34638236 PMCID: PMC8507529 DOI: 10.3390/cancers13194750] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/09/2021] [Accepted: 09/17/2021] [Indexed: 12/30/2022] Open
Abstract
An outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) started in December 2019 in China and then become pandemic in February 2020. Several publications investigated the possible increased rate of COVID-19 infection in hematological malignancies. Based on the published data, strategies for the management of chronic Philadelphia-negative chronic myeloproliferative neoplasms (MPNs) are provided. The risk of severe COVID-19 seems high in MPN, particularly in patients with essential thrombocythemia, but not negligible in myelofibrosis. MPN patients are at high risk of both thrombotic and hemorrhagic complications and this must be accounted in the case of COVID-19 deciding on a case-by-case basis. There are currently no data to suggest that hydroxyurea or interferon may influence the risk or severity of COVID-19 infection. Conversely, while the immunosuppressive activity of ruxolitinib might pose increased risk of infection, its abrupt discontinuation during COVID-19 syndrome is associated with worse outcome. All MPN patients should receive vaccine against COVID-19; reassuring data are available on efficacy of mRNA vaccines in MPNs.
Collapse
|
17
|
Reeves BN, Moliterno AR. Thrombosis in myeloproliferative neoplasms: update in pathophysiology. Curr Opin Hematol 2021; 28:285-291. [PMID: 34183535 DOI: 10.1097/moh.0000000000000664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW This review summarizes high-impact research in myeloproliferative neoplasms (MPN) from the last 18 months, with a particular focus on basic science findings. RECENT FINDINGS A pseudo-hypoxia state with stabilization of hypoxia-inducible factor (HIFα exists that is central to cell growth, cell renewal, inflammation, and thrombotic potential in MPN hematopoietic cells. SUMMARY HIFα and inflammatory pathways are new therapeutic targets in MPN, with the potential to ameliorate thrombotic risk and perhaps eradicate mutant progenitor cells.
Collapse
Affiliation(s)
- Brandi N Reeves
- Hematology Division, Department of Medicine, Blood Research Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alison R Moliterno
- Hematology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Kashari O, Alghamdi B, Al-Hebshi A, Asiri A, Fallatah E, Alshehri F, Alsamiri S, Masmali H, Nabulsi M, Assiri M, Alwasaidi TA. COVID-19 in Saudi Patients With Sickle Cell Disease: A Retrospective Multi-Center Study. Cureus 2021; 13:e17238. [PMID: 34422504 PMCID: PMC8369254 DOI: 10.7759/cureus.17238] [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] [Accepted: 08/16/2021] [Indexed: 12/15/2022] Open
Abstract
Background The prevalence of sickle cell disease (SCD) within Saudi Arabia is relatively high, with an estimated 145/10,000 cases. There is an urgent need for researching many aspects of the Coronavirus disease of 2019 (COVID-19) due to the widespread of the virus among SCD patients in Saudi Arabia. The aim of this study is to determine how COVID-19 affects SCD patients in order to reach the best strategy for their management protocols. Methods This is a retrospective chart review study from a multi-center in Saudi Arabia that evaluated a total of 33 patients with sickle cell anemia/disease who were confirmed to have COVID-19. The diagnosis of COVID-19 was confirmed by using the reverse transcription-polymerase chain reaction (RT-PCR) tests based on the nasopharyngeal swabs of the included patients. Results The mean age of patients was 10.75+9.11 years, and nearly all patients (n= 32; 96.9%) were Saudi, and 48.4% of them were females. Twenty-two patients were admitted (59.5%); the main reasons for admission included vaso-occlusive crisis (VOC) only (n= 6; 27.3%), fever (n= 6; 27.3%), acute chest syndrome (n= 5; 22.7%), and VOC combined with other conditions (n= 4; 18.2%). During hospitalization, 54.1% of the patients received at least one medication, while antibiotics (54.1%), analgesia (32.4%), anticoagulants (16.2%), and steroids (16.2%) were the most commonly administered drugs. The mean length of hospitalization was 7.6±4.5 days, with only one patient (2.7%) requiring intensive care unit admission and assisted ventilation. Conclusion The overall prognosis was good since only one patient has passed away, while all others recovered and, subsequently, were discharged. Manifestations, laboratory investigations, and management modalities should be utilized promptly to enhance the prognosis and obtain better outcomes.
Collapse
Affiliation(s)
- Ohoud Kashari
- Department of Pediatrics, East Jeddah General Hospital, Jeddah, SAU
| | - Badriah Alghamdi
- Department of Pediatrics, East Jeddah General Hospital, Jeddah, SAU
| | - Abdulqader Al-Hebshi
- Division of Hematology, Department of Pediatrics, Prince Mohammed bin AbdulAziz Hospital, Ministry of National Guard - Health Affairs, Medina, SAU.,Department of Pediatrics, King Abdullah International Medical Research Center (KAIMRC), Riyadh, SAU.,Faculty of Medicine, Department of Pediatrics, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, SAU
| | - Aljawharah Asiri
- Department of Pediatrics, East Jeddah General Hospital, Jeddah, SAU
| | - Ebtehal Fallatah
- Department of Pediatrics, East Jeddah General Hospital, Jeddah, SAU
| | - Fayez Alshehri
- Department of Pediatrics, Maternity and Children Hospital, Khamis Mushayt, SAU
| | - Salihah Alsamiri
- Medicine, Royal College of Surgeons, University of Medicine and Health Sciences, Dublin, IRL
| | - Hassan Masmali
- Department of Pediatrics, Maternity and Children Hospital, Mecca, SAU
| | - Mohammad Nabulsi
- Department of Medicine, King Abdullah Medical Complex, Jeddah, SAU
| | - Mona Assiri
- Department of Pediatrics, Maternity and Children Hospital, Abha, SAU
| | - Turki A Alwasaidi
- Division of Hematology, Department of Medicine, Prince Mohammed bin Abdulaziz Hospital, Ministry of National Guard - Health Affairs, Madinah, SAU.,Department of Medicine, King Abdullah International Medical Research Centers (KAIMRC), Medina, SAU.,Department of Medicine, College of Medicine, Taibah University, Medinah, SAU.,Faculty of Medicine, Department of Pediatrics, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Jeddah, SAU
| |
Collapse
|
19
|
Sayad B, Karimi M, Rahimi Z. Sickle cell disease and COVID-19: Susceptibility and severity. Pediatr Blood Cancer 2021; 68:e29075. [PMID: 34061431 PMCID: PMC8209850 DOI: 10.1002/pbc.29075] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/25/2022]
Abstract
We surveyed published papers and an international sickle cell disease (SCD) registry to detect susceptibility and clinical course of coronavirus disease 2019 (COVID-19) in SCD patients. COVID-19 presentation was mild in children and moderate in many SCD adults. Regarding increased comorbidities with age, it seems severe COVID-19 to be more common in older SCD patients. Although the overall outcome of COVID-19 was favorable in SCD children, a high rate of pediatric intensive care unit admission should be considered in managing these patients. To explain COVID-19 outcome in SCD patients, the possible benefits of hydroxyurea therapy could be considered. The obtained results should be interpreted, considering low cases from sub-Saharan people, younger age of SCD patients compared to general population, a bias toward registry of the more severe form of disease, the effect of pre-existing comorbidities with multisystem organ damage, and the role of health socio-economic determinants.
Collapse
Affiliation(s)
- Babak Sayad
- Infectious Diseases Research CenterKermanshah University of Medical SciencesKermanshahIran
| | - Mehran Karimi
- Hematology Research CenterShiraz University of Medical SciencesShirazIran
| | - Zohreh Rahimi
- Department of Clinical Biochemistry, Medical SchoolKermanshah University of Medical SciencesKermanshahIran
- Medical Biology Research CenterKermanshah University of Medical SciencesKermanshahIran
| |
Collapse
|
20
|
Hasselbalch HC, Skov V, Kjær L, Ellervik C, Poulsen A, Poulsen TD, Nielsen CH. COVID-19 as a mediator of interferon deficiency and hyperinflammation: Rationale for the use of JAK1/2 inhibitors in combination with interferon. Cytokine Growth Factor Rev 2021; 60:28-45. [PMID: 33992887 PMCID: PMC8045432 DOI: 10.1016/j.cytogfr.2021.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 02/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) elicits an interferon (IFN) deficiency state, which aggravates the type I interferon deficiency and slow IFN responses, which associate with e.g. aging and obesity. Additionally, SARS-CoV-2 may also elicit a cytokine storm, which accounts for disease progression and ultimately the urgent need of ventilator support. Based upon several reports, it has been argued that early treatment with IFN-alpha2 or IFN-beta, preferentially in the early disease stage, may prohibit disease progression. Similarly, preliminary studies have shown that JAK1/2 inhibitor treatment with ruxolitinib or baricitinib may decrease mortality by dampening the deadly cytokine storm, which - in addition to the virus itself - also contributes to multi-organ thrombosis and multi-organ failure. Herein, we describe the rationale for treatment with IFNs (alpha2 or beta) and ruxolitinib emphasizing the urgent need to explore these agents in the treatment of SARS-CoV-2 - both as monotherapies and in combination. In this context, we take advantage of several safety and efficacy studies in patients with the chronic myeloproliferative blood cancers (essential thrombocythemia, polycythemia vera and myelofibrosis) (MPNs), in whom IFN-alpha2 and ruxolitinib have been used successfully for the last 10 (ruxolitinib) to 30 years (IFN) as monotherapies and most recently in combination as well. In the context of these agents being highly immunomodulating (IFN boosting immune cells and JAK1/2 inhibitors being highly immunosuppressive and anti-inflammatory), we also discuss if statins and hydroxyurea, both agents possessing anti-inflammatory, antithrombotic and antiviral potentials, might be inexpensive agents to be repurposed in the treatment of SARS-CoV-2.
Collapse
Affiliation(s)
- H C Hasselbalch
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark.
| | - V Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - L Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - C Ellervik
- Department of Research, Production, Innovation, Region Zealand, Denmark; Department of Pathology, Harvard Medical School, Boston, MA, United States; Department of Laboratory Medicine, Boston Children's Hospital, Boston, MA, United States
| | - A Poulsen
- Department of Anestesiology and Intensive Care Unit, Zealand University Hospital, Roskilde, Denmark
| | - T D Poulsen
- Department of Anestesiology and Intensive Care Unit, Zealand University Hospital, Roskilde, Denmark
| | - C H Nielsen
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
21
|
Cominal JG, Cacemiro MDC, Berzoti-Coelho MG, Pereira IEG, Frantz FG, Souto EX, Covas DT, de Figueiredo-Pontes LL, Oliveira MC, Malmegrim KCR, de Castro FA. Bone Marrow Soluble Mediator Signatures of Patients With Philadelphia Chromosome-Negative Myeloproliferative Neoplasms. Front Oncol 2021; 11:665037. [PMID: 34084749 PMCID: PMC8167065 DOI: 10.3389/fonc.2021.665037] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/21/2021] [Indexed: 12/18/2022] Open
Abstract
Background Essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis (PMF) are clonal hematological diseases classified as Philadelphia chromosome-negative myeloproliferative neoplasms (MPN). MPN pathogenesis is associated with the presence of somatic driver mutations, bone marrow (BM) niche alterations, and tumor inflammatory status. The relevance of soluble mediators in the pathogenesis of MPN led us to analyze the levels of cytokines, chemokines, and growth factors related to inflammation, angiogenesis and hematopoiesis regulation in the BM niche of MPN patients. Methods Soluble mediator levels in BM plasma samples from 17 healthy subjects, 28 ET, 19 PV, and 16 PMF patients were determined using a multiplex assay. Soluble mediator signatures were created from categorical analyses of high mediator producers. Soluble mediator connections and the correlation between plasma levels and clinic-laboratory parameters were also analyzed. Results The soluble mediator signatures of the BM niche of PV patients revealed a highly inflammatory and pro-angiogenic milieu, with increased levels of chemokines (CCL2, CCL5, CXCL8, CXCL12, CXCL10), and growth factors (GM-CSF M-CSF, HGF, IFN-γ, IL-1β, IL-6Ra, IL-12, IL-17, IL-18, TNF-α, VEGF, and VEGF-R2). ET and PMF patients presented intermediate inflammatory and pro-angiogenic profiles. Deregulation of soluble mediators was associated with some clinic-laboratory parameters of MPN patients, including vascular events, treatment status, risk stratification of disease, hemoglobin concentration, hematocrit, and red blood cell count. Conclusions Each MPN subtype exhibits a distinct soluble mediator signature. Deregulated production of BM soluble mediators may contribute to MPN pathogenesis and BM niche modification, provides pro-tumor stimuli, and is a potential target for future therapies.
Collapse
Affiliation(s)
- Juçara Gastaldi Cominal
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maira da Costa Cacemiro
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maria Gabriela Berzoti-Coelho
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Illy Enne Gomes Pereira
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fabiani Gai Frantz
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Elizabeth Xisto Souto
- Department of Clinical Hematology, Euryclides de Jesus Zerbini Transplant Hospital, São Paulo, Brazil
| | - Dimas Tadeu Covas
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Lorena Lobo de Figueiredo-Pontes
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Division of Hematology, Hemotherapy and Cellular Therapy, Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maria Carolina Oliveira
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Division of Rheumatology, Allergy and Immunotherapy, Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Kelen Cristina Ribeiro Malmegrim
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fabíola Attié de Castro
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
22
|
Kosiyo P, Otieno W, Gitaka J, Munde EO, Ouma C. Haematological abnormalities in children with sickle cell disease and non-severe malaria infection in western Kenya. BMC Infect Dis 2021; 21:329. [PMID: 33827455 PMCID: PMC8028187 DOI: 10.1186/s12879-021-06025-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/30/2021] [Indexed: 12/29/2022] Open
Abstract
Background In Plasmodium falciparum infection, clinical conditions such as anaemia, thrombocytopenia and leukocytosis are common. Mutation in haemoglobin sub-unit beta gene (HBB) may be a genetic factor responsible for these haematological changes during infection. However, the contributions of the carriage of different HBB genotypes on these changes remain largely unknown. Methodology In this cross-sectional study, we evaluated haematological abnormalities in P. falciparum-infected children (n = 217, aged 1–192 months) with different haemoglobin sub-unit beta (HBB) genotypes (HbAA, HbAS and HbSS). Children with acute febrile conditions were recruited at Jaramogi Oginga Odinga Teaching and Referral Hospital at the outpatient clinic. Haematological parameters were determined using Beckman Coulter counter ACTdiff2™ while HBB genotyping was done using TaqMan® SNP genotyping assay. Chi-square (χ2) was used to determine differences between proportions. Differences in haematological parameters were compared across groups using Kruskal Wallis test and between groups using Mann Whitney U test. Partial correlation test was used to determine correlation between haematological parameters and sickle cell genotypes while controlling for age and sex. Results Haemoglobin (Hb), [median (IQR); 7.3 (1.3), P = 0.001], haematocrit (HCT), [median (IQR); 26.4 (4.4), P = 0.009], red blood cells (RBC), [median (IQR); 3.2 (1.7), P = 0.048] were markedly reduced in HbSS, however, red cell distribution with (RDW) [median (IQR); 14.9 (3.3), P = 0.030] was increased in malaria infected children with HbSS. Severe anaemia was highest in HbSS (23.1%) followed by HbAA (8.6%) and HbAS (7.1%). There were no differences in platelet count (P = 0.399) hence no severe thrombocytopeania across the genotypes. Leukocytosis was highest in HbSS (69.2%), 42% in HbAS and 31% in HbAA. The RBC, HCT and Hb had negative correlation with RDW in HbSS in malarial-infected children (r = − 0.725, P = 0.008), (r = − 0.718, P = 0.009) and (r = − 0.792, P = 0.002), respectively. Conclusion Our study reveals that anaemia is the most common abnormality in malaria-infected children with carriage of HbSS. The RBC, HCT and Hb concentration decrease with increase in RDW levels in infected children with carriage of HbSS compared to other HBB genotypes. Therefore, carriage of HbSS genotype is correlated with severity of haematological abnormalities. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06025-7.
Collapse
Affiliation(s)
- Paul Kosiyo
- Department of Biomedical Science and Technology, School of Public Health and Community Development, Maseno University, Private Bag, Maseno, Kenya.,Department of Medical Laboratory Sciences, School of Medicine, Maseno University, Private Bag, Maseno, Kenya
| | - Walter Otieno
- Department of Paediatrics and Child Health, School of Medicine, Maseno University, Private Bag, Maseno, Kenya
| | - Jesse Gitaka
- Directorate of Research and Innovation, Mount Kenya University, General Kago Road, P.O. Box 342-01000, Thika, Kenya
| | - Elly O Munde
- Department of Clinical Medicine, School of Health Sciences, Kirinyaga University, P.O. Box 143-10300, Kerugoya, Kenya
| | - Collins Ouma
- Department of Biomedical Science and Technology, School of Public Health and Community Development, Maseno University, Private Bag, Maseno, Kenya.
| |
Collapse
|
23
|
Connes P, Möckesch B, Tudor Ngo Sock E, Hardy-Dessources MD, Reminy K, Skinner S, Billaud M, Nader E, Tressieres B, Etienne-Julan M, Guillot N, Lemonne N, Hue O, Romana M, Antoine-Jonville S. Oxidative stress, inflammation, blood rheology, and microcirculation in adults with sickle cell disease: Effects of hydroxyurea treatment and impact of sickle cell syndrome. Eur J Haematol 2021; 106:800-807. [PMID: 33629431 DOI: 10.1111/ejh.13607] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/16/2022]
Abstract
Inflammation and oxidative stress play a key role in the pathophysiology of sickle cell disease (SCD). However, the potential influence of different sickle genotypes, or hydroxyurea (HU) treatment, on these factors remains poorly documented. The present study compared several plasma markers of inflammation and oxidative stress, as well as microvascular function, between patients with sickle SC disease (HbSC, n = 19) and patients with sickle cell anemia (HbSS) under hydroxyurea (HU) treatment (n = 16), or not (n = 13). Hemorheological parameters and levels of inflammatory (IL-6, IL-8, IFN-γ, MCP-1, MIP-1β, TNF-α) and oxidative stress (AOPP, MDA, MPO) markers were determined. Peripheral microcirculatory cutaneous blood flow and immediate microvascular response to local heat were evaluated using laser Doppler flowmetry. Oxidative stress and inflammation were lower in HbSC patients and HbSS patients under HU therapy compared to HbSS patients not treated with HU. Blood viscosity was higher in HbSC than in HbSS patients treated with or not with HU. Vasodilation response of the cutaneous microcirculation to heat stress was higher in HbSS patients receiving HU treatment. Our results clearly established that both sickle cell genotype and HU treatment modulate inflammation and oxidative stress.
Collapse
Affiliation(s)
- Philippe Connes
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Université Lyon 1 (COMUE Lyon), Equipe "Biologie Vasculaire et du Globule Rouge", Lyon, France.,Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| | - Berenike Möckesch
- EA "ACTES": Adaptation, Climat Tropical, Exercice et Santé, Université des Antilles, Pointe-à-Pitre, France
| | - Emilienne Tudor Ngo Sock
- EA "ACTES": Adaptation, Climat Tropical, Exercice et Santé, Université des Antilles, Pointe-à-Pitre, France
| | - Marie-Dominique Hardy-Dessources
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France.,Université des Antilles, UMR_S1134, BIGR Inserm, Pointe-à-Pitre, France.,Université de Paris, UMR_S1134, BIGR, INSERM, Paris, France
| | - Karen Reminy
- EA "ACTES": Adaptation, Climat Tropical, Exercice et Santé, Université des Antilles, Pointe-à-Pitre, France
| | - Sarah Skinner
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Université Lyon 1 (COMUE Lyon), Equipe "Biologie Vasculaire et du Globule Rouge", Lyon, France.,Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| | - Marie Billaud
- Unité Transversale de la Drépanocytose, CHU de la Guadeloupe, Pointe-à-Pitre, France
| | - Elie Nader
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Université Lyon 1 (COMUE Lyon), Equipe "Biologie Vasculaire et du Globule Rouge", Lyon, France.,Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| | - Benoit Tressieres
- Centre d'Investigation Clinique Antilles Guyane, Pointe-à-Pitre, France
| | - Maryse Etienne-Julan
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France.,Université des Antilles, UMR_S1134, BIGR Inserm, Pointe-à-Pitre, France.,Université de Paris, UMR_S1134, BIGR, INSERM, Paris, France.,Unité Transversale de la Drépanocytose, CHU de la Guadeloupe, Pointe-à-Pitre, France
| | - Nicolas Guillot
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Université Lyon 1 (COMUE Lyon), Equipe "Biologie Vasculaire et du Globule Rouge", Lyon, France.,Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| | - Nathalie Lemonne
- Unité Transversale de la Drépanocytose, CHU de la Guadeloupe, Pointe-à-Pitre, France
| | - Olivier Hue
- EA "ACTES": Adaptation, Climat Tropical, Exercice et Santé, Université des Antilles, Pointe-à-Pitre, France
| | - Marc Romana
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France.,Université des Antilles, UMR_S1134, BIGR Inserm, Pointe-à-Pitre, France.,Université de Paris, UMR_S1134, BIGR, INSERM, Paris, France
| | - Sophie Antoine-Jonville
- EA "ACTES": Adaptation, Climat Tropical, Exercice et Santé, Université des Antilles, Pointe-à-Pitre, France.,Université d'Avignon, LAPEC EA4278, Avignon, France
| |
Collapse
|
24
|
Clinical predictors of poor outcomes in patients with sickle cell disease and COVID-19 infection. Blood Adv 2021; 5:207-215. [PMID: 33570644 PMCID: PMC7802524 DOI: 10.1182/bloodadvances.2020003456] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/25/2020] [Indexed: 12/19/2022] Open
Abstract
SCD patients with COVID-19 display a broad range of severity, with a higher case fatality than the non-SCD population (10.9% vs 3.3%). Older patients not treated with hydroxyurea with end organ damage who present with acute kidney injury, and elevated LDH and D-dimer level are at higher risk of death.
We aimed to identify predictors of outcomes and survival in patients living in 4 major metropolitan areas who had sickle cell disease (SCD) and COVID-19 to inform best approaches to prevention and care. Data were collected at baseline and during the clinical course in SCD patients diagnosed with COVID-19 in four COVID-19 epicenters. Patients were followed up posthospital discharge for up to 3 months. Of sixty-six SCD patients with COVID-19, fifty patients (75%) required hospitalization, and seven died (10.6%). Patients with preexisting kidney disease (chronic kidney disease) were more likely to be hospitalized. The most common presenting symptom was vaso-occlusive pain. Acute chest syndrome occurred in 30 (60%) of the 50 hospitalized patients and in all who died. Older age and histories of pulmonary hypertension, congestive heart failure, chronic kidney disease, and stroke were more prevalent in patients who died, as were higher creatinine, lactate dehydrogenase, and D-dimer levels. Anticoagulation use while inpatient was twice less common in patients who died. All deaths occurred in individuals not taking hydroxyurea or any other SCD-modifying therapy. Patients with SCD and COVID-19 exhibited a broad range of disease severity. We cannot definitively state that the overall mortality is higher in patients with SCD, although our case fatality rate was ∼10% compared with ∼3% in the general population, despite a median age of 34 years. Individuals with SCD aged >50 years, with preexisting cardiopulmonary, renal disease, and/or stroke not receiving hydroxyurea, who present with high serum creatinine, lactate dehydrogenase, and D-dimer levels, are at higher risk of death, irrespective of genotype or sex.
Collapse
|
25
|
Sesti-Costa R, Borges MD, Lanaro C, de Albuquerque DM, Saad STO, Costa FF. Inflammatory Dendritic Cells Contribute to Regulate the Immune Response in Sickle Cell Disease. Front Immunol 2021; 11:617962. [PMID: 33613546 PMCID: PMC7890087 DOI: 10.3389/fimmu.2020.617962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/21/2020] [Indexed: 11/13/2022] Open
Abstract
Sickle cell disease (SCD), one of the most common hemoglobinopathies worldwide, is characterized by a chronic inflammatory component, with systemic release of inflammatory cytokines, due to hemolysis and vaso-occlusive processes. Patients with SCD demonstrate dysfunctional T and B lymphocyte responses, and they are more susceptible to infection. Although dendritic cells (DCs) are the main component responsible for activating and polarizing lymphocytic function, and are able to produce pro-inflammatory cytokines found in the serum of patients with SCD, minimal studies have thus far been devoted to these cells. In the present study, we identified the subpopulations of circulating DCs in patients with SCD, and found that the bloodstream of the patients showed higher numbers and percentages of DCs than that of healthy individuals. Among all the main DCs subsets, inflammatory DCs (CD14+ DCs) were responsible for this rise and correlated with higher reticulocyte count. The patients had more activated monocyte-derived DCs (mo-DCs), which produced MCP-1, IL-6, and IL-8 in culture. We found that a CD14+ mo-DC subset present in culture from some of the patients was the more activated subset and was mainly responsible for cytokine production, and this subset was also responsible for IL-17 production in co-culture with T lymphocytes. Finally, we suggest an involvement of heme oxygenase in the upregulation of CD14 in mo-DCs from the patients, indicating a potential mechanism for inducing inflammatory DC differentiation from circulating monocytes in the patients, which correlated with inflammatory cytokine production, T lymphocyte response skewing, and reticulocyte count.
Collapse
Affiliation(s)
- Renata Sesti-Costa
- Hematology and Hemotherapy Center, University of Campinas, UNICAMP, Campinas, Brazil
| | | | - Carolina Lanaro
- Hematology and Hemotherapy Center, University of Campinas, UNICAMP, Campinas, Brazil
| | | | | | | |
Collapse
|
26
|
Gaur S, Philipovskiy A, Orazi A. Post-operative hyperleukocytosis and leukostasis as the initial presentation of chronic myelomonocytic leukemia: A case report and review of literature. Leuk Res Rep 2021; 16:100283. [PMID: 34934616 PMCID: PMC8654613 DOI: 10.1016/j.lrr.2021.100283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 10/22/2021] [Accepted: 11/27/2021] [Indexed: 11/25/2022] Open
Abstract
Leukemoid reactions following surgery are commonly caused by infections or tissue injury. Management is directed towards underlying condition and cytoreduction is not indicated. Chronic myelo-monocytic leukemia (CMML) is a clonal hematological malignancy characterized by persistent monocytosis and overlapping features of myelodysplastic and myeloproliferative neoplasms.In this case report we describe a 51-year-old Hispanic female without any significant prior medical history, who underwent a cholecystectomy for calculous cholecystitis. Post-operative course was complicated by hyperleukocytosis leading to splenic infarction and intracranial hemorrhage. Further investigations led to a diagnosis of CMML-2. A literature review of patients with CMML who developed post-operative leukocytosis and leukostasis (POLL) is presented.Case high lights two critical points: Post-operative hyperleukocytosis with leukostasis can be the first presentation of CMML Rapid diagnosis and institution of cytoreductive therapy with hydroxyurea is critical to avoid high morbidity and mortality.
Collapse
|
27
|
Morrone KA, Strumph K, Liszewski MJ, Jackson J, Rinke ML, Silver EJ, Minniti C, Davila J, Mitchell WB, Manwani D. Acute chest syndrome in the setting of SARS-COV-2 infections-A case series at an urban medical center in the Bronx. Pediatr Blood Cancer 2020; 67:e28579. [PMID: 32893954 DOI: 10.1002/pbc.28579] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/26/2020] [Accepted: 06/27/2020] [Indexed: 12/21/2022]
Abstract
New York City has emerged as one of the epicenters of the SARS-COV-2 pandemic, with the Bronx being disproportionately affected. This novel coronavirus has caused significant respiratory manifestations raising the concern for development of acute chest syndrome (ACS) in patients with sickle cell disease (SCD). We report a series of pediatric SCD SARS-COV-2-positive patients admitted with ACS. SARS-COV-2-positive SCD patients, who did not develop ACS, were the comparison group. Hydroxyurea use (P-value = .02) and lower absolute monocyte counts (P-value = .04) were noted in patients who did not develop ACS. These preliminary findings need to be further evaluated in larger cohorts.
Collapse
Affiliation(s)
- Kerry A Morrone
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Albert Einstein College of Medicine, New York City, New York
| | - Kaitlin Strumph
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Albert Einstein College of Medicine, New York City, New York
| | - Mark J Liszewski
- Department of Radiology, Division of Pediatric Radiology, Albert Einstein College of Medicine, New York City, New York
| | - Jenai Jackson
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Albert Einstein College of Medicine, New York City, New York
| | - Michael L Rinke
- Department of Pediatrics, Division of Academic General Pediatrics, Albert Einstein College of Medicine, New York City, New York
| | - Ellen J Silver
- Department of Pediatrics, Division of Academic General Pediatrics and Montefiore School Health Program, Albert Einstein College of Medicine, New York City, New York
| | - Caterina Minniti
- Department of Medicine, Division of Hematology, Albert Einstein College of Medicine, New York City, New York
| | - Jennifer Davila
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Albert Einstein College of Medicine, New York City, New York
| | - William B Mitchell
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Albert Einstein College of Medicine, New York City, New York
| | - Deepa Manwani
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Albert Einstein College of Medicine, New York City, New York
| |
Collapse
|
28
|
Hasselbalch HC. Cytokine Profiling as a Novel Complementary Tool to Predict Prognosis in MPNs? Hemasphere 2020; 4:e407. [PMID: 32647805 PMCID: PMC7306306 DOI: 10.1097/hs9.0000000000000407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 02/03/2023] Open
Affiliation(s)
- Hans C Hasselbalch
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| |
Collapse
|
29
|
Ballas SK. The Evolving Pharmacotherapeutic Landscape for the Treatment of Sickle Cell Disease. Mediterr J Hematol Infect Dis 2020; 12:e2020010. [PMID: 31934320 PMCID: PMC6951351 DOI: 10.4084/mjhid.2020.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 12/17/2019] [Indexed: 01/02/2023] Open
Abstract
Sickle cell disease (SCD) is an extremely heterogeneous disease that has been associated with global morbidity and early mortality. More effective and inexpensive therapies are needed. During the last five years, the landscape of the pharmacotherapy of SCD has changed dramatically. Currently, 54 drugs have been used or under consideration to use for the treatment of SCD. These fall into 3 categories: the first category includes the four drugs (Hydroxyurea, L-Glutamine, Crizanlizumab tmca and Voxelotor) that have been approved by the United States Food and Drug Administration (FDA) based on successful clinical trials. The second category includes 22 drugs that failed, discontinued or terminated for now and the third category includes 28 drugs that are actively being considered for the treatment of SCD. Crizanlizumab and Voxelotor are included in the first and third categories because they have been used in more than one trial. New therapies targeting multiple pathways in the complex pathophysiology of SCD have been achieved or are under continued investigation. The emerging trend seems to be the use of multimodal drugs (i.e. drugs that have different mechanisms of action) to treat SCD similar to the use of multiple chemotherapeutic agents to treat cancer.
Collapse
Affiliation(s)
- Samir K Ballas
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|