1
|
Gao C, Dai Y, Spezza PA, Boasiako P, Tang A, Rasquinha G, Zhong H, Shao B, Liu Y, Shi PA, Lobo CA, An X, Guo A, Mitchell WB, Manwani D, Yazdanbakhsh K, Mendelson A. Megakaryocytes transfer mitochondria to bone marrow mesenchymal stromal cells to lower platelet activation. J Clin Invest 2025; 135:e189801. [PMID: 40014405 PMCID: PMC11996913 DOI: 10.1172/jci189801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
Newly produced platelets acquire a low activation state, but whether the megakaryocyte plays a role in this outcome has not been fully uncovered. Mesenchymal stem cells (MSCs) were previously shown to promote platelet production and lower platelet activation. We found that healthy megakaryocytes transfer mitochondria to MSCs, which is mediated by connexin 43 (Cx43) gap junctions on MSCs and leads to platelets at a low energetic state with increased LYN activation, characteristic of resting platelets with increased LYN activation, characteristic of resting platelets. On the contrary, MSCs have a limited ability to transfer mitochondria to megakaryocytes. Sickle cell disease (SCD) is characterized by hemolytic anemia and results in heightened platelet activation, contributing to numerous disease complications. Platelets in SCD mice and human samples had a heightened energetic state with increased glycolysis. MSC exposure to heme in SCD led to decreased Cx43 expression and a reduced ability to uptake mitochondria from megakaryocytes. This prevented LYN activation in platelets and contributed to increased platelet activation at steady state. Altogether, our findings demonstrate an effect of hemolysis in the microenvironment leading to increased platelet activation in SCD. These findings have the potential to inspire new therapeutic targets to relieve thrombosis-related complications of SCD and other hemolytic conditions.
Collapse
Affiliation(s)
| | - Yitian Dai
- Laboratory of Stem Cell Biology and Engineering Research
| | - Paul A. Spezza
- Laboratory of Stem Cell Biology and Engineering Research
| | - Paul Boasiako
- Laboratory of Stem Cell Biology and Engineering Research
| | - Alice Tang
- Laboratory of Stem Cell Biology and Engineering Research
| | | | | | - Bojing Shao
- Laboratory of Vascular Inflammation and Thrombosis Research
| | | | | | - Cheryl A. Lobo
- Laboratory of Blood Borne Parasites, New York Blood Center, New York, New York, USA
| | | | - Anqi Guo
- Laboratory of Complement Biology
| | - William B. Mitchell
- Department of Pediatrics, Montefiore Health Center, Albert Einstein College of Medicine, Children’s Hospital at Montefiore, Bronx, New York, USA
| | - Deepa Manwani
- Department of Pediatrics, Montefiore Health Center, Albert Einstein College of Medicine, Children’s Hospital at Montefiore, Bronx, New York, USA
| | | | | |
Collapse
|
2
|
Qiu Y, Lin J, Wang A, Fang Z, Sakurai Y, Choi H, Williams EK, Hardy ET, Maher K, Coskun AF, Woods G, Lam WA. Clinically relevant clot resolution via a thromboinflammation-on-a-chip. Nature 2025:10.1038/s41586-025-08804-7. [PMID: 40175551 DOI: 10.1038/s41586-025-08804-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/18/2025] [Indexed: 04/04/2025]
Abstract
Thromboinflammation occurs in various diseases, leading to life-threatening microvascular occlusion with resulting end-organ failure1-4. Importantly, how microvascular thromboinflammation resolves remains poorly understood due to the small size-scale of microvasculature and the long duration (weeks to months) of this process. Here we introduce a hydrogel-based thromboinflammation-on-a-chip model with long-term culture capabilities to model microvascular thromboinflammation and monitor clot resolution over clinically and physiologically relevant timescales (up to months). Using this system, we mapped out the distinct temporal phases of clot resolution in microvascular thromboinflammation. Using multiplexed RNA fluorescence in situ hybridization in combination with our thromboinflammation-on-a-chip model, we observed that inflammation shifts the endothelium fibrinolytic balance to favour thrombosis and pinpointed neutrophil elastase as a double-edged sword that induces clot resolution but also tissue damage. We then investigated the mechanisms of potential therapeutic agents that either prevent microvascular thrombosis or accelerate clot resolution. Specifically, we observed that, in thromboinflammation, (1) early tissue plasminogen activator administration within 3 h directly improves endothelial barrier function; (2) prophylactic defibrotide and enoxaparin suppress microvascular thromboinflammation through endothelium-mediated mechanisms; and (3) combining enoxaparin with crizanlizumab reduces microvascular occlusion and protects endothelial function in sickle cell disease. These data introduce a paradigm in investigating the underlying mechanisms of thromboinflammatory clot resolution and conducting drug discovery thereof.
Collapse
Affiliation(s)
- Yongzhi Qiu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Jessica Lin
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA
| | - Audrey Wang
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhou Fang
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Yumiko Sakurai
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hyoann Choi
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA
| | - Evelyn K Williams
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA
| | - Elaissa T Hardy
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA
| | - Kristin Maher
- Division of Hematology, Oncology, Bone Marrow Transplant, and Cellular Therapy, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Ahmet F Coskun
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Gary Woods
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Wilbur A Lam
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
- The Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
3
|
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
|
4
|
Illa AC, Hvid H, Elm T, Frederiksen CA, Bangshof LF, Danielsen DF, Skov S, Dan Ley C. From early development to maturity: a phenotypic analysis of the Townes sickle cell disease mice. Biol Open 2025; 14:bio061828. [PMID: 39912492 PMCID: PMC11832121 DOI: 10.1242/bio.061828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
Well-characterised mouse models of disease may provide valuable insights into pathophysiology. This study characterises the Townes mouse model of sickle cell disease (SCD) and establishes a time window in which the disease is present but does not progress significantly in terms of severity. We examined Townes mice with the HbAA, HbAS, and HbSS genotypes from young (4 weeks) to mature (5 months) stages of life to assess the disease state at different ages and any progression. We conducted blood tests, histological organ damage evaluations, and metabolic assessments to identify a suitable time frame for study based on welfare considerations. Townes HbSS mice displayed key SCD features such as anaemia, haemolysis, thromboinflammation and organ pathology. Notably, these manifestations remained relatively stable over the study period, indicating a stable phase suitable for conducting intervention studies. Mice with HbAS and HbAA genotypes served as comparative controls, showing minimal to no pathology throughout. These findings are valuable for future research on SCD and may ultimately lead to the development of more effective treatments for this debilitating disease.
Collapse
Affiliation(s)
- Ariadna Carol Illa
- Rare Disease Research, Global Drug Discovery, Novo Nordisk A/S, 2760 Måløv, Denmark
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | - Henning Hvid
- Global Discovery & Development Sciences, Global Drug Discovery, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Torben Elm
- Rare Disease Research, Global Drug Discovery, Novo Nordisk A/S, 2760 Måløv, Denmark
| | | | | | | | - Søren Skov
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | - Carsten Dan Ley
- Rare Disease Research, Global Drug Discovery, Novo Nordisk A/S, 2760 Måløv, Denmark
| |
Collapse
|
5
|
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
|
6
|
Choi SS, Kim EJ, Shin SK, Lee JY, Han JW, Kwon EY, Bae HR. Pathway Analysis of Allulose as a Sugar Substitute in Mitigating Thrombotic Risks in Sickle Cell Disease Patients. Nutrients 2024; 16:4295. [PMID: 39770916 PMCID: PMC11678832 DOI: 10.3390/nu16244295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/07/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Long-term consumption of erythritol, a widely used sugar substitute, has been associated with increased risks of thrombosis and cardiometabolic diseases. In this study, we investigated the effects and mechanisms of allulose in mitigating these risks compared to erythritol using the clusterProfiler tool in R (version 4.12.6). Since a high-fat diet (HFD) is known to enhance platelet aggregation, we compared the pathways related to these processes between groups of mice treated with allulose and those treated with erythritol. While erythritol exacerbated HFD-induced increased platelet aggregation, allulose treatment significantly reduced it. Further analysis of platelet gene expression in sickle cell disease (SCD) patients to explore the potential of using sugar substitutes revealed that platelet coagulation mechanisms could be exacerbated by HFD. Additionally, the top up- and downregulated pathways in SCD were significantly reduced in the allulose-treated group compared to the erythritol group. Specific mechanisms related to this include the mitochondrial complex I and mitochondrial translational process as potential pathological factors in platelet coagulation related to SCD. Therefore, this study demonstrates that allulose may offer a safer alternative to erythritol in dietary applications, especially in individuals susceptible to thrombotic events, by modulating critical pathways associated with platelet function and mitochondrial activity.
Collapse
Affiliation(s)
- Seong Su Choi
- Department of Food Science and Nutrition, Kyungpook National University, 80 Daehak-ro, Buk-ku, Daegu 41566, Republic of Korea; (S.S.C.); (E.J.K.); (J.-Y.L.)
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Eun Ji Kim
- Department of Food Science and Nutrition, Kyungpook National University, 80 Daehak-ro, Buk-ku, Daegu 41566, Republic of Korea; (S.S.C.); (E.J.K.); (J.-Y.L.)
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Su-Kyung Shin
- Department of Food Science and Nutrition, Kyungpook National University, 80 Daehak-ro, Buk-ku, Daegu 41566, Republic of Korea; (S.S.C.); (E.J.K.); (J.-Y.L.)
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Ji-Yoon Lee
- Department of Food Science and Nutrition, Kyungpook National University, 80 Daehak-ro, Buk-ku, Daegu 41566, Republic of Korea; (S.S.C.); (E.J.K.); (J.-Y.L.)
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Ji Won Han
- Department of Food Science and Nutrition, Kyungpook National University, 80 Daehak-ro, Buk-ku, Daegu 41566, Republic of Korea; (S.S.C.); (E.J.K.); (J.-Y.L.)
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Eun-Young Kwon
- Department of Food Science and Nutrition, Kyungpook National University, 80 Daehak-ro, Buk-ku, Daegu 41566, Republic of Korea; (S.S.C.); (E.J.K.); (J.-Y.L.)
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
- Center for Beautiful Aging, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Heekyong R. Bae
- Department of Food Science and Nutrition, Kyungpook National University, 80 Daehak-ro, Buk-ku, Daegu 41566, Republic of Korea; (S.S.C.); (E.J.K.); (J.-Y.L.)
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
7
|
Ita MI, Olesen P, Rosing M, Mørk M, Einarsson HB, Riis JJ. Spontaneous Extradural Hematoma in a Sickle Cell Anemia Patient with Hyperinflammation and Thrombotic Microangiopathy Successfully Treated with Eculizumab: A Case Report and Review of the Literature. J Neurol Surg A Cent Eur Neurosurg 2024; 85:625-632. [PMID: 38378045 DOI: 10.1055/a-2271-8772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
BACKGROUND The event of extradural hematoma in the absence of head trauma is a rare central nervous system complication of sickle cell disease. We report here a case of spontaneous extradural hematoma in a patient being treated for sickle cell vasoocclusive crisis complicated by hyperinflammation and thrombotic microangiopathy. The significance of inflammation as an integral component of the pathomechanism of vasoocclusive crisis in patients with sickle cell disease and the role of heme in activating the complement system's alternative pathway are highlighted in this case report. CASE PRESENTATION A teenage patient with sickle cell disease developed a spontaneous right parietal extradural hematoma while receiving treatment for sickle cell vasoocclusive crisis. The concurrent events of hyperinflammation, disseminated intravascular coagulation, hyperhemolysis syndrome, thrombotic microangiopathy, and refractory postoperative bleeding complicated this patient's clinical course after surgical evacuation of extradural hematoma. This patient was subsequently treated with eculizumab and improved in the days following. CONCLUSION Treatment with the anti-C5 monoclonal antibody eculizumab, which targets and inhibits terminal complement system activation, reversed the deleterious cascade of events in this patient with sickle cell disease.
Collapse
Affiliation(s)
- Michael Itak Ita
- Department of Neurological Surgery, Aalborg University Hospital, Aalborg, Denmark
| | - Pia Olesen
- Department of Neurological Surgery, Aalborg University Hospital, Aalborg, Denmark
| | - Maria Rosing
- Department of Neurological Surgery, Aalborg University Hospital, Aalborg, Denmark
| | - Morten Mørk
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | | | - Jens Jakob Riis
- Department of Neurological Surgery, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
8
|
Abiri A, Marmarchinia S, Shah P, Thuptimdang W, Coates TD, Khoo MCK, Khine M. Beat-to-beat analysis of hemodynamic response to mental and psychological stress in sickle cell anemia. JOURNAL OF SICKLE CELL DISEASE 2024; 1:yoae010. [PMID: 40160500 PMCID: PMC11951424 DOI: 10.1093/jscdis/yoae010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/22/2024] [Accepted: 09/19/2024] [Indexed: 04/02/2025]
Abstract
Objectives Vaso-occlusive crises (VOCs) are a hallmark symptom of sickle cell disease (SCD). Physical stressors can trigger decreased microvascular blood flow and increase the risk for VOCs. However, the effect of mental and psychological stressors on vascular physiology in SCD is not well-established. We hereby examined fluctuations in continuous blood pressure (BP) to evaluate hemodynamic changes in SCD patients during mental and psychological stress. Methods Thirteen SCD (HbSS) subjects from the Children's Hospital Los Angeles and 11 healthy (HbAA) volunteers were recruited. Continuous BP was recorded as subjects participated in two mental and one psychological stress tasks. Systolic beat-to-beat BP variability (BPV) measurements were calculated for each subject. Three very short-term BPV metrics served as outcome measures: standard deviation, coefficient of variation, and average real variability. Linear mixed effects models evaluated associations between patient factors and outcome measures. Results SCD patients were associated with increased systolic BPV and exhibited a distinct increase in BPV in response to psychological stress. All subjects exhibited a decrease in systolic BPV in response to mental stress tasks. During mental stress, both groups displayed increased augmentation index, reflective of stress-induced vasoconstriction, while psychological stress in SCD patients led to both decreased mean arterial pressure and increased AI, suggestive of uncompensated vasoconstriction. Conclusion These findings emphasize the impact of mental and psychological stressors on vascular function in SCD, the potential for monitoring physiological signals to predict VOC events, and the importance of counseling SCD patients on lifestyle practices to reduce their stress to prevent pain.
Collapse
Affiliation(s)
- Arash Abiri
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, 92697, United States
| | - Sara Marmarchinia
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, 92697, United States
| | - Payal Shah
- Division of Hematology, Children’s Center for Cancer and Blood Diseases, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States
| | - Wanwara Thuptimdang
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Thomas D Coates
- Division of Hematology, Children’s Center for Cancer and Blood Diseases, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States
| | - Michael C K Khoo
- Department of Biomedical Engineering, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90007, United States
| | - Michelle Khine
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, 92697, United States
| |
Collapse
|
9
|
Ahmed B, Arigliani M, Gupta A. Respiratory management of acute chest syndrome in children with sickle cell disease. Eur Respir Rev 2024; 33:240005. [PMID: 39293855 PMCID: PMC11409057 DOI: 10.1183/16000617.0005-2024] [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: 01/08/2024] [Accepted: 07/01/2024] [Indexed: 09/20/2024] Open
Abstract
Acute chest syndrome (ACS) is a leading cause of respiratory distress and hospitalisation in children with sickle cell disease (SCD). The aetiology is multifactorial and includes fat embolism, venous thromboembolism, alveolar hypoventilation and respiratory infections, with the latter being particularly common in children. These triggers contribute to a vicious cycle of erythrocyte sickling, adhesion to the endothelium, haemolysis, vaso-occlusion and ventilation-perfusion mismatch in the lungs, resulting in the clinical manifestations of ACS. The clinical presentation includes fever, chest pain, dyspnoea, cough, wheeze and hypoxia, accompanied by a new pulmonary infiltrate on chest radiography. Respiratory symptoms may overlap with those of acute asthma, which may be difficult to distinguish. Patients with ACS may deteriorate rapidly; thus prevention, early recognition and aggressive, multidisciplinary team management is essential. In this narrative review, we highlight the current evidence regarding the epidemiology, pathophysiology, treatment and preventative strategies for ACS, focusing on the aspects of major interest for the paediatric pulmonologist and multidisciplinary team who manage children with SCD.
Collapse
Affiliation(s)
- Bushra Ahmed
- UCL GOS Institute of Child Health, University College London, London, UK
| | - Michele Arigliani
- UCL GOS Institute of Child Health, University College London, London, UK
| | - Atul Gupta
- King's College Hospital, King's College London, London, UK
- Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
10
|
Fortuna V, Lima J, Oliveira GF, Oliveira YS, Getachew B, Nekhai S, Aschner M, Tizabi Y. Ferroptosis as an emerging target in sickle cell disease. Curr Res Toxicol 2024; 7:100181. [PMID: 39021403 PMCID: PMC11252799 DOI: 10.1016/j.crtox.2024.100181] [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: 12/30/2023] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Sickle cell disease (SCD) is an inherited hemoglobin disorder marked by red blood cell sickling, resulting in severe anemia, painful episodes, extensive organ damage, and shortened life expectancy. In SCD, increased iron levels can trigger ferroptosis, a specific type of cell death characterized by reactive oxygen species (ROS) and lipid peroxide accumulation, leading to damage and organ impairments. The intricate interplay between iron, ferroptosis, inflammation, and oxidative stress in SCD underscores the necessity of thoroughly understanding these processes for the development of innovative therapeutic strategies. This review highlights the importance of balancing the complex interactions among various factors and exploitation of the knowledge in developing novel therapeutics for this devastating disease.
Collapse
Affiliation(s)
- Vitor Fortuna
- Department of Biochemistry and Biophysics, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Jaqueline Lima
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Gabriel F. Oliveira
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Yasmin S. Oliveira
- Postgraduate Program in Immunology, Health Sciences Institute, Federal University of Bahia, BA 40231-300, Brazil
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Sergei Nekhai
- Center for Sickle Cell Disease, Departments of Microbiology and Medicine, Howard University College of Medicine, Washington, DC, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
11
|
Hassan A, Maki R, Aljawad M, Alzayer A, Habeeb A, Alzaher A, Alawami A, Alaithan F, Adnan J. Beyond pulmonary embolism: Alternative diagnosis and incidental findings on CT pulmonary angiography in sickle cell disease. Emerg Radiol 2024; 31:321-330. [PMID: 38619803 DOI: 10.1007/s10140-024-02229-x] [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: 03/06/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Sickle cell disease (SCD) is a genetic hematological disorder associated with severe complications, such as vaso-occlusive crises, acute chest syndrome (ACS), and an increased risk of thromboembolic events, including pulmonary embolism (PE). The diagnosis of PE in SCD patients presents challenges due to the overlapping symptoms with other pulmonary conditions. Our previous study revealed that nearly 96% of computed tomography pulmonary angiography (CTPA) scans in SCD patients were negative for PE, highlighting a gap in understanding the significance of CTPA findings when PE is absent. METHODS In this retrospective follow-up study conducted at the Salmaniya Medical Complex in Bahrain, we examined SCD patients with HbSS genotypes who underwent CTPA from January 1, 2018, to December 31, 2021, for suspected PE, but the results were negative. The aim of this study was to identify alternative diagnoses and incidental findings from CTPA scans. Experienced radiologists reviewed the CTPA images and reports to assess potential alternative diagnoses and incidental findings, incorporating an additional analysis of chest X-rays to evaluate the diagnostic value of CTPA. Incidental findings were classified based on their location and clinical significance. RESULTS Among the 230 evaluated SCD patients (average age 39.7 years; 53% male) who were CTPA negative for PE, 142 (61.7%) had identifiable alternative diagnoses, primarily pneumonia (49.1%). Notably, 88.0% of these alternative diagnoses had been previously suggested by chest radiographs. Furthermore, incidental findings were noted in 164 (71.3%) patients, with 11.0% deemed clinically significant, necessitating immediate action, and 87.8% considered potentially significant, requiring further assessment. Notable incidental findings included thoracic abnormalities such as cardiomegaly (12.2%) and an enlarged pulmonary artery (11.3%), as well as upper abdominal pathologies such as hepatomegaly (19.6%), splenomegaly (20.9%), and gallstones (10.4%). CONCLUSION This study underscores the limited additional diagnostic yield of CTPA for identifying alternative diagnoses to PE in SCD patients, with the majority of diagnoses, such as pneumonia, already suggested by chest radiographs. The frequent incidental findings, most of which necessitate further evaluation, highlight the need for a cautious and tailored approach to using CTPA in the SCD population.
Collapse
Affiliation(s)
- Ali Hassan
- Radiology Department, Governmental Hospitals, Salmaniya Medical Complex, Manama, Bahrain.
| | - Reem Maki
- Radiology Department, Governmental Hospitals, Salmaniya Medical Complex, Manama, Bahrain
| | - Mahdi Aljawad
- Radiology Department, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Ali Alzayer
- Radiology Department, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Ali Habeeb
- Radiology Department, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Aqeel Alzaher
- Radiology Department, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Adnan Alawami
- Radiology Department, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Fatimah Alaithan
- Radiology Department, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Jalila Adnan
- Radiology Department, Governmental Hospitals, Salmaniya Medical Complex, Manama, Bahrain
| |
Collapse
|
12
|
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
|
13
|
Hopp MT, Ugurlar D, Pezeshkpoor B, Biswas A, Ramoji A, Neugebauer U, Oldenburg J, Imhof D. In-depth structure-function profiling of the complex formation between clotting factor VIII and heme. Thromb Res 2024; 237:184-195. [PMID: 38631156 DOI: 10.1016/j.thromres.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND AND AIMS Blood disorders, such as sickle cell disease, and other clinical conditions are often accompanied by intravascular hemolytic events along with the development of severe coagulopathies. Hemolysis, in turn, leads to the accumulation of Fe(II/III)-protoporphyrin IX (heme) in the intravascular compartment, which can trigger a variety of proinflammatory and prothrombotic reactions. As such, heme binding to the blood coagulation proteins factor VIII (FVIII), fibrinogen, and activated protein C with functional consequences has been demonstrated earlier. METHODS We herein present an in-depth characterization of the FVIII-heme interaction at the molecular level and its (patho-)physiological relevance through the application of biochemical, biophysical, structural biology, bioinformatic, and diagnostic tools. RESULTS FVIII has a great heme-binding capacity with seven heme molecules associating with the protein. The respective binding sites were identified by investigating heme binding to FVIII-derived peptides in combination with molecular docking and dynamic simulation studies of the complex as well as cryo-electron microscopy, revealing three high-affinity and four moderate heme-binding motifs (HBMs). Furthermore, the relevance of the FVIII-heme complex formation was characterized in physiologically relevant assay systems, revealing a ~ 50 % inhibition of the FVIII cofactor activity even in the protein-rich environment of blood plasma. CONCLUSION Our study provides not only novel molecular insights into the FVIII-heme interaction and its physiological relevance, but also strongly suggests the reduction of the intrinsic pathway and the accentuation of the final clotting step (by, for example, fibrinogen crosslinking) in hemolytic conditions as well as a future perspective in the context of FVIII substitution therapy of hemorrhagic events in hemophilia A patients.
Collapse
Affiliation(s)
- Marie-T Hopp
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, Bonn, Germany; Department of Chemistry, Institute for Integrated Natural Sciences, University of Koblenz, Koblenz, Germany.
| | - Deniz Ugurlar
- Center for Electron Microscopy, Thermo Fisher Scientific, Eindhoven, the Netherlands
| | - Behnaz Pezeshkpoor
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Arijit Biswas
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Anuradha Ramoji
- Leibniz Institute of Photonic Technology, Jena, Germany; Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Jena, Germany
| | - Ute Neugebauer
- Leibniz Institute of Photonic Technology, Jena, Germany; Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Johannes Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, Bonn, Germany.
| |
Collapse
|
14
|
Schrottmaier WC, Assinger A. The Concept of Thromboinflammation. Hamostaseologie 2024; 44:21-30. [PMID: 38417802 DOI: 10.1055/a-2178-6491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2024] Open
Abstract
Inflammation and thrombosis are intricate and closely interconnected biological processes that are not yet fully understood and lack effective targeted therapeutic approaches. Thrombosis initiated by inflammatory responses, known as immunothrombosis, can confer advantages to the host by constraining the spread of pathogens within the bloodstream. Conversely, platelets and the coagulation cascade can influence inflammatory responses through interactions with immune cells, endothelium, or complement system. These interactions can lead to a state of heightened inflammation resulting from thrombotic processes, termed as thromboinflammation. This review aims to comprehensively summarize the existing knowledge of thromboinflammation and addressing its significance as a challenging clinical issue.
Collapse
Affiliation(s)
- Waltraud C Schrottmaier
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Alice Assinger
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
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
|
16
|
Hamali HA. Hypercoagulability in Sickle Cell Disease: A Thrombo-Inflammatory Mechanism. Hemoglobin 2023; 47:205-214. [PMID: 38189099 DOI: 10.1080/03630269.2023.2301026] [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: 04/20/2023] [Accepted: 12/28/2023] [Indexed: 01/09/2024]
Abstract
Sickle cell disease (SCD) is a group of inherited disorders characterized by the presence of abnormal hemoglobin S. Patients with SCD suffer from frequent episodes of anemia, chronic hemolysis, pain crisis, and vaso-occlusion. Additionally, SCD is associated with diverse and serious clinical complications, including thrombosis, which can lead to organ failure, increased morbidity, and eventually, mortality. SCD is known to be a hypercoagulable condition, and the cause of hypercoagulability is multifactorial, with the molecular basis of hemoglobin S being the main driver. The presence of hemoglobin S induces sickling of the RBCs and their subsequent hemolysis, as well as oxidative stress. Both of these processes can alter the hemostatic system, through the activation of platelets, coagulation system, and fibrinolysis, as well as depletion of coagulation inhibitors. These changes can also induce the formation of microvesicles and expression of tissue factor, leading to activation of WBCs, endothelial cell damage, and inflammatory response. Understanding the various factors that drive hypercoagulability as a thrombo-inflammatory mechanism in SCD can help provide explanations for the pathogenesis and other complications of the disease.
Collapse
Affiliation(s)
- Hassan A Hamali
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| |
Collapse
|
17
|
Liang T, Guo K, Ni P, Duan G, Zhang R. The association of sickle cell disorder with adverse outcomes in COVID-19 patients: A meta-analysis. J Med Virol 2023; 95:e29120. [PMID: 37772453 DOI: 10.1002/jmv.29120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/30/2023]
Abstract
The aim is to elucidate the relationship between sickle cell disorder and severe COVID-19. We systematically searched the required articles in three electronic databases, extracting and pooling effect sizes (ES) and 95% confidence interval (CI) from each eligible study to evaluate the effect of combined sickle cell disorder on adverse consequences in patients with COVID-19. This meta-analysis included 21 studies. Sickle cell disease (SCD) was a risk factor for mortality (pooled ES = 1.70, 95% CI: 1.00-2.92, p = 0.001), hospitalization (pooled ES = 6.21, 95% CI: 3.60-10.70, p = 0.000) and intensive care unit (ICU) admission (pooled ES = 2.29, 95% CI: 1.61-3.24, p = 0.099) in COVID-19 patients. Patients with SCD had an increased risk of respiratory failure/mechanical ventilation, but a statistical association was not found (pooled ES = 1.21, 95%CI: 0.74-1.98, p = 0.036). There was significant heterogeneity between SCD and death, hospitalization, and respiratory failure/mechanical ventilation. The results of meta-regression of SCD and hospitalization suggested that the tested variables including Area (p = 0.642), study design (p = 0.739), sample size (p = 0.397), proportion of males (p = 0.708), effect type (p = 0.723), whether confounding factors are adjusted (p = 0.606) might not be the source of heterogeneity. In addition, sickle cell trait (SCT) was significantly associated with the mortality (pooled ES = 1.54, 95% CI: 1.28-1.85, p = 0.771) and hospitalization (pooled ES = 1.20, 95% CI: 1.07-1.35,p = 0.519) in patients with COVID-19. But any increased risk of ICU admission/severe (pooled ES = 1.24, 95% CI: 0.95-1.62, p = 0.520) and mechanical ventilation (OR = 1.00, 95%CI:0.59-1.69) in COVID-19 patients with SCT was not observed. Sensitivity analysis demonstrated that the results were robust. The results of the funnel plot and Egger's test did not support the existence of publication bias. Current meta-analysis indicated that sickle cell disorder has a meaningful impact on COVID-19 progression to severe cases and associated deaths. However, further investigations and research to validate the current findings is indispensable.
Collapse
Affiliation(s)
- Tianyi Liang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Kaixin Guo
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Peng Ni
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Guangcai Duan
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Rongguang Zhang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, China
- International School of Public Health and One Health and The First Affiliated Hospital, Hainan Medical University, Haikou, China
| |
Collapse
|
18
|
Cavalcante JS, de Almeida DEG, Santos-Filho NA, Sartim MA, de Almeida Baldo A, Brasileiro L, Albuquerque PL, Oliveira SS, Sachett JAG, Monteiro WM, Ferreira RS. Crosstalk of Inflammation and Coagulation in Bothrops Snakebite Envenoming: Endogenous Signaling Pathways and Pathophysiology. Int J Mol Sci 2023; 24:11508. [PMID: 37511277 PMCID: PMC10380640 DOI: 10.3390/ijms241411508] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/26/2023] [Accepted: 06/05/2023] [Indexed: 07/30/2023] Open
Abstract
Snakebite envenoming represents a major health problem in tropical and subtropical countries. Considering the elevated number of accidents and high morbidity and mortality rates, the World Health Organization reclassified this disease to category A of neglected diseases. In Latin America, Bothrops genus snakes are mainly responsible for snakebites in humans, whose pathophysiology is characterized by local and systemic inflammatory and degradative processes, triggering prothrombotic and hemorrhagic events, which lead to various complications, organ damage, tissue loss, amputations, and death. The activation of the multicellular blood system, hemostatic alterations, and activation of the inflammatory response are all well-documented in Bothrops envenomings. However, the interface between inflammation and coagulation is still a neglected issue in the toxinology field. Thromboinflammatory pathways can play a significant role in some of the major complications of snakebite envenoming, such as stroke, venous thromboembolism, and acute kidney injury. In addition to exacerbating inflammation and cell interactions that trigger vaso-occlusion, ischemia-reperfusion processes, and, eventually, organic damage and necrosis. In this review, we discuss the role of inflammatory pathways in modulating coagulation and inducing platelet and leukocyte activation, as well as the inflammatory production mediators and induction of innate immune responses, among other mechanisms that are altered by Bothrops venoms.
Collapse
Affiliation(s)
- Joeliton S Cavalcante
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP-Univ Estadual Paulista), Botucatu 18618-687, São Paulo, Brazil
| | - Denis Emanuel Garcia de Almeida
- Department of Bioprocess and Biotechnology, School of Agriculture, Agronomic Sciences School, São Paulo State University (UNESP-Univ Estadual Paulista), Botucatu 18618-687, São Paulo, Brazil
| | - Norival A Santos-Filho
- Institute of Chemistry, São Paulo State University (UNESP-Univ Estadual Paulista), Araraquara 14800-900, São Paulo, Brazil
| | - Marco Aurélio Sartim
- Laboratory of Bioprospection, University Nilton Lins, Manaus 69058-030, Amazonas, Brazil
- Research & Development Department, Nilton Lins Foundation, Manaus 69058-030, Amazonas, Brazil
- Graduate Program in Tropical Medicine, Department of Research at Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Amazonas State University, Manaus 69850-000, Amazonas, Brazil
| | - Amanda de Almeida Baldo
- Institute of Biosciences, São Paulo State University (UNESP-Univ Estadual Paulista), Botucatu 18618-687, São Paulo, Brazil
| | - Lisele Brasileiro
- Graduate Program in Tropical Medicine, Department of Research at Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Amazonas State University, Manaus 69850-000, Amazonas, Brazil
| | - Polianna L Albuquerque
- Toxicological Information and Assistance Center, Instituto Doutor Jose Frota Hospital, Fortaleza 60025-061, Ceará, Brazil
- Faculty of Medicine, University of Fortaleza, Fortaleza 60430-140, Ceará, Brazil
| | - Sâmella S Oliveira
- Research Management, Hospital Foundation of Hematology and Hemotherapy of Amazonas, Manaus 69050-001, Amazonas, Brazil
| | - Jacqueline Almeida Gonçalves Sachett
- Research & Development Department, Nilton Lins Foundation, Manaus 69058-030, Amazonas, Brazil
- Graduate Program in Tropical Medicine, Department of Research at Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Amazonas State University, Manaus 69850-000, Amazonas, Brazil
| | - Wuelton Marcelo Monteiro
- Research & Development Department, Nilton Lins Foundation, Manaus 69058-030, Amazonas, Brazil
- Graduate Program in Tropical Medicine, Department of Research at Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Amazonas State University, Manaus 69850-000, Amazonas, Brazil
| | - Rui Seabra Ferreira
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP-Univ Estadual Paulista), Botucatu 18618-687, São Paulo, Brazil
- Center for Translational Science and Development of Biopharmaceuticals FAPESP/CEVAP-UNESP, Botucatu 18610-307, São Paulo, Brazil
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP-Univ Estadual Paulista), Botucatu 18610-307, São Paulo, Brazil
| |
Collapse
|
19
|
Vital EF, Lam WA. Hidden behind thromboinflammation: revealing the roles of von Willebrand factor in sickle cell disease pathophysiology. Curr Opin Hematol 2023; 30:86-92. [PMID: 36853830 PMCID: PMC10065920 DOI: 10.1097/moh.0000000000000755] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
PURPOSE OF REVIEW This review provides an update on the pathophysiology of sickle cell disease (SCD) with a particular focus on the dysregulation of the von Willebrand factor (VWF) - ADAMTS13 axis that contributes to its pathogenesis. In discussing recent developments, we hope to encourage new and ongoing discussions surrounding therapeutic targets for SCD. RECENT FINDINGS Within the last 5 years, the role of VWF in the pathophysiology of SCD has been further elucidated and is now a target of study in ongoing clinical trials. SUMMARY The pathophysiology of SCD is multifaceted, as it involves systemwide vascular activation, altered blood rheology, and the activation of immune responses and coagulative pathways. The presence of VWF in excess in SCD, particularly in its largest multimeric form, greatly contributes to its pathogenesis. Understanding the molecular mechanisms that underly the presence of large VWF multimers in SCD will provide further insight into the pathogenesis of SCD and provide specific targets for therapy.
Collapse
Affiliation(s)
- Eudorah F. Vital
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University
- Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta and Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Wilbur A. Lam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University
- Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta and Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
20
|
Gotardo ÉMF, Brito PL, Gushiken LFS, Chweih H, Leonardo FC, Costa FF, Conran N. Molecular and cellular effects of in vivo chronic intravascular hemolysis and anti-inflammatory therapeutic approaches. Vascul Pharmacol 2023; 150:107176. [PMID: 37116732 DOI: 10.1016/j.vph.2023.107176] [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: 01/24/2023] [Revised: 04/11/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023]
Abstract
Intravascular hemolysis (IVH) occurs in numerous inherited and acquired disorders, including sickle cell disease (SCD), malaria and sepsis. These diseases display unique symptoms, but often share complications, such as vasomotor dysfunction and pulmonary hypertension. Consequently, in vivo models are needed to study the effects of continuous intravascular hemolytic processes, independently of the molecular or extrinsic alteration that leads to erythrocyte destruction. We gave twice-weekly low-dose phenylhydrazine (LDPHZ) to C57BL/6 J mice for 4 weeks, and measured parameters indicative of anemia, hemoglobin-clearance pathways, inflammation and iron turnover, comparing these to those of a murine model of SCD, which displays associated IVH. LDPHZ administration provoked discreet anemia in mice and significant reticulocytosis, in association with hemoglobin/heme-clearance pathway protein depletion. Mice subjected to chronic hemolysis displayed elevated leukocyte counts and plasma levels of interleukin (IL)-1β, TNF-α, IL-6, soluble ICAM-1, endothelin-1 and anti-inflammatory IL-10, closely emulating alterations indicative of systemic inflammatory and endothelial activation in SCD, and confirming chronic IVH in itself as a serious complication. Discreet accelerations in hepatic and splenic iron turnover also occurred in LDPHZ mice, without alterations in liver damage markers. Examining the effects of two therapies on hemolysis-induced inflammation, the administration of hydroxyurea (and to a lesser extent, l-glutamine) significantly abrogated hemolytic inflammation in mice, without apparent inhibition of hemolysis. In conclusion, the isolation of chronic IVH, a common disease mechanism, using this model, may allow the study of hemolysis-specific sequelae at the cellular and systemic level, and the investigation of candidate agents that could potentially counter hemolytic inflammation.
Collapse
Affiliation(s)
- Érica M F Gotardo
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil.
| | - Pâmela L Brito
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Lucas F S Gushiken
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Hanan Chweih
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Flavia C Leonardo
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Fernando F Costa
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Nicola Conran
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil.
| |
Collapse
|
21
|
Shet AS. FXII and sickle cell: the clot thickens. Blood 2023; 141:1787-1789. [PMID: 37052945 PMCID: PMC10122103 DOI: 10.1182/blood.2023019642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023] Open
|
22
|
de Lima F, Moraes CRP, Barbosa MS, Bombassaro B, Palma AC, Dertkigil SSJ, Moretti ML, Orsi FA, Annichino-Bizzacchi JM, Mansour E, Velloso LA, De Paula EV. Association of heme-oxygenase 1, hemopexin, and heme levels with markers of disease severity in COVID-19. Exp Biol Med (Maywood) 2023; 248:309-316. [PMID: 36740756 PMCID: PMC9902789 DOI: 10.1177/15353702221139185] [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] [Indexed: 02/07/2023] Open
Abstract
Heme-oxygenase 1 (HO-1) is an enzyme with well-known anti-inflammatory and antioxidant properties, whose levels have been previously associated with disease severity in the context of sterile and infectious diseases. Moreover, the heme/HO-1 pathway has been associated with prothrombotic changes in other diseases. Accordingly, the potential of modulating HO-1 levels for the treatment of COVID-19 was extensively speculated during the COVID-19 pandemic, but very few actual data were generated. The aim of our study was to explore the association of HO-1, heme, and hemopexin (HPX) levels with COVID-19 severity and with markers of inflammation and coagulation activation. The study was conducted in 30 consecutive patients with COVID-19 admitted due to hypoxemia, and 30 healthy volunteers matched by sex, age, and geographic region. HO-1 and HPX levels were measured by enzyme immunoassay (ELISA) and heme levels were measured by a colorimetric method. A comprehensive panel of coagulation and fibrinolysis activation was also used. Patients with COVID-19 presented increased levels of HO-1 when compared to controls (5741 ± 2696 vs 1953 ± 612 pg/mL, respectively, P < 0.0001), as well as a trend toward increased levels of HPX (3.724 ± 0.880 vs 3.254 ± 1.022 mg/mL, respectively; P = 0.06). In addition, HO-1 and HPX levels reduced from admission to day + 4. HO-1 levels were associated with duration of intensive care unit stay and with several markers of coagulation activation. In conclusion, modulation of HO-1 could be associated with the prothrombotic state observed in COVID-19, and HO-1 could also represent a relevant biomarker for COVID-19. New independent studies are warranted to explore and expand these findings.
Collapse
Affiliation(s)
- Franciele de Lima
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil,Franciele de Lima.
| | | | - Mayck Silva Barbosa
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, University of Campinas, Campinas 13083-864, Brazil
| | - André C Palma
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | | | - Maria Luiza Moretti
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | | | - Joyce M Annichino-Bizzacchi
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil,Hematology and Hemotherapy Center, University of Campinas, Campinas 13083-878, Brazil
| | - Eli Mansour
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | - Licio A Velloso
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil,Obesity and Comorbidities Research Center, University of Campinas, Campinas 13083-864, Brazil
| | - Erich Vinicius De Paula
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil,Hematology and Hemotherapy Center, University of Campinas, Campinas 13083-878, Brazil
| |
Collapse
|
23
|
Knisely MR, Yang Q, Stauffer N, Kenney M, Ashley-Koch A, Myers J, Walker JKL, Tanabe PJ, Shah NR. Evaluating Associations between Average Pain Intensity and Genetic Variation in People with Sickle Cell Disease: An Exploratory Study. Pain Manag Nurs 2023; 24:12-18. [PMID: 36096903 PMCID: PMC9925395 DOI: 10.1016/j.pmn.2022.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/18/2022] [Accepted: 08/08/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Pain is one of the most common and deleterious symptoms experienced by individuals with sickle cell disease (SCD). There is a paucity of studies identifying potential genetic mechanisms of pain in this population. AIM Examine associations between 11 functional single nucleotide polymorphisms in 9 candidate genes with reports of average pain intensity in individuals with sickle cell disease. METHOD Cross-sectional analyses were performed on data and blood samples collected through the Duke SCD Implementation Consortium Registry. Participants were asked to rate their pain "on the average" using an 11-point numeric rating scale (0 = no pain; 10 = pain as bad as you can imagine). We genotyped 11 single nucleotide polymorphisms in 9 pain-related genes using TaqMan® Genotyping Assays. Associations between each polymorphism and reports of average pain were evaluated. RESULTS The 86 participants (mean age: 28.7 years; 64% female) included in this study reported moderate pain on average (Mean = 4, Standard Deviation = 2.4). ICAM1 rs1799969 was the only genetic polymorphism that was significantly associated with pain (p = .01). Individuals with one or more minor alleles had lower average pain (Mean = 1.25, Standard Deviation = 1.50) than individuals without a minor allele (Mean = 4.13, Standard Deviation = 2.25). The effect size for ICAM1 rs1799969 was 1.30, which is considered large. The effect sizes for all other single nucleotide polymorphisms ranged from small to medium (range: 0-0.3). CONCLUSIONS Our findings provide preliminary evidence that the minor allele in ICAM1 rs1799969 had protective effects against experiencing more severe pain in sickle cell disease.
Collapse
Affiliation(s)
| | - Qing Yang
- Duke University School of Nursing, Durham, North Carolina
| | - Nic Stauffer
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, North Carolina
| | - Martha Kenney
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina
| | - Allison Ashley-Koch
- Departments of Medicine and Biostatistics and Bioinformatics, Duke Molecular Physiology Institute, Durham, North Carolina; Duke University School of Medicine, Durham, North Carolina
| | - John Myers
- Duke University School of Nursing, Durham, North Carolina
| | | | - Paula J Tanabe
- Duke University School of Nursing, Durham, North Carolina
| | - Nirmish R Shah
- Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
24
|
Chiang KC, Gupta A, Sundd P, Krishnamurti L. Thrombo-Inflammation in COVID-19 and Sickle Cell Disease: Two Faces of the Same Coin. Biomedicines 2023; 11:338. [PMID: 36830874 PMCID: PMC9953430 DOI: 10.3390/biomedicines11020338] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 01/26/2023] Open
Abstract
People with sickle cell disease (SCD) are at greater risk of severe illness and death from respiratory infections, including COVID-19, than people without SCD (Centers for Disease Control and Prevention, USA). Vaso-occlusive crises (VOC) in SCD and severe SARS-CoV-2 infection are both characterized by thrombo-inflammation mediated by endothelial injury, complement activation, inflammatory lipid storm, platelet activation, platelet-leukocyte adhesion, and activation of the coagulation cascade. Notably, lipid mediators, including thromboxane A2, significantly increase in severe COVID-19 and SCD. In addition, the release of thromboxane A2 from endothelial cells and macrophages stimulates platelets to release microvesicles, which are harbingers of multicellular adhesion and thrombo-inflammation. Currently, there are limited therapeutic strategies targeting platelet-neutrophil activation and thrombo-inflammation in either SCD or COVID-19 during acute crisis. However, due to many similarities between the pathobiology of thrombo-inflammation in SCD and COVID-19, therapies targeting one disease may likely be effective in the other. Therefore, the preclinical and clinical research spurred by the COVID-19 pandemic, including clinical trials of anti-thrombotic agents, are potentially applicable to VOC. Here, we first outline the parallels between SCD and COVID-19; second, review the role of lipid mediators in the pathogenesis of these diseases; and lastly, examine the therapeutic targets and potential treatments for the two diseases.
Collapse
Affiliation(s)
| | - Ajay Gupta
- KARE Biosciences, Orange, CA 89128, USA
- Division of Nephrology, Hypertension and Kidney Transplantation, Department of Medicine, University of California Irvine (UCI) School of Medicine, Irvine, CA 92868, USA
| | - Prithu Sundd
- Vascular Medicine Institute and Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Lakshmanan Krishnamurti
- Division of Pediatric Hematology-Oncology, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
25
|
Changes in Heme Levels During Acute Vaso-occlusive Crisis in Sickle Cell Anemia. Hematol Oncol Stem Cell Ther 2023; 16:124-132. [PMID: 34450106 DOI: 10.1016/j.hemonc.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/20/2021] [Accepted: 08/09/2021] [Indexed: 01/31/2023] Open
Abstract
OBJECTIVE/BACKGROUND Sickle cell anemia (SCA) is associated with increased levels of extracellular heme, which is a key mediator of inflammation in this condition. Despite abundant evidence supporting this concept in cell and animal models, few studies addressed the association between heme levels and the development and severity of acute vasoocclusive crises (VOC) in humans. METHODS A cross-sectional study was conducted in patients with acute VOC. Total extracellular heme levels were measured in both plasma and serum at admission and after convalescence, and correlated with other clinical and laboratory markers of SCA severity. RESULTS A total of 28 episodes of VOC in 25 patients were included. Heme levels were similar between admission and convalescence, and correlated with the difference between pre and post hemoglobin, and SCA severity estimated by a composite score of clinical and laboratory markers. Heme levels were neither associated with VOC severity nor with markers of hemostasis activation, and were similar to those reported in an independent population of SCA patients at steady state. DISCUSSION Acute VOC are not characterized by significant increases in total extracellular heme levels. Studies measuring the fraction of free extracellular heme unbound to proteins are warranted to further refine our understanding of the role of heme in acute VOC.
Collapse
|
26
|
Thangaraju K, Setua S, Lisk C, Swindle D, Stephenson D, Dzieciatkowska M, Lamb DR, Moitra P, Pak D, Hassell K, George G, Nuss R, Davizon-Castillo P, Stenmark KR, D’Alessandro A, Irwin DC, Buehler PW. Extracellular Vesicle Size Reveals Cargo Specific to Coagulation and Inflammation in Pediatric and Adult Sickle Cell Disease. Clin Appl Thromb Hemost 2023; 29:10760296231186144. [PMID: 37469147 PMCID: PMC10363884 DOI: 10.1177/10760296231186144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/02/2023] [Accepted: 06/18/2023] [Indexed: 07/21/2023] Open
Abstract
Aberrant coagulation in sickle cell disease (SCD) is linked to extracellular vesicle (EV) exposure. However, there is no consensus on the contributions of small EVs (SEVs) and large EVs (LEVs) toward underlying coagulopathy or on their molecular cargo. The present observational study compared the thrombin potential of SEVs and LEVs isolated from the plasma of stable pediatric and adult SCD patients. Further, EV lipid and protein contents were analyzed to define markers consistent with activation of thrombin and markers of underlying coagulopathy. Results suggested that LEVs-but not SEVs-from pediatrics and adults similarly enhanced phosphatidylserine (PS)-dependent thrombin generation, and cell membrane procoagulant PS (18:0;20:4 and 18:0;18:1) were the most abundant lipids found in LEVs. Further, LEVs showed activated coagulation in protein pathway analyses, while SEVs demonstrated high levels of cholesterol esters and a protein pathway analysis that identified complement factors and inflammation. We suggest that thrombin potential of EVs from both stable pediatric and adult SCD patients is similarly dependent on size and show lipid and protein contents that identify underlying markers of coagulation and inflammation.
Collapse
Affiliation(s)
- Kiruphagaran Thangaraju
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Saini Setua
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Christina Lisk
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, USA
| | - Delaney Swindle
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, USA
| | - Daniel Stephenson
- Department of Biochemistry & Molecular Genetics, Graduate School, University of Colorado, Anschutz, Medical Campus, Aurora, CO, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry & Molecular Genetics, Graduate School, University of Colorado, Anschutz, Medical Campus, Aurora, CO, USA
| | - Derek R. Lamb
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Parikshit Moitra
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David Pak
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, USA
| | - Kathryn Hassell
- Division of Hematology Colorado Sickle Cell Treatment and Research Center, School of Medicine, Anschutz Medical Campus, University of Colorado-Denver School of Medicine, Aurora, CO, USA
| | - Gemlyn George
- Division of Hematology Colorado Sickle Cell Treatment and Research Center, School of Medicine, Anschutz Medical Campus, University of Colorado-Denver School of Medicine, Aurora, CO, USA
| | - Rachelle Nuss
- Division of Hematology Colorado Sickle Cell Treatment and Research Center, School of Medicine, Anschutz Medical Campus, University of Colorado-Denver School of Medicine, Aurora, CO, USA
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Pavel Davizon-Castillo
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Kurt R. Stenmark
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Angelo D’Alessandro
- Department of Biochemistry & Molecular Genetics, Graduate School, University of Colorado, Anschutz, Medical Campus, Aurora, CO, USA
| | - David C. Irwin
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, USA
| | - Paul W. Buehler
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
27
|
Xu JZ, Conrey A, Frey I, Gwaabe E, Menapace LA, Tumburu L, Lundt M, Lequang T, Li Q, Glass K, Dunkelberger EB, Iyer V, Mangus H, Kung C, Dang L, Kosinski PA, Hawkins P, Jeffries N, Eaton WA, Lay Thein S. A phase 1 dose escalation study of the pyruvate kinase activator mitapivat (AG-348) in sickle cell disease. Blood 2022; 140:2053-2062. [PMID: 35576529 PMCID: PMC9837441 DOI: 10.1182/blood.2022015403] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/11/2022] [Indexed: 01/21/2023] Open
Abstract
Polymerization of deoxygenated hemoglobin S underlies the pathophysiology of sickle cell disease (SCD). In activating red blood cell pyruvate kinase and glycolysis, mitapivat (AG-348) increases adenosine triphosphate (ATP) levels and decreases the 2,3-diphosphoglycerate (2,3-DPG) concentration, an upstream precursor in glycolysis. Both changes have therapeutic potential for patients with SCD. Here, we evaluated the safety and tolerability of multiple ascending doses of mitapivat in adults with SCD with no recent blood transfusions or changes in hydroxyurea or l-glutamine therapy. Seventeen subjects were enrolled; 1 subject was withdrawn shortly after starting the study. Sixteen subjects completed 3 ascending dose levels of mitapivat (5, 20, and 50 mg, twice daily [BID]) for 2 weeks each; following a protocol amendment, the dose was escalated to 100 mg BID in 9 subjects. Mitapivat was well tolerated at all dose levels, with the most common treatment-emergent adverse events (AEs) being insomnia, headache, and hypertension. Six serious AEs (SAEs) included 4 vaso-occlusive crises (VOCs), non-VOC-related shoulder pain, and a preexisting pulmonary embolism. Two VOCs occurred during drug taper and were possibly drug related; no other SAEs were drug related. Mean hemoglobin increase at the 50 mg BID dose level was 1.2 g/dL, with 9 of 16 (56.3%) patients achieving a hemoglobin response of a ≥1 g/dL increase compared with baseline. Mean reductions in hemolytic markers and dose-dependent decreases in 2,3-DPG and increases in ATP were also observed. This study provides proof of concept that mitapivat has disease-modifying potential in patients with SCD. This trial was registered at www.clinicaltrials.gov as #NCT04000165.
Collapse
Affiliation(s)
- Julia Z. Xu
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Anna Conrey
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Ingrid Frey
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Eveline Gwaabe
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Laurel A. Menapace
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Laxminath Tumburu
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Maureen Lundt
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Timothy Lequang
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Quan Li
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Kristen Glass
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Emily B. Dunkelberger
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | | | | | | | - Lenny Dang
- Agios Pharmaceuticals, Inc., Cambridge, MA
| | | | | | - Neal Jeffries
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - William A. Eaton
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
28
|
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
|
29
|
Mubeen S, Domingo-Fernández D, Díaz del Ser S, Solanki DM, Kodamullil AT, Hofmann-Apitius M, Hopp MT, Imhof D. Exploring the Complex Network of Heme-Triggered Effects on the Blood Coagulation System. J Clin Med 2022; 11:jcm11195975. [PMID: 36233841 PMCID: PMC9572022 DOI: 10.3390/jcm11195975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/16/2022] Open
Abstract
Excess labile heme, occurring under hemolytic conditions, displays a versatile modulator in the blood coagulation system. As such, heme provokes prothrombotic states, either by binding to plasma proteins or through interaction with participating cell types. However, despite several independent reports on these effects, apparently contradictory observations and significant knowledge gaps characterize this relationship, which hampers a complete understanding of heme-driven coagulopathies and the development of suitable and specific treatment options. Thus, the computational exploration of the complex network of heme-triggered effects in the blood coagulation system is presented herein. Combining hemostasis- and heme-specific terminology, the knowledge available thus far was curated and modeled in a mechanistic interactome. Further, these data were incorporated in the earlier established heme knowledge graph, "HemeKG", to better comprehend the knowledge surrounding heme biology. Finally, a pathway enrichment analysis of these data provided deep insights into so far unknown links and novel experimental targets within the blood coagulation cascade and platelet activation pathways for further investigation of the prothrombotic nature of heme. In summary, this study allows, for the first time, a detailed network analysis of the effects of heme in the blood coagulation system.
Collapse
Affiliation(s)
- Sarah Mubeen
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
| | - Daniel Domingo-Fernández
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
- Enveda Biosciences, Inc., San Francisco, CA 94080, USA
| | - Sara Díaz del Ser
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
- Polytechnic University of Madrid, E-28040 Madrid, Spain
| | - Dhwani M. Solanki
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Alpha T. Kodamullil
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
- Causality Biomodels, Kinfra Hi-Tech Park, Kalamassery, Cochin 683503, Kerala, India
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
| | - Marie-T. Hopp
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Correspondence: (M.-T.H.); (D.I.); Tel.: +49-228-73-5231 (M.-T.H.); +49-228-73-5254 (D.I.)
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Correspondence: (M.-T.H.); (D.I.); Tel.: +49-228-73-5231 (M.-T.H.); +49-228-73-5254 (D.I.)
| |
Collapse
|
30
|
Feugray G, Kasonga F, Grall M, Dumesnil C, Benhamou Y, Brunel V, Le Cam Duchez V, Lahary A, Billoir P. Investigation of thrombin generation assay to predict vaso-occlusive crisis in adulthood with sickle cell disease. Front Cardiovasc Med 2022; 9:883812. [PMID: 36277754 PMCID: PMC9579298 DOI: 10.3389/fcvm.2022.883812] [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: 02/25/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Sickle cell disease (SCD) is an inherited hemoglobinopathy disorder. The main consequence is synthesis of hemoglobin S leading to chronic hemolysis associated with morbidity. The aim of this study was to investigate Thrombin Generation Assay (TGA) to assess hypercoagulability in SCD and TGA parameters as biomarkers of vaso-occlusive crisis (VOC) risk and hospitalization within 1 year. Materials and methods We performed TGA in platelet poor plasma (PPP) with 1 pM of tissue factor and 4 μM of phospholipid-standardized concentration, in duplicate for patients and controls. We measured thrombomodulin (TM), soluble endothelial Protein C Receptor and Tissue Factor Pathway Inhibitor (TFPI). Results A total of 113 adult patients with SCD, 83 at steady state and 30 during VOC, and 25 healthy controls matched on age and gender were included. Among the 83 patients at steady state, (36 S/S-1 S/β0, 20 S/Sα3.7, and 19 S/C-7 S/β+) 28 developed a VOC within 1 year (median: 4 months [2.25–6]). We observed an increase of peak and velocity associated with a shortening of lagtime and time to peak (TTP) and no difference of endogenous thrombin potential (ETP) in patients compared to controls. TFPI (p < 0.001) and TM (p = 0.006) were significantly decreased. TGA confirmed hypercoagulability in all SCD genotypes and clinical status. The association of ETP > 1,207 nM.min and peak >228.5 nM presented a sensitivity of 73.5% and a specificity of 93.9% to predict VOC development within 1 year. Conclusion We have demonstrated a hypercoagulable state in SCD associated with chronic hemolysis. These preliminary findings suggest that TGA parameters, as ETP and peak, could be used to predict VOC development within 1 year.
Collapse
Affiliation(s)
- Guillaume Feugray
- Vascular Hemostasis Unit, CHU Rouen, Normandie Université, UNIROUEN, INSERM U1096, Rouen, France
| | | | | | - Cécile Dumesnil
- Department of Pediatric Onco-Hematology, CHU Rouen, Rouen, France
| | - Ygal Benhamou
- Department of Internal Medicine, CHU Rouen, Normandie Université, UNIROUEN, INSERM U1096, Rouen, France
| | - Valery Brunel
- Department of General Biochemistry, CHU Rouen, Rouen, France
| | - Véronique Le Cam Duchez
- Vascular Hemostasis Unit, CHU Rouen, Normandie Université, UNIROUEN, INSERM U1096, Rouen, France
| | | | - Paul Billoir
- Vascular Hemostasis Unit, CHU Rouen, Normandie Université, UNIROUEN, INSERM U1096, Rouen, France,*Correspondence: Paul Billoir, ; orcid.org/0000-0001-5632-7713
| |
Collapse
|
31
|
Heeney MM, Abboud MR, Githanga J, Inusa BPD, Kanter J, Michelson AD, Nduba V, Musiime V, Apte M, Inati A, Taksande AM, Andersson M, Åstrand M, Maklad N, Niazi M, Himmelmann A, Berggren AR. Ticagrelor vs placebo for the reduction of vaso-occlusive crises in pediatric sickle cell disease: the HESTIA3 study. Blood 2022; 140:1470-1481. [PMID: 35849650 PMCID: PMC9523370 DOI: 10.1182/blood.2021014095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 05/24/2022] [Indexed: 11/28/2022] Open
Abstract
The phase 3 HESTIA3 study assessed the efficacy and safety of the reversible P2Y12 inhibitor ticagrelor vs placebo in preventing vaso-occlusive crises in pediatric patients with sickle cell disease (SCD). Patients aged 2 to 17 years were randomly assigned 1:1 to receive weight-based doses of ticagrelor or matching placebo. The primary end point was the rate of vaso-occlusive crises, a composite of painful crises and/or acute chest syndrome (ACS). Key secondary end points included number and duration of painful crises, number of ACS events, and number of vaso-occlusive crises requiring hospitalization or emergency department visits. Exploratory end points included the effect of ticagrelor on platelet activation. In total, 193 patients (ticagrelor, n = 101; placebo, n = 92) underwent randomization at 53 sites across 16 countries. The study was terminated 4 months before planned completion for lack of efficacy. Median ticagrelor exposure duration was 296.5 days. The primary end point was not met: estimated yearly incidence of vaso-occlusive crises was 2.74 in the ticagrelor group and 2.60 in the placebo group (rate ratio, 1.06; 95% confidence interval, 0.75-1.50; P = .7597). There was no evidence of efficacy for ticagrelor vs placebo across secondary end points. Median platelet inhibition with ticagrelor at 6 months was 34.9% predose and 55.7% at 2 hours' postdose. Nine patients (9%) in the ticagrelor group and eight patients (9%) in the placebo group had at least one bleeding event. In conclusion, no reduction of vaso-occlusive crises was seen with ticagrelor vs placebo in these pediatric patients with SCD. This trial was registered at www.clinicaltrials.gov as #NCT03615924.
Collapse
Affiliation(s)
- Matthew M Heeney
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Miguel R Abboud
- American University of Beirut Medical Center, Beirut, Lebanon
| | - Jessie Githanga
- Hematology and Blood Transfusion Unit, Department of Human Pathology, University of Nairobi, Nairobi, Kenya
| | - Baba P D Inusa
- Paediatric Haematology, Evelina London Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Julie Kanter
- Hematology-Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Alan D Michelson
- Center for Platelet Research Studies, Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Videlis Nduba
- Kenya Medical Research Institute, Center for Respiratory Diseases Research, Nairobi, Kenya
| | - Victor Musiime
- Department of Pediatrics and Child Health, College of Health Sciences, Makerere University, Kampala, Uganda
- Joint Clinical Research Centre, Kampala, Uganda
| | - Mohini Apte
- Department of Pediatrics, Government Medical College Nagpur, Nagpur, India
| | - Adlette Inati
- School of Medicine, Lebanese American University, Byblos and Nini Hospital, Tripoli, Lebanon
| | - Amar M Taksande
- Department of Pediatrics, Jawaharlal Nehru Medical College, Sawangi (Meghe), Wardha, Maharashta, India
| | - Marielle Andersson
- AstraZeneca BioPharmaceuticals R&D, Late-stage Development, Cardiovascular, Renal and Metabolic, Gothenburg, Sweden
| | - Magnus Åstrand
- AstraZeneca BioPharmaceuticals R&D, Clinical Pharmacology and Safety Sciences, Clinical Pharmacology and Quantitative Pharmacology, Cardiovascular, Renal and Metabolic, Gothenburg, Sweden; and
| | - Noha Maklad
- AstraZeneca BioPharmaceuticals R&D, Late-stage Development, Cardiovascular, Renal and Metabolic, Gaithersburg, MD
| | - Mohammad Niazi
- AstraZeneca BioPharmaceuticals R&D, Clinical Pharmacology and Safety Sciences, Clinical Pharmacology and Quantitative Pharmacology, Cardiovascular, Renal and Metabolic, Gothenburg, Sweden; and
| | - Anders Himmelmann
- AstraZeneca BioPharmaceuticals R&D, Late-stage Development, Cardiovascular, Renal and Metabolic, Gothenburg, Sweden
| | - Anders R Berggren
- AstraZeneca BioPharmaceuticals R&D, Late-stage Development, Cardiovascular, Renal and Metabolic, Gothenburg, Sweden
| |
Collapse
|
32
|
Smith WR, McClish DK, Bovbjerg VE, Singh HK. Development and validation of the sickle cell stress scale-adult. Eur J Haematol 2022; 109:215-225. [PMID: 35585659 PMCID: PMC9531901 DOI: 10.1111/ejh.13789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 11/28/2022]
Abstract
Disease-specific stress can partly explain Sickle Cell Disease (SCD) healthcare utilization. We developed and validated two measures of adult SCD-specific stress for research and clinical care. A large cohort of adults with SCD completed both the 3-item Likert-scale adapted from a previous disease stress measure and a 10-item Likert-scale questionnaire drafted specifically to measure SCD stress. They concurrently completed a psychosocial and health-related quality of life scale battery, then subsequently daily pain diaries. Diaires measured: daily intensity, distress and interference of pain; self-defined vaso-occlusive crises (VOC), opioid use, and types of healthcare utilization for up to 24 weeks. Analyses tested Cronbach's alpha, correlation of the three-item and 10-item stress scales with the concurrent battery, with percentages of pain days, VOC days, opioid use days, and healthcare utilization days, and correlation of baseline stress and 6-month stress for the 10-item scale. Cronbach's alpha was high for both the 3-item (0.73) and 10-item (0.83) SCD stress scales, test-retest correlation of 0.55, expected correlation with the concurrent battery, and correlation with diary-measured healthcare utilization over 6 months. The correlations with the 3-item scale were stronger, but only statistically significant for depression-anxiety. The correlation between the two stress scales was 0.59. Both the 3-item and the 10-item stress scales exhibited good face, construct, concurrent, and predictive validity as well as moderate test-retest reliability. Further scale validation should determine population norms and response to interventions.
Collapse
Grants
- U10 HL083732 NHLBI NIH HHS
- U54 HL090516 NHLBI NIH HHS
- R01 HL064122 NHLBI NIH HHS
- R18 HL112737 NHLBI NIH HHS
- Pain in Sickle Cell Epidemiology Study, 1 R01 HL 64122, National Heart, Lung, and Blood Institute
- A.D. Williams Faculty Research Grant, Virginia Commonwealth University
- Virginia Basic and Translational Research Program in Sickle Cell Disease. 1U54HL090516, National Heart, Lung, and Blood Institute
- A.D. Williams Student Research Fellowships, Virgin ia Commonwealth University
- Enhancing Use of Hydroxyurea In Sickle Cell Disease Using Patient Navigators
- Clinical Research Training Program, National Institutes of Health
- Sickle Cell Disease Clinical Research Network, National Heart, Lung, and Blood Institute 1U10HL083732
- Robert Wood Johnson Foundation Generalist Physician Faculty Scholar Award (1993-1997)
- VCU School of Medicine Student Fellowships
- Enhancing Use of Hydroxyurea In Sickle Cell Disease Using Patient Navigators (NCT02197845, 1 R18 HL 112737, National Heart, Lung, and Blood Institute
Collapse
Affiliation(s)
- Wally R. Smith
- Section of Research, Division of General Internal MedicineVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Donna K. McClish
- Department of BiostatisticsVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Viktor E. Bovbjerg
- College of Health and Human SciencesOregon State UniversityCorvallisOregonUSA
| | - Harjot K. Singh
- Division of Infectious DiseasesWeill Cornell MedicineNew YorkNew YorkUSA
| |
Collapse
|
33
|
Hounkpe BW, Moraes CRP, Lanaro C, Santos MNN, Costa FF, De Paula EV. Evaluation of the mechanisms of heme-induced tissue factor activation: Contribution of innate immune pathways. Exp Biol Med (Maywood) 2022; 247:1542-1547. [PMID: 35775605 PMCID: PMC9554166 DOI: 10.1177/15353702221106475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hemolytic diseases such as Sickle Cell Disease (SCD) are characterized by a natural propensity for both arterial and venous thrombosis. The ability of heme to induce tissue factor (TF) activation has been shown both in animal models of SCD, and in human endothelial cells and monocytes. Moreover, it was recently demonstrated that heme can induce coagulation activation in the whole blood of healthy volunteers in a TF-dependent fashion. Herein, we aim to further explore the cellular mechanisms by which heme induces TF-coagulation activation, using human mononuclear cells, which have been shown to be relevant to in vivo hemostasis. TF mRNA expression was evaluated by qPCR and TF procoagulant activity was evaluated using a 2-stage assay based on the generation of activated factor X (FXa). Heme was capable of inducing both TF expression and activation in a TLR4-dependent pathway. This activity was further amplified after TNF-α-priming. Our results provide additional details on the mechanisms by which heme is involved in the pathogenesis of hypercoagulability in hemolytic diseases.
Collapse
Affiliation(s)
| | | | - Carolina Lanaro
- Hematology and Hemotherapy Center, University of Campinas, CEP 13083-970 Campinas, Brazil
| | | | - Fernando Ferreira Costa
- School of Medical Sciences, University of Campinas, CEP 13083-894 Campinas, Brazil,Hematology and Hemotherapy Center, University of Campinas, CEP 13083-970 Campinas, Brazil
| | - Erich Vinicius De Paula
- School of Medical Sciences, University of Campinas, CEP 13083-894 Campinas, Brazil,Hematology and Hemotherapy Center, University of Campinas, CEP 13083-970 Campinas, Brazil,Erich Vinicius De Paula.
| |
Collapse
|
34
|
Verma A, Huffman JE, Gao L, Minnier J, Wu WC, Cho K, Ho YL, Gorman BR, Pyarajan S, Rajeevan N, Garcon H, Joseph J, McGeary JE, Suzuki A, Reaven PD, Wan ES, Lynch JA, Petersen JM, Meigs JB, Freiberg MS, Gatsby E, Lynch KE, Zekavat SM, Natarajan P, Dalal S, Jhala DN, Arjomandi M, Bonomo RA, Thompson TK, Pathak GA, Zhou JJ, Donskey CJ, Madduri RK, Wells QS, Gelernter J, Huang RDL, Polimanti R, Chang KM, Liao KP, Tsao PS, Sun YV, Wilson PWF, O’Donnell CJ, Hung AM, Gaziano JM, Hauger RL, Iyengar SK, Luoh SW. Association of Kidney Comorbidities and Acute Kidney Failure With Unfavorable Outcomes After COVID-19 in Individuals With the Sickle Cell Trait. JAMA Intern Med 2022; 182:796-804. [PMID: 35759254 PMCID: PMC9237798 DOI: 10.1001/jamainternmed.2022.2141] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Importance Sickle cell trait (SCT), defined as the presence of 1 hemoglobin beta sickle allele (rs334-T) and 1 normal beta allele, is prevalent in millions of people in the US, particularly in individuals of African and Hispanic ancestry. However, the association of SCT with COVID-19 is unclear. Objective To assess the association of SCT with the prepandemic health conditions in participants of the Million Veteran Program (MVP) and to assess the severity and sequelae of COVID-19. Design, Setting, and Participants COVID-19 clinical data include 2729 persons with SCT, of whom 353 had COVID-19, and 129 848 SCT-negative individuals, of whom 13 488 had COVID-19. Associations between SCT and COVID-19 outcomes were examined using firth regression. Analyses were performed by ancestry and adjusted for sex, age, age squared, and ancestral principal components to account for population stratification. Data for the study were collected between March 2020 and February 2021. Exposures The hemoglobin beta S (HbS) allele (rs334-T). Main Outcomes and Measures This study evaluated 4 COVID-19 outcomes derived from the World Health Organization severity scale and phenotypes derived from International Classification of Diseases codes in the electronic health records. Results Of the 132 577 MVP participants with COVID-19 data, mean (SD) age at the index date was 64.8 (13.1) years. Sickle cell trait was present in 7.8% of individuals of African ancestry and associated with a history of chronic kidney disease, diabetic kidney disease, hypertensive kidney disease, pulmonary embolism, and cerebrovascular disease. Among the 4 clinical outcomes of COVID-19, SCT was associated with an increased COVID-19 mortality in individuals of African ancestry (n = 3749; odds ratio, 1.77; 95% CI, 1.13 to 2.77; P = .01). In the 60 days following COVID-19, SCT was associated with an increased incidence of acute kidney failure. A counterfactual mediation framework estimated that on average, 20.7% (95% CI, -3.8% to 56.0%) of the total effect of SCT on COVID-19 fatalities was due to acute kidney failure. Conclusions and Relevance In this genetic association study, SCT was associated with preexisting kidney comorbidities, increased COVID-19 mortality, and kidney morbidity.
Collapse
Affiliation(s)
- Anurag Verma
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania
- Perelman School of Medicine, Department of Medicine, University of Pennsylvania, Philadelphia
| | | | - Lina Gao
- Knight Cancer Institute, Biostatistics Shared Resource, Oregon Health & Science University, Portland
- VA Portland Health Care System, Portland, Oregon
| | - Jessica Minnier
- VA Portland Health Care System, Portland, Oregon
- OHSU-PSU School of Public Health, Oregon Health & Science University, Portland
- Knight Cancer Institute, Biostatistics Shared Resource, Oregon Health & Science University, Portland
| | - Wen-Chih Wu
- Department of Medicine, Cardiology, Providence VA Healthcare System, Providence, Rhode Island
- Alpert Medical School & School of Public Health, Brown University, Providence, Rhode Island
| | - Kelly Cho
- MAVERIC, VA Boston Healthcare System, Boston, Massachusetts
- Medicine, Aging, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yuk-Lam Ho
- MAVERIC, VA Boston Healthcare System, Boston, Massachusetts
| | | | - Saiju Pyarajan
- VA Boston Healthcare System, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Nallakkandi Rajeevan
- Yale Center for Medical Informatics, Yale School of Medicine, New Haven, Connecticut
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven
| | - Helene Garcon
- MAVERIC, VA Boston Healthcare System, Boston, Massachusetts
| | - Jacob Joseph
- Department of Medicine, VA Boston Healthcare System, Boston, Massachusetts
- Brigham & Women’s Hospital, Boston, Massachusetts
| | - John E. McGeary
- Department of Psychiatry and Human Behavior, Providence VA Medical Center, Providence, Rhode Island
- Brown University Medical School, Providence, Rhode Island
| | - Ayako Suzuki
- Department of Medicine, Gastroenterology, Durham VA Medical Center, Durham, North Carolina
- Department of Medicine, Gastroenterology, Duke University, Durham, North Carolina
| | - Peter D. Reaven
- Department of Medicine, Phoenix VA Healthcare System, Phoenix, Arizona
- University of Arizona, Phoenix
| | - Emily S. Wan
- Department of Medicine, Pulmonary, Critical Care, Sleep, and Allergy Section, VA Boston Healthcare System, Boston, Massachusetts
- Channing Division of Network Medicine, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Julie A. Lynch
- VA Informatics & Computing Infrastructure, VA Salt Lake City Utah & University of Utah, School of Medicine, Salt Lake City
| | - Jeffrey M. Petersen
- Pathology and Laboratory Medicine, Corporal Michael Crescenz VA Medical Center, Philadelphia, Pennsylvania
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - James B. Meigs
- Medicine, General Internal Medicine, Massachusetts General Hospital, Boston
| | | | - Elise Gatsby
- VA Informatics and Computing Infrastructure (VINCI), VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Kristine E. Lynch
- VA Informatics and Computing Infrastructure (VINCI), VA Salt Lake City Healthcare System, Salt Lake City, Utah
- Internal Medicine, Epidemiology, University of Utah School of Medicine, Salt Lake City
| | - Seyedeh Maryam Zekavat
- Computational Biology & Bioinformatics, Yale School of Medicine, New Haven, Connecticut
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Pradeep Natarajan
- Program in Medical and Population Genetics, Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Clinical Data Science Research Group, ORD, Portland VA Medical Center, Portland, Oregon
| | - Sharvari Dalal
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Pathology and Laboratory Medicine, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania
| | - Darshana N. Jhala
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Pathology and Laboratory Medicine, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania
| | - Mehrdad Arjomandi
- Medicine, Pulmonary and Critical Care, San Francisco VA Healthcare System, San Francisco, California
- University of California San Francisco
| | - Robert A. Bonomo
- Cleveland VA Medical Center, Cleveland, Ohio
- Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | | | - Gita A. Pathak
- Department of Psychiatry, Division of Human Genetics, Yale School of Medicine, New Haven, Connecticut
- VA Connecticut Healthcare System, West Haven
| | - Jin J. Zhou
- Medicine, University of California, Los Angeles
- Epidemiology and Biostatistics, University of Arizona, Phoenix
| | - Curtis J. Donskey
- Infectious Disease Section, Louis Stokes Cleveland VA, Cleveland, Ohio
- Case Western Reserve University, Cleveland, Ohio
| | - Ravi K. Madduri
- Data Science and Learning, Argonne National Laboratory, Lemont, Illinois
| | - Quinn S. Wells
- Departments of Medicine, Biomedical Informatics, and Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joel Gelernter
- VA Connecticut Healthcare System, West Haven
- Psychiatry, Human Genetics, Yale University School of Medicine, West Haven, Connecticut
| | | | - Renato Polimanti
- Departments of Medicine, Biomedical Informatics, and Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
- Psychiatry, Human Genetics, Yale University School of Medicine, West Haven, Connecticut
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Katherine P. Liao
- Medicine, Rheumatology, VA Boston Healthcare System, Boston, Massachusetts
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine & Biomedical Informatics, Harvard Medical School, Boston, Massachusetts
| | - Philip S. Tsao
- Precision Medicine, VA Palo Alto Health Care System, Palo Alto, California
| | - Yan V. Sun
- Epidemiology, Emory University School of Public Health, Atlanta, Georgia
- Atlanta VA Health Care System, Decatur, Georgia
| | - Peter W. F. Wilson
- Atlanta VA Health Care System, Decatur, Georgia
- Emory University School of Medicine, Atlanta, Georgia
| | | | - Adriana M. Hung
- Vanderbilt University Medical Center, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, Tennessee
| | - J. Michael Gaziano
- VA Boston Health Care System, Boston, Massachusetts
- Medicine, Harvard Medical School, Boston, Massachusetts
| | - Richard L. Hauger
- Center of Excellence for Stress & Mental Health, VA San Diego Healthcare System, San Diego, California
- Center for Behavioral Genetics of Aging, University of California, San Diego, La Jolla
| | - Sudha K. Iyengar
- Departments of Population and Quantitative Health Sciences, Ophthalmology and Visual Sciences and Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
- Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio
| | - Shiuh-Wen Luoh
- VA Portland Health Care System, Portland, Oregon
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland
| |
Collapse
|
35
|
Torres DG, Paes J, da Costa AG, Malheiro A, Silva GV, Mourão LPDS, Tarragô AM. JAK2 Variant Signaling: Genetic, Hematologic and Immune Implication in Chronic Myeloproliferative Neoplasms. Biomolecules 2022; 12:291. [PMID: 35204792 PMCID: PMC8961666 DOI: 10.3390/biom12020291] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 02/04/2023] Open
Abstract
The JAK2V617F variant constitutes a genetic alteration of higher frequency in BCR/ABL1 negative chronic myeloproliferative neoplasms, which is caused by a substitution of a G ˃ T at position 1849 and results in the substitution of valine with phenylalanine at codon 617 of the polypeptide chain. Clinical, morphological and molecular genetic features define the diagnosis criteria of polycythemia vera, essential thrombocythemia and primary myelofibrosis. Currently, JAK2V617F is associated with clonal hematopoiesis, genomic instability, dysregulations in hemostasis and immune response. JAK2V617F clones induce an inflammatory immune response and lead to a process of immunothrombosis. Recent research has shown great interest in trying to understand the mechanisms associated with JAK2V617F signaling and activation of cellular and molecular responses that progressively contribute to the development of inflammatory and vascular conditions in association with chronic myeloproliferative neoplasms. Thus, the aim of this review is to describe the main genetic, hematological and immunological findings that are linked to JAK2 variant signaling in chronic myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Dania G. Torres
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
| | - Jhemerson Paes
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
| | - Allyson G. da Costa
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus 69067-005, AM, Brazil
| | - Adriana Malheiro
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus 69067-005, AM, Brazil
| | - George V. Silva
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
- Fundação Oswaldo Cruz–Instituto Leônidas e Maria Deane (Fiocruz), Manaus 69027-070, AM, Brazil
- Fundação Centro de Controle de Oncologia do Amazonas (FCECON), Manaus 69040-010, AM, Brazil
| | - Lucivana P. de Souza Mourão
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
| | - Andréa M. Tarragô
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (FHEMOAM), Manaus 69050-001, AM, Brazil
| |
Collapse
|
36
|
Rankine-Mullings AE. Ulcerative colitis in patients with sickle cell disease: a rare but important co-morbidity. Paediatr Int Child Health 2022; 42:1-4. [PMID: 35694868 DOI: 10.1080/20469047.2022.2084964] [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
COVID-19: Coronavirus disease 2019; HIC: high-income countries; IBD: inflammatory bowel disease; LMIC: low- and middle-income countries; PUCAL: paediatric ulcerative colitis activity index; SCD: sickle cell disease; UC: ulcerative colitis.
Collapse
Affiliation(s)
- Angela E Rankine-Mullings
- Sickle Cell Unit, Caribbean Institute for Health Research, The University of the West Indies, Kingston, Jamaica
| |
Collapse
|
37
|
Rozi W, Ali EAH, Al-Mashdali AF, Abdelrazek M, Yassin MA. Use of rivaroxaban in sickle cell disease and venous thromboembolism: A case report. Medicine (Baltimore) 2021; 100:e27988. [PMID: 34941038 PMCID: PMC8702265 DOI: 10.1097/md.0000000000027988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/07/2021] [Accepted: 11/11/2021] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION Sickle cell disease (SCD) is a hematological disorder characterized by sickling of red blood cells. Patients are at increased risk of venous thromboembolism. There are no guidelines for the management of venous thromboembolism in sickle cell disease specifically in terms of the anticoagulant of choice. PATIENT CONCERNS Here, we report a case of a 30-year-old lady with past medical history of sickle cell disease who came with chest pain and shortness of breath. DIAGNOSIS We found that she has bilateral pulmonary embolism (PE). INTERVENTION She was started on rivaroxaban. OUTCOME The patient was followed for 18 months, she did not suffer from recurrence of PE, and she did not develop any complications related to rivaroxaban. CONCLUSION We concluded that rivaroxaban is effective in treating PE in sicklers and also it is safe.
Collapse
Affiliation(s)
- Waail Rozi
- Internal Medicine, Hamad Medical Corporation, Doha, Qatar
| | | | | | | | | |
Collapse
|
38
|
Annarapu GK, Nolfi-Donegan D, Reynolds M, Wang Y, Kohut L, Zuckerbraun B, Shiva S. Heme stimulates platelet mitochondrial oxidant production to induce targeted granule secretion. Redox Biol 2021; 48:102205. [PMID: 34891098 PMCID: PMC8661700 DOI: 10.1016/j.redox.2021.102205] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/24/2021] [Accepted: 12/04/2021] [Indexed: 10/19/2022] Open
Abstract
Hemolysis, a pathological component of many diseases, is associated with thrombosis and vascular dysfunction. Hemolytic products, including cell-free hemoglobin and free heme directly activate platelets. However, the effect of hemolysis on platelet degranulation, a central process in not only thrombosis, but also inflammatory and mitogenic signaling, remains less clear. Our group showed that hemoglobin-induced platelet activation involved the production of mitochondrial reactive oxygen species (mtROS). However, the molecular mechanism by which extracellular hemolysis induces platelet mtROS production, and whether these mtROS regulate platelet degranulation remains unknown. Here, we demonstrate using isolated human platelets that cell free heme is a more potent agonist for platelet activation than hemoglobin, and stimulates the release of a specific set of molecules, including the glycoprotein thrombospondin-1 (TSP-1), from the α-granule of platelets. We uncover the mechanism of heme-mediated platelet mtROS production which is dependent on the activation of platelet toll-like receptor 4 (TLR4) signaling and leads to the downstream phosphorylation and inhibition of complex-V by the serine kinase Akt. Notably, inhibition of platelet TLR4 or Akt, or scavenging of mtROS prevents heme-induced granule release in vitro. Further, heme-dependent granule release is significantly attenuated in vivo in mice lacking TLR4 or those treated with the mtROS scavenger MitoTEMPO. These data elucidate a novel mechanism of TLR4-mediated mitochondrial regulation, establish the mechanistic link between hemolysis and platelet degranulation, and begin to define the heme and mtROS-dependent platelet secretome. These data have implications for hemolysis-induced thrombo-inflammatory signaling and for the consideration of platelet mitochondria as a therapeutic target in hemolytic disorders.
Collapse
Affiliation(s)
- Gowtham K Annarapu
- Pittsburgh Heart, Lung, Blood, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Deirdre Nolfi-Donegan
- Pittsburgh Heart, Lung, Blood, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA; Department of Pediatrics, Division of Hematology/Oncology, Children's Hospital of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Michael Reynolds
- Pittsburgh Heart, Lung, Blood, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Yinna Wang
- Pittsburgh Heart, Lung, Blood, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Lauryn Kohut
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Brian Zuckerbraun
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
39
|
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
|
40
|
Silva CM, de Souza Medina S, de Melo Campos P, Costa FF, Saad STO, Benites BD. Platelet counts on peripheral blood and Mean Platelet Volume as markers of clinical severity in Sickle Cell Disease. Blood Cells Mol Dis 2021; 91:102592. [PMID: 34273711 DOI: 10.1016/j.bcmd.2021.102592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/25/2021] [Accepted: 06/25/2021] [Indexed: 11/29/2022]
|
41
|
Ellsworth P, Little JA. Sevuparin trial for acute pain in sickle cell disease: the dog that did not bark. LANCET HAEMATOLOGY 2021; 8:e307-e309. [PMID: 33894164 DOI: 10.1016/s2352-3026(21)00100-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 10/21/2022]
Affiliation(s)
| | - Jane A Little
- UNC Blood Research Center, Chapel Hill, NC 27599, USA
| |
Collapse
|
42
|
Vogel S, Kamimura S, Arora T, Smith ML, Almeida LEF, Combs CA, Thein SL, Quezado ZMN. NLRP3 inflammasome and bruton tyrosine kinase inhibition interferes with upregulated platelet aggregation and in vitro thrombus formation in sickle cell mice. Biochem Biophys Res Commun 2021; 555:196-201. [PMID: 33831782 DOI: 10.1016/j.bbrc.2021.03.115] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 03/21/2021] [Indexed: 12/20/2022]
Abstract
The nucleotide-binding domain leucine-rich repeat containing protein 3 (NLRP3) inflammasome is a critical inflammatory mechanism identified in platelets, which controls platelet activation and aggregation. We have recently shown that the platelet NLRP3 inflammasome is upregulated in sickle cell disease (SCD), which is mediated by Bruton tyrosine kinase (BTK). Here, we investigated the effect of pharmacological inhibition of NLRP3 and BTK on platelet aggregation and the formation of in vitro thrombi in Townes SCD mice. Mice were injected for 4 weeks with the NLRP3 inhibitor MCC950, the BTK inhibitor ibrutinib or vehicle control. NLRP3 activity, as monitored by caspase-1 activation, was upregulated in platelets from SCD mice, which was dependent on BTK. Large areas of platelet aggregates detected in the liver of SCD mice were decreased when mice were treated with MCC950 or ibrutinib. Moreover, platelet aggregation and in vitro thrombus formation were upregulated in SCD mice and were inhibited when mice were subjected to pharmacological inhibition of NLRP3 and BTK. Targeting the NLRP3 inflammasome might be a novel approach for antiplatelet therapy in SCD.
Collapse
Affiliation(s)
- Sebastian Vogel
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Sayuri Kamimura
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Taruna Arora
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Meghann L Smith
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Luis E F Almeida
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Christian A Combs
- Light Microscopy Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zenaide M N Quezado
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA; Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
43
|
Conran N, Embury SH. Sickle cell vaso-occlusion: The dialectic between red cells and white cells. Exp Biol Med (Maywood) 2021; 246:1458-1472. [PMID: 33794696 DOI: 10.1177/15353702211005392] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The pathophysiology of sickle cell anemia, a hereditary hemoglobinopathy, has fascinated clinicians and scientists alike since its description over 100 years ago. A single gene mutation in the HBB gene results in the production of abnormal hemoglobin (Hb) S, whose polymerization when deoxygenated alters the physiochemical properties of red blood cells, in turn triggering pan-cellular activation and pathological mechanisms that include hemolysis, vaso-occlusion, and ischemia-reperfusion to result in the varied and severe complications of the disease. Now widely regarded as an inflammatory disease, in recent years attention has included the role of leukocytes in vaso-occlusive processes in view of the part that these cells play in innate immune processes, their inherent ability to adhere to the endothelium when activated, and their sheer physical and potentially obstructive size. Here, we consider the role of sickle red blood cell populations in elucidating the importance of adhesion vis-a-vis polymerization in vaso-occlusion, review the direct adhesion of sickle red cells to the endothelium in vaso-occlusive processes, and discuss how red cell- and leukocyte-centered mechanisms are not mutually exclusive. Given the initial clinical success of crizanlizumab, a specific anti-P selectin therapy, we suggest that it is appropriate to take a holistic approach to understanding and exploring the complexity of vaso-occlusive mechanisms and the adhesive roles of the varied cell types, including endothelial cells, platelets, leukocytes, and red blood cells.
Collapse
Affiliation(s)
- Nicola Conran
- Hematology Center, University of Campinas-UNICAMP, Barão Geraldo 13083-8, Campinas, SP, Brazil
| | | |
Collapse
|
44
|
Silva-Junior AL, Garcia NP, Cardoso EC, Dias S, Tarragô AM, Fraiji NA, Gomes MS, Amaral LR, Teixeira-Carvalho A, Martins-Filho OA, De Paula EV, Costa AG, Malheiro A. Immunological Hallmarks of Inflammatory Status in Vaso-Occlusive Crisis of Sickle Cell Anemia Patients. Front Immunol 2021; 12:559925. [PMID: 33776989 PMCID: PMC7990896 DOI: 10.3389/fimmu.2021.559925] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
Sickle Cell Anemia (SCA) is the most common genetic disorder around the world. The mutation in the β-globin gene is responsible for a higher hemolysis rate, with further involvement of immunological molecules, especially cytokines, chemokines, growth factors, and anaphylatoxins. These molecules are responsible for inducing and attracting immune cells into circulation, thus contributing to increases in leukocytes and other pro-inflammatory mediators, and can culminate in a vaso-occlusive crisis (VOC). This study aimed to characterize the levels of these molecules in SCA patients in different clinical conditions in order to identify potential hallmarks of inflammation in these patients. An analytical prospective study was conducted using the serum of SCA patients in steady-state (StSt; n = 27) and VOC (n = 22), along with 53 healthy donors (HD). Samples from the VOC group were obtained on admission and on discharge, in the convalescent phase (CV). Levels of chemokines (CXCL8, CXCL10, CL2, CLL3, CCL4, CL5, and CCL11), cytokines (IL-1β, IL-1ra, IL-2, IL-4, IL-5, IL-6, IL-7, IL-10, IL-12p70, IL-13, IL-17A, TNF-α, and IFN-γ) and growth factors (VEGF, FGFb, PDGF-BB, GM-CSF, and G-CSF) were measured using a Luminex assay, and anaphylatoxins (C3a, C4a, and C5a) were measured using Cytometric Bead Array. SCA patients in StSt showed a pro-inflammatory profile, and were indicated as being higher producers of CCL2, IL-1β, IL-12p70, IFN-γ, IL-17A, and GM-CSF, while VOC is highlighted by molecules IL-4 and IL-5, but also IL-2, IL-7, PDGF-BB, and G-CSF. PDGF-BB and IL-1ra seemed to be two important hallmarks for the acute-to-chronic stage, due to their significant decrease after crisis inflammation and statistical difference in VOC and CV groups. These molecules show higher levels and a strong correlation with other molecules in VOC. Furthermore, they remain at higher levels even after crisis recovery, which suggest their importance in the role of inflammation during crisis and participation in immune cell adhesion and activation. These results support a relevant role of cytokines, neutrophil and monocytes, since these may act as markers of VOC inflammation in SCA patients.
Collapse
Affiliation(s)
- Alexander Leonardo Silva-Junior
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Nadja Pinto Garcia
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Evilázio Cunha Cardoso
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Stephanny Dias
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Andrea Monteiro Tarragô
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Nelson Abrahim Fraiji
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Matheus Souza Gomes
- Laboratório de Bioinformática e Análises Moleculares, Rede Multidisciplinar de Pesquisa, Ciência e Tecnologia, Universidade Federal de Uberlândia, Patos de Minas, Brazil
| | - Laurence Rodrigues Amaral
- Laboratório de Bioinformática e Análises Moleculares, Rede Multidisciplinar de Pesquisa, Ciência e Tecnologia, Universidade Federal de Uberlândia, Patos de Minas, Brazil
| | - Andréa Teixeira-Carvalho
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Grupo Integrado de Pesquisas em Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
| | - Olindo Assis Martins-Filho
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Grupo Integrado de Pesquisas em Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
| | - Erich Vinicius De Paula
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Escola de Ciências Médicas, Universidade de Campinas, Campinas, Brazil
| | - Allyson Guimarães Costa
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
- Escola de Enfermagem de Manaus, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Adriana Malheiro
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
| |
Collapse
|
45
|
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: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
46
|
Hounkpe BW, Chenou F, Domingos IDF, Cardoso EC, Costa Sobreira MJDV, Araujo AS, Lucena‐Araújo AR, da Silva Neto PV, Malheiro A, Fraiji NA, Costa FF, Bezerra MAC, Santos MNN, De Paula EV. Neutrophil extracellular trap regulators in sickle cell disease: Modulation of gene expression of PADI4, neutrophil elastase, and myeloperoxidase during vaso-occlusive crisis. Res Pract Thromb Haemost 2021; 5:204-210. [PMID: 33537545 PMCID: PMC7845058 DOI: 10.1002/rth2.12463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 10/28/2020] [Accepted: 10/31/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Recent evidence suggests that generation of neutrophil extracellular traps (NETosis), one of the components of immunothrombosis, is associated with the pathogenesis of both venous thromboembolism and sickle cell disease (SCD). NETosis is a complex process regulated by several proteins such as peptidyl arginine deaminase 4 (PADI4), neutrophil elastase (ELANE), and myeloperoxidase (MPO). Among these regulators, PADI4 is responsible of histone citrullination, an essential step for NETosis. Accordingly, its inhibition has been recently cited as a promising therapeutic strategy for diseases such as SCD. Although attractive, this strategy requires supportive evidence of its role in the pathogenesis of SCD. PATIENTS AND METHODS Patients from two independent cohorts were enrolled in this study. Samples were obtained at steady state (53 patients) or during acute episodes of vaso-occlusive crisis (VOC; 28 patients) in patients from cohort 1. mRNA was extracted from granulocytes to analyze PADI4, ELANE, and MPO expression by qPCR. Furthermore, plasma activity of PADI4 was assessed from an independent cohort in 15 patients, within 24 hours from admission for VOC. Race-matched healthy individuals from the same geographic regions were used as controls for each cohort. RESULTS AND CONCLUSIONS Higher levels of gene expression of PADI4 and ELANE were observed during VOC. Furthermore, plasma activity of PADI4 was higher in acute VOC when compared to healthy individuals. These results demonstrate that NETosis regulators are modulated during acute VOC, and pave the way for studies of PADI4 inhibition as a therapeutic strategy for acute VOC in SCD.
Collapse
Affiliation(s)
| | - Francine Chenou
- School of Medical SciencesUniversity of CampinasCampinasBrazil
| | | | | | | | - Aderson S. Araujo
- Hematology and Hemotherapy Foundation of Pernambuco ‐ HEMOPERecifeBrazil
| | | | | | - Adriana Malheiro
- Hematology and Hemotherapy Foundation from Amazonas State (HEMOAM)ManausBrazil
| | | | - Fernando Ferreira Costa
- School of Medical SciencesUniversity of CampinasCampinasBrazil
- Hematology and Hemotherapy CenterUniversity of CampinasCampinasBrazil
| | | | | | - Erich Vinicius De Paula
- School of Medical SciencesUniversity of CampinasCampinasBrazil
- Hematology and Hemotherapy CenterUniversity of CampinasCampinasBrazil
| |
Collapse
|
47
|
Santaterra VAG, Fiusa MML, Hounkpe BW, Chenou F, Tonasse WV, da Costa LNG, Garcia-Weber D, Domingos IDF, de Lima F, Borba-Junior IT, Araújo ADS, Lucena-Araújo AR, Bezerra MAC, Dos Santos MNN, Costa FF, Millán J, De Paula EV. Endothelial Barrier Integrity Is Disrupted In Vitro by Heme and by Serum From Sickle Cell Disease Patients. Front Immunol 2020; 11:535147. [PMID: 33381108 PMCID: PMC7767881 DOI: 10.3389/fimmu.2020.535147] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Free extracellular heme has been shown to activate several compartments of innate immunity, acting as a danger-associated molecular pattern (DAMP) in hemolytic diseases. Although localized endothelial barrier (EB) disruption is an important part of inflammation that allows circulating leukocytes to reach inflamed tissues, non-localized/deregulated disruption of the EB can lead to widespread microvascular hyperpermeability and secondary tissue damage. In mouse models of sickle cell disease (SCD), EB disruption has been associated with the development of a form of acute lung injury that closely resembles acute chest syndrome (ACS), and that can be elicited by acute heme infusion. Here we explored the effect of heme on EB integrity using human endothelial cell monolayers, in experimental conditions that include elements that more closely resemble in vivo conditions. EB integrity was assessed by electric cell-substrate impedance sensing in the presence of varying concentrations of heme and sera from SCD patients or healthy volunteers. Heme caused a dose-dependent decrease of the electrical resistance of cell monolayers, consistent with EB disruption, which was confirmed by staining of junction protein VE-cadherin. In addition, sera from SCD patients, but not from healthy volunteers, were also capable to induce EB disruption. Interestingly, these effects were not associated with total heme levels in serum. However, when heme was added to sera from SCD patients, but not from healthy volunteers, EB disruption could be elicited, and this effect was associated with hemopexin serum levels. Together our in vitro studies provide additional support to the concept of heme as a DAMP in hemolytic conditions.
Collapse
Affiliation(s)
| | | | | | - Francine Chenou
- School of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | - Loredana Nilkenes Gomes da Costa
- School of Medical Sciences, University of Campinas, Campinas, Brazil.,Department of Biomedicine, Federal University of Piaui, Parnaiba, Brazil
| | - Diego Garcia-Weber
- Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas, Universidad Autonoma de Madrid, Madrid, Spain
| | - Igor de Farias Domingos
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Brazil.,Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Franciele de Lima
- School of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | - Aderson da Silva Araújo
- Department of Internal Medicine, Hematology and Hemotherapy Foundation of Pernambuco (HEMOPE), Recife, Brazil
| | | | | | | | - Fernando Ferreira Costa
- School of Medical Sciences, University of Campinas, Campinas, Brazil.,Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | - Jaime Millán
- Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas, Universidad Autonoma de Madrid, Madrid, Spain
| | - Erich Vinicius De Paula
- School of Medical Sciences, University of Campinas, Campinas, Brazil.,Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| |
Collapse
|
48
|
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
|
49
|
CXCR4 hi effector neutrophils in sickle cell anemia: potential role for elevated circulating serotonin (5-HT) in CXCR4 hi neutrophil polarization. Sci Rep 2020; 10:14262. [PMID: 32868775 PMCID: PMC7459317 DOI: 10.1038/s41598-020-71078-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/07/2020] [Indexed: 01/01/2023] Open
Abstract
Leukocyte recruitment and heterocellular aggregate formation drive the inflammatory vaso-occlusive processes associated with sickle cell anemia (SCA). We characterized neutrophils in a population of patients with SCA and investigated whether platelet-derived molecules can induce phenotypic alterations in this cell type. Imaging flow cytometry analysis demonstrated that the frequency of circulating CXCR4hi neutrophils was significantly higher in steady-state SCA individuals than in healthy control individuals and that these cells presented increased CD11b activation and toll-like receptor-4 expression. SCA neutrophils display increased neutrophil-platelet aggregation, and CXCR4hi neutrophils demonstrated augmented neutrophil-platelet aggregate frequency with a higher mean number of platelets adhered per neutrophil. Importantly, incubation of neutrophils with platelets significantly elevated their CXCR4 expression, while SCA plasma was found to induce CXCR4hi neutrophil polarization significantly more than control plasma. SCA individuals had significantly increased plasma levels of serotonin (5-HT), and serotonin molecule and SCA plasma induced neutrophil CXCR4 expression in a serotonin-receptor-dependent manner. Thus, the augmented CXCR4hi neutrophil population may contribute to mechanisms that promote vaso-occlusion in SCA; furthermore, circulating serotonin, derived from platelet activation, may play a role in the polarization of neutrophils, suggesting that serotonin-receptor antagonists or serotonin reuptake inhibitors could represent therapeutic approaches to reduce neutrophil activation in SCA.
Collapse
|
50
|
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
|