1
|
Chauhan W, Ferdowsi S, Sudharshan SJ, Zennadi R. Rpl13a snoRNAs-regulated NADPH oxidase 1-dependent ROS generation: A novel RBC pathway mediating complement C3a deposition and triggering thrombosis in aging and venous blood clotting disorders. Free Radic Biol Med 2025; 230:138-150. [PMID: 39938620 PMCID: PMC11936428 DOI: 10.1016/j.freeradbiomed.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/30/2025] [Accepted: 02/08/2025] [Indexed: 02/14/2025]
Abstract
Adults older than 45 years old are at higher risk of developing venous blood clotting known as venous thrombosis/thromboembolism than a cohort <45 years old. Complement activation, which can be mediated by oxidative stress, plays a central role in venous thrombosis. Yet, whether RBCs contribute to complement activation triggering thrombosis in aging and in patients with venous thrombosis/thromboembolism remains an open question. RBCs from healthy Mid-life stage (55-68 years old) adults and patients with venous thrombosis/thromboembolism showed higher deposition of the complement C3 and the anaphylatoxin C3a, and NADPH oxidase (Nox)1 expression than a younger cohort (21-30 years old). Increased C3/C3a deposition on RBCs from mid-life stage adults and patients with venous thrombosis/thromboembolism triggered prothrombin activation via Nox1-dependent reactive oxygen species (ROS) generation, and G protein-coupled receptor kinase 2 (GRK2) activation. Interaction of C3/C3a positive RBCs from mid-life stage adults with endothelial cells led to increased endothelial ROS production. TGF-β1-stimulated GRK2 and Nox1 activation in RBCs from the younger and older adults exacerbated RBC C3/C3a deposition and C3/C3a-mediated prothrombotic activation, which appears to result from ROS-mediated increased RBC phosphatidylserine exposure. Using human RBCs, and Rpl13a snoRNA knockout aged mice, we show that Rpl13a snoRNAs, the master regulators of ROS levels and oxidative stress response, regulate human and murine RBC C3a deposition and prothrombic activation in aging by modulating Nox1 mRNA expression. In vivo Rpl13a snoRNA knockout in aged mice decreased thrombi size by blunting RBC C3a deposition, and RBCs-triggering prothrombin activation. These findings point out to a novel role of RBC Rpl13a snoRNAs in dysregulating RBC ROS-induced C3a deposition promoting venous thrombosis in aging.
Collapse
Affiliation(s)
- Waseem Chauhan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Shirin Ferdowsi
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - S J Sudharshan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Rahima Zennadi
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
2
|
Tozatto‐Maio K, Rós FA, Weinlich R, Rocha V. Inflammatory pathways and anti-inflammatory therapies in sickle cell disease. Hemasphere 2024; 8:e70032. [PMID: 39698332 PMCID: PMC11655128 DOI: 10.1002/hem3.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 07/12/2024] [Accepted: 09/17/2024] [Indexed: 12/20/2024] Open
Abstract
Sickle cell disease (SCD) is a monogenic disease, resulting from a single-point mutation, that presents a complex pathophysiology and high clinical heterogeneity. Inflammation stands as a prominent characteristic of SCD. Over the past few decades, the role of different cells and molecules in the regulation of the inflammatory process has been elucidated. In conjunction with the polymerization of hemoglobin S (HbS), intravascular hemolysis, which releases free heme, HbS, and hemoglobin-related damage-associated molecular patterns, initiates multiple inflammatory pathways that are not yet fully comprehended. These complex phenomena lead to a vicious cycle that perpetuates vaso-occlusion, hemolysis, and inflammation. To date, few inflammatory biomarkers can predict disease complications; conversely, there is a plethora of therapies that reduce inflammation in SCD, although clinical outcomes vary widely. Importantly, whether the clinical heterogeneity and complications are related to the degree of inflammation is not known. This review aims to further our understanding of the roles of main immune cells, and other inflammatory factors, as potential prognostic biomarkers for predicting clinical outcomes or identifying novel treatments for SCD.
Collapse
Affiliation(s)
- Karina Tozatto‐Maio
- Centro de Ensino e PesquisaHospital Israelita Albert EinsteinSão PauloBrazil
- Divisão de Hematologia, Hemoterapia e Terapia CelularHospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão PauloBrazil
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco‐Immuno‐Hematology (LIM‐31), Department of Hematology and Cell TherapyHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
| | - Felipe A. Rós
- Divisão de Hematologia, Hemoterapia e Terapia CelularHospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão PauloBrazil
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco‐Immuno‐Hematology (LIM‐31), Department of Hematology and Cell TherapyHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
| | - Ricardo Weinlich
- Centro de Ensino e PesquisaHospital Israelita Albert EinsteinSão PauloBrazil
| | - Vanderson Rocha
- Divisão de Hematologia, Hemoterapia e Terapia CelularHospital das Clínicas da Faculdade de Medicina da Universidade de São PauloSão PauloBrazil
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco‐Immuno‐Hematology (LIM‐31), Department of Hematology and Cell TherapyHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
- Instituto D'Or de Ensino e Pesquisa, Rede D'OrSao PauloBrazil
- Department of Hematology, Churchill HospitalUniversity of OxfordOxfordUK
| |
Collapse
|
3
|
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
|
4
|
Frostegård A, Haegerstrand A. New Therapeutic Strategies in Retinal Vascular Diseases: A Lipid Target, Phosphatidylserine, and Annexin A5-A Future Theranostic Pairing in Ophthalmology. Pharmaceuticals (Basel) 2024; 17:979. [PMID: 39204083 PMCID: PMC11357257 DOI: 10.3390/ph17080979] [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: 05/28/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 09/03/2024] Open
Abstract
Despite progress in the management of patients with retinal vascular and degenerative diseases, there is still an unmet clinical need for safe and effective therapeutic options with novel mechanisms of action. Recent mechanistic insights into the pathogenesis of retinal diseases with a prominent vascular component, such as retinal vein occlusion (RVO), diabetic retinopathy (DR) and wet age-related macular degeneration (AMD), may open up new treatment paradigms that reach beyond the inhibition of vascular endothelial growth factor (VEGF). Phosphatidylserine (PS) is a novel lipid target that is linked to the pathophysiology of several human diseases, including retinal diseases. PS acts upstream of VEGF and complement signaling pathways. Annexin A5 is a protein that targets PS and inhibits PS signaling. This review explores the current understanding of the potential roles of PS as a target and Annexin A5 as a therapeutic. The clinical development status of Annexin A5 as a therapeutic and the potential utility of PS-Annexin A5 as a theranostic pairing in retinal vascular conditions in particular is described.
Collapse
Affiliation(s)
- Anna Frostegård
- Annexin Pharmaceuticals AB, Kammakargatan 48, S-111 60 Stockholm, Sweden
- Unit of Immunology and Chronic Disease, IMM, Karolinska Institute, S-171 77 Stockholm, Sweden
| | | |
Collapse
|
5
|
Evangelidis P, Evangelidis N, Vlachaki E, Gavriilaki E. What is the role of complement in bystander hemolysis? Old concept, new insights. Expert Rev Hematol 2024; 17:107-116. [PMID: 38708453 DOI: 10.1080/17474086.2024.2348662] [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: 01/07/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
INTRODUCTION Bystander hemolysis occurs when antigen-negative red blood cells (RBCs) are lysed by the complement system. Many clinical entities including passenger lymphocyte syndrome, hyperhemolysis following blood transfusion, and paroxysmal nocturnal hemoglobinuria are complicated by bystander hemolysis. AREAS COVERED The review provides data about the role of the complement system in the pathogenesis of bystander hemolysis. Moreover, future perspectives on the understanding and management of this syndrome are described. EXPERT OPINION Complement system can be activated via classical, alternative, and lectin pathways. Classical pathway activation is mediated by antigen-antibody (autoantibodies and alloantibodies against autologous RBCs, infectious agents) complexes. Alternative pathway initiation is triggered by heme, RBC microvesicles, and endothelial injury that is a result of intravascular hemolysis. Thus, C5b is formed, binds with C6-C9 compomers, and MAC (C5b-9) is formulated in bystander RBCs membranes, leading to cell lysis. Intravascular hemolysis, results in activation of the alternative pathway, establishing a vicious cycle between complement activation and bystander hemolysis. C5 inhibitors have been used effectively in patients with hyperhemolysis syndrome and other entities characterized by bystander hemolysis.
Collapse
Affiliation(s)
- Paschalis Evangelidis
- Second Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Evangelidis
- Second Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Efthymia Vlachaki
- Adult Thalassemia Unit, 2nd Department of Internal Medicine, Aristotle University of Thessaloniki, Hippocration General Hospital, Thessaloniki, Greece
| | - Eleni Gavriilaki
- Second Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
6
|
Meuleman MS, Roumenina LT, Grunenwald A. Complement involvement in sickle cell disease. Presse Med 2023; 52:104205. [PMID: 37972851 DOI: 10.1016/j.lpm.2023.104205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023] Open
Abstract
Sickle Cell Disease (SCD) is a hereditary blood disorder characterized by the presence of abnormal hemoglobin, leading to the formation of sickle-shaped red blood cells, causing vaso-occlusion. Inflammation is a key component of the pathophysiology of SCD, contributing to the vascular complications and tissue damage. This review is centered on exploring the role of the inflammatory complement system in the pathophysiology of SCD. Our goal is to offer a comprehensive summary of the existing evidence regarding complement activation in patients with SCD, encompassing both steady-state conditions and episodes of vaso-occlusive events. Additionally, we will discuss the proposed mechanisms by which the complement system may contribute to tissue injury in this pathology. Finally, we will provide an overview of the available evidence concerning the effectiveness of therapeutic interventions aimed at blocking the complement system in the context of SCD and discuss the perspective of complement inhibition.
Collapse
Affiliation(s)
- Marie-Sophie Meuleman
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer team, Paris, France
| | - Lubka T Roumenina
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer team, Paris, France.
| | - Anne Grunenwald
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer team, Paris, France; CHI de Poissy-St Germain en Laye, Service de néphrologie - hémodialyse, Poissy, France.
| |
Collapse
|
7
|
Ivy ZK, Belcher JD, Khasabova IA, Chen C, Juliette JP, Abdulla F, Ruan C, Allen K, Nguyen J, Rogness VM, Beckman JD, Khasabov SG, Gupta K, Taylor RP, Simone DA, Vercellotti GM. Cold exposure induces vaso-occlusion and pain in sickle mice that depend on complement activation. Blood 2023; 142:1918-1927. [PMID: 37774369 PMCID: PMC10731576 DOI: 10.1182/blood.2022019282] [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] [Received: 12/05/2022] [Revised: 08/30/2023] [Accepted: 09/18/2023] [Indexed: 10/01/2023] Open
Abstract
Vaso-occlusive pain episodes (VOE) cause severe pain in patients with sickle cell disease (SCD). Vaso-occlusive events promote ischemia/reperfusion pathobiology that activates complement. We hypothesized that complement activation is linked to VOE. We used cold to induce VOE in the Townes sickle homozygous for hemoglobin S (HbSS) mouse model and complement inhibitors to determine whether anaphylatoxin C5a mediates VOE. We used a dorsal skinfold chamber to measure microvascular stasis (vaso-occlusion) and von Frey filaments applied to the plantar surface of the hind paw to assess mechanical hyperalgesia in HbSS and control Townes mice homozygous for hemoglobin A (HbAA) mice after cold exposure at 10°C/50°F for 1 hour. Cold exposure induced more vaso-occlusion in nonhyperalgesic HbSS mice (33%) than in HbAA mice (11%) or HbSS mice left at room temperature (1%). Cold exposure also produced mechanical hyperalgesia as measured by paw withdrawal threshold in HbSS mice compared with that in HbAA mice or HbSS mice left at room temperature. Vaso-occlusion and hyperalgesia were associated with an increase in complement activation fragments Bb and C5a in plasma of HbSS mice after cold exposure. This was accompanied by an increase in proinflammatory NF-κB activation and VCAM-1 and ICAM-1 expression in the liver. Pretreatment of nonhyperalgesic HbSS mice before cold exposure with anti-C5 or anti-C5aR monoclonal antibodies (mAbs) decreased vaso-occlusion, mechanical hyperalgesia, complement activation, and liver inflammatory markers compared with pretreatment with control mAb. Anti-C5 or -C5aR mAb infusion also abrogated mechanical hyperalgesia in HbSS mice with ongoing hyperalgesia at baseline. These findings suggest that C5a promotes vaso-occlusion, pain, and inflammation during VOE and may play a role in chronic pain.
Collapse
Affiliation(s)
- Zalaya K. Ivy
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - John D. Belcher
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Iryna A. Khasabova
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, MN
| | - Chunsheng Chen
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Joseph P. Juliette
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, MN
| | - Fuad Abdulla
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Conglin Ruan
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Kaje Allen
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, MN
| | - Julia Nguyen
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Victoria M. Rogness
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, MN
| | - Joan D. Beckman
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Sergey G. Khasabov
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, MN
| | - Kalpna Gupta
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, CA
| | - Ronald P. Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA
| | - Donald A. Simone
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, MN
| | - Gregory M. Vercellotti
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| |
Collapse
|
8
|
Aboderin FI, Oduola T, Davison GM, Oguntibeju OO. A Review of the Relationship between the Immune Response, Inflammation, Oxidative Stress, and the Pathogenesis of Sickle Cell Anaemia. Biomedicines 2023; 11:2413. [PMID: 37760854 PMCID: PMC10525295 DOI: 10.3390/biomedicines11092413] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/09/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
Sickle cell anaemia (SCD) is a life-threatening haematological disorder which is predominant in sub-Saharan Africa and is triggered by a genetic mutation of the β-chain haemoglobin gene resulting in the substitution of glutamic acid with valine. This mutation leads to the production of an abnormal haemoglobin molecule called haemoglobin S (HbS). When deoxygenated, haemoglobin S (HbS) polymerises and results in a sickle-shaped red blood cell which is rigid and has a significantly shortened life span. Various reports have shown a strong link between oxidative stress, inflammation, the immune response, and the pathogenesis of sickle cell disease. The consequence of these processes leads to the development of vasculopathy (disease of the blood vessels) and several other complications. The role of the immune system, particularly the innate immune system, in the pathogenesis of SCD has become increasingly clear in recent years of research; however, little is known about the roles of the adaptive immune system in this disease. This review examines the interaction between the immune system, inflammation, oxidative stress, blood transfusion, and their effects on the pathogenesis of sickle cell anaemia.
Collapse
Affiliation(s)
- Florence Ifechukwude Aboderin
- Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville 7535, South Africa;
| | - Taofeeq Oduola
- Department of Chemical Pathology, Usmanu Danfodiyo University, Sokoto 840004, Nigeria;
| | - Glenda Mary Davison
- SAMRC/CPUT Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville 7535, South Africa;
| | - Oluwafemi Omoniyi Oguntibeju
- Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville 7535, South Africa;
| |
Collapse
|
9
|
Green A, Jones H, Nero A, Ibrahim IF, Sarode R, Scheid LM, Webb CB, Adkins BD, Yates SG. A case of hyperhemolysis syndrome in sickle cell disease and concomitant COVID-19. Transfus Apher Sci 2023; 62:103712. [PMID: 37149492 PMCID: PMC10101481 DOI: 10.1016/j.transci.2023.103712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/05/2023] [Accepted: 04/12/2023] [Indexed: 05/08/2023]
Abstract
BACKGROUND Hyperhemolysis syndrome (HHS) is an uncommon transfusion reaction described in several hematologic disorders, including sickle cell disease (SCD). HHS is characterized by a decline in hemoglobin (Hb) values below pre-transfusion levels following transfusion of red blood cells (RBCs), coupled with laboratory markers consistent with hemolysis. The proposed pathophysiologic mechanisms underlying HHS include increased phosphatidylserine expression, macrophage activation, and complement dysregulation. Many pathophysiologic mechanisms thought to contribute to HHS have been similarly described in cases of severe COVID-19. CASE REPORT A 28-year-old male with a history of HbSS presented with shortness of breath, right-sided chest pain, and a two-day history of fever. Polymerase chain reaction (PCR) detected SARS-CoV-2 infection with the omicron variant. The patient required an RBC transfusion (pre-transfusion hemoglobin [Hb]5.8 g/dL) with an immediate post-transfusion Hb of 6.3 g/dL. However, Hb rapidly declined to 1.7 g/dL, and lactate dehydrogenase (LDH) rose to 8701 u/L. The absolute reticulocyte count of 538 × 109/L correspondingly fell to 29 × 109/L. Despite additional RBC transfusions and initiation of immunosuppressive therapy, he expired on Day 9(D9). CONCLUSION Given the similarities in their proposed pathophysiology, patients with SCD and concomitant SARS-CoV-2 infection may be predisposed to developing HHS.
Collapse
Affiliation(s)
- Allen Green
- Department of Pathology, Division of Transfusion Medicine and Hemostasis, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Heather Jones
- Department of Pathology, Division of Transfusion Medicine and Hemostasis, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alecia Nero
- Department of Internal Medicine, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ibrahim F Ibrahim
- Department of Internal Medicine, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ravi Sarode
- Department of Pathology, Division of Transfusion Medicine and Hemostasis, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lisa M Scheid
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christopher B Webb
- Department of Pathology, Division of Transfusion Medicine and Hemostasis, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Brian D Adkins
- Department of Pathology, Division of Transfusion Medicine and Hemostasis, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sean G Yates
- Department of Pathology, Division of Transfusion Medicine and Hemostasis, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
10
|
Wu M, Fields JJ, Sachdev V, Belcik JT, Chen J, Reed F, Fu X, Hodovan J, Harmann LM, Swistara G, Lindner JR. Increased Susceptibility for Adverse Reactions to Ultrasound Enhancing Agents in Sickle Cell Disease. J Am Soc Echocardiogr 2023; 36:208-215. [PMID: 36113741 DOI: 10.1016/j.echo.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/28/2022] [Accepted: 09/03/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pain-related adverse events (AEs) to ultrasound enhancing agents (UEAs) have been reported in patients with sickle cell disease (SCD). The aims of this study were to characterize the scope of these AEs in the SCD population and to investigate potential mechanisms on the basis of pathways involved in SCD vaso-occlusive crisis (VOC) and pain. METHODS The prevalence and classification of AEs were analyzed from two clinical trials in which high-dose Definity infusions were used in patients with SCD (n = 55) or matched control subjects (n = 43) to study muscle or myocardial microvascular perfusion. Because complement (C') activation can trigger VOC in SCD, C' activation and surface adhesion of C' proteins on lipid UEAs were studied in vitro. C'-mediated UEA attachment to bone marrow immune cells was assessed using flow cytometry in a murine SCD model (Townes mice). Blood from patients receiving Definity was obtained to measure specific lysophospholipid metabolites of lipids in Definity thought to mediate SCD pain. RESULTS Moderate or greater AEs, all of which were nociceptive (back or bone pain), occurred in one control subject and nine SCD subjects (2% vs 16%, P = .02). Patients with SCD who had AEs tended to have more severe manifestations of SCD. Three of the subjects with SCD had previously received Definity without complications. In patients with SCD, four AEs were classified as severe in intensity and as serious AEs on the basis of need for medical intervention. AEs were described to be similar to SCD-related pain, but there was no evidence for VOC, hemolysis, hypotension, or hypoxemia. At baseline, markers of C' activation were greater in patients with SCD than control subjects. However, after administration of lipid UEAs, SCD and control subjects were similar with regard to C' activation response, anaphylatoxin production, bone marrow microbubble retention, and production of lysophospholipids. There was a trend toward increased deposition of C3b and C3bi on lipid UEAs exposed to serum from patients with SCD. CONCLUSIONS Patients with SCD are particularly susceptible to nociceptive AEs when given Definity at high doses. The mechanism for these AEs remains unclear but most are not related to the triggering of classic VOC.
Collapse
Affiliation(s)
- Melinda Wu
- Doernbecher Children's Hospital and Pape Research Center, Oregon Health & Science University, Portland, Oregon
| | - Joshua J Fields
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - J Todd Belcik
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Junmei Chen
- Bloodworks Research Institute, Seattle, Washington
| | | | - Xiaoyun Fu
- Bloodworks Research Institute, Seattle, Washington
| | - James Hodovan
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Leanne M Harmann
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Gabriella Swistara
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
11
|
Moghimi SM, Haroon HB, Yaghmur A, Simberg D, Trohopoulos PN. Nanometer- and angstrom-scale characteristics that modulate complement responses to nanoparticles. J Control Release 2022; 351:432-443. [PMID: 36152807 PMCID: PMC10200249 DOI: 10.1016/j.jconrel.2022.09.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/15/2022] [Accepted: 09/18/2022] [Indexed: 11/26/2022]
Abstract
The contribution of the complement system to non-specific host defence and maintenance of homeostasis is well appreciated. Many particulate systems trigger complement activation but the underlying mechanisms are still poorly understood. Activation of the complement cascade could lead to particle opsonisation by the cleavage products of the third complement protein and might promote inflammatory reactions. Antibody binding in a controlled manner and/or sensing of particles by the complement pattern-recognition molecules such as C1q and mannose-binding lectin can trigger complement activation. Particle curvature and spacing arrangement/periodicity of surface functional groups/ligands are two important parameters that modulate complement responses through multivalent engagement with and conformational regulation of surface-bound antibodies and complement pattern-recognition molecules. Thus, a better fundamental understanding of nanometer- and angstrom-scale parameters that modulate particle interaction with antibodies and complement proteins could portend new possibilities for engineering of particulate drug carriers and biomedical platforms with tuneable complement responses and is discussed here.
Collapse
Affiliation(s)
- S Moein Moghimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Center, Aurora, CO, USA.
| | - Hajira B Haroon
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Anan Yaghmur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen Ø, Denmark
| | - Dmitri Simberg
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Center, Aurora, CO, USA; Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | | |
Collapse
|
12
|
Gadó K, Khodier M, Virág A, Domján G, Dörnyei G. Anemia of geriatric patients. Physiol Int 2022; 109:119-134. [DOI: 10.1556/2060.2022.00218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/14/2022] [Accepted: 02/22/2022] [Indexed: 11/19/2022]
Abstract
Abstract
Anemia is a common finding in the elderly. Approximately 10 percent of the elderly suffers from anemia. Anemia per se is an independent factor of mortality in older patients regardless its cause. Frailty is also frequent in geriatric patients. That means that there is a decreased reserve capacity to react to different stress factors including anemia. The frequent presence of heart failure and also impaired cerebrovascular circulation makes more difficult to tolerate anemia in older age.
Anemia is a symptom, finding and treating the underlying cause is also important.
Treatment always depends on clinical findings: the more severe the symptoms, the more important to treat them. Severity of anemia depends not only the underlying cause, degree of anemia, co-morbidities and frailty of the patients, but also the speed of its development. Sudden blood loss due to an accident is less well tolerated than the same degree of anemia due to B12 deficiency.
Main causes of anemia in the elderly include nutritional deficiencies, chronic diseases, tumors, and certain hematological malignancies such as chronic lymphocytic leukemia, multiple myeloma, myelodysplastic syndrome.
Collapse
Affiliation(s)
- Klara Gadó
- Department of Clinical Studies, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
- Department of Geriatrics and Center of Nursing Sciences, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Malaz Khodier
- Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Andrea Virág
- Department of Geriatrics and Center of Nursing Sciences, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Gyula Domján
- Department of Clinical Studies, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Gabriella Dörnyei
- Department of Morphology and Physiotherapy, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| |
Collapse
|
13
|
Hendrickson JE. Management of hemolytic transfusion reactions. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2021; 2021:704-709. [PMID: 34889404 PMCID: PMC8791106 DOI: 10.1182/hematology.2021000308] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Delayed hemolytic transfusion reactions (DHTRs) in patients with sickle cell disease are underappreciated and potentially fatal. Patients with DHTRs typically have symptoms of pain or dark urine days to weeks following a red blood cell (RBC) transfusion. In instances of DHTRs with hyperhemolysis, the patient's hemoglobin (Hgb) may be significantly lower than it was pretransfusion, and the Hgb A may drop by more than 50%. In most cases, at least 1 RBC alloantibody and sometimes multiple RBC alloantibodies can be identified during the DHTR, with those antibodies presumably having fallen below the level of detection at the time of the implicated transfusion. However, in up to one-third of cases, no new RBC alloantibodies can be identified posttransfusion. Complement is increasingly being appreciated to play a role in DHTRs and hyperhemolysis, not only due to classic pathway activation (with complement fixed antibody bound to RBCs) but also due to alternative pathway activation (resulting in part from plasma free heme). As such, anti-C5 inhibition has recently been reported to be effective at mitigating hemolysis in the setting of some severe DHTRs. Transfusion avoidance during DHTRs is recommended if possible, with long-term transfusion support advice being less clear; for example, a history of a severe DHTR may lead to questions regarding the safety of transfusions prior to curative therapies such as stem cell transplantation or gene therapy. A better understanding of antibody-positive and antibody-negative DHTRs, including patient- or disease-specific risk factors, is necessary to improve transfusion safety.
Collapse
Affiliation(s)
- Jeanne E. Hendrickson
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
- Correspondence Jeanne E. Hendrickson, Yale University Department of Laboratory Medicine, 330 Cedar Street, Clinic Building 405, PO Box 208035, New Haven, CT 06520-0835; e-mail:
| |
Collapse
|
14
|
Rausch L, Lutz K, Schifferer M, Winheim E, Gruber R, Oesterhaus EF, Rinke L, Hellmuth JC, Scherer C, Muenchhoff M, Mandel C, Bergwelt‐Baildon M, Simons M, Straub T, Krug AB, Kranich J, Brocker T. Binding of phosphatidylserine-positive microparticles by PBMCs classifies disease severity in COVID-19 patients. J Extracell Vesicles 2021; 10:e12173. [PMID: 34854246 PMCID: PMC8636722 DOI: 10.1002/jev2.12173] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/02/2021] [Indexed: 01/08/2023] Open
Abstract
Infection with SARS-CoV-2 is associated with thromboinflammation, involving thrombotic and inflammatory responses, in many COVID-19 patients. In addition, immune dysfunction occurs in patients characterised by T cell exhaustion and severe lymphopenia. We investigated the distribution of phosphatidylserine (PS), a marker of dying cells, activated platelets and platelet-derived microparticles (PMP), during the clinical course of COVID-19. We found an unexpectedly high amount of blood cells loaded with PS+ PMPs for weeks after the initial COVID-19 diagnosis. Elevated frequencies of PS+ PMP+ PBMCs correlated strongly with increasing disease severity. As a marker, PS outperformed established laboratory markers for inflammation, leucocyte composition and coagulation, currently used for COVID-19 clinical scoring. PS+ PMPs preferentially bound to CD8+ T cells with gene expression signatures of proliferating effector rather than memory T cells. As PS+ PMPs carried programmed death-ligand 1 (PD-L1), they may affect T cell expansion or function. Our data provide a novel marker for disease severity and show that PS, which can trigger the blood coagulation cascade, the complement system, and inflammation, resides on activated immune cells. Therefore, PS may serve as a beacon to attract thromboinflammatory processes towards lymphocytes and cause immune dysfunction in COVID-19.
Collapse
Affiliation(s)
- Lisa Rausch
- Institute for ImmunologyBiomedical Center (BMC), Faculty of Medicine, LMU MunichMunichGermany
| | - Konstantin Lutz
- Institute for ImmunologyBiomedical Center (BMC), Faculty of Medicine, LMU MunichMunichGermany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Munich Cluster of Systems Neurology (Synergy)MunichGermany
| | - Elena Winheim
- Institute for ImmunologyBiomedical Center (BMC), Faculty of Medicine, LMU MunichMunichGermany
| | - Rudi Gruber
- bene pharmaChem GmbH & Co.KG., GeretsriedGermany
| | - Elina F. Oesterhaus
- COVID‐19 Registry of the LMU Munich (CORKUM)University Hospital, LMU MunichMunichGermany
- Max von Pettenkofer Institute & Gene CenterVirologyNational Reference Center for RetrovirusesLMU MünchenMunichGermany
| | - Linus Rinke
- Institute for ImmunologyBiomedical Center (BMC), Faculty of Medicine, LMU MunichMunichGermany
| | - Johannes C. Hellmuth
- COVID‐19 Registry of the LMU Munich (CORKUM)University Hospital, LMU MunichMunichGermany
- Department of Medicine IIIUniversity Hospital, LMU MunichMunichGermany
| | - Clemens Scherer
- COVID‐19 Registry of the LMU Munich (CORKUM)University Hospital, LMU MunichMunichGermany
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
| | - Maximilian Muenchhoff
- COVID‐19 Registry of the LMU Munich (CORKUM)University Hospital, LMU MunichMunichGermany
- Max von Pettenkofer Institute & Gene CenterVirologyNational Reference Center for RetrovirusesLMU MünchenMunichGermany
- German Center for Infection Research (DZIF)partner site MunichGermany
| | - Christopher Mandel
- COVID‐19 Registry of the LMU Munich (CORKUM)University Hospital, LMU MunichMunichGermany
- Department of Medicine IVUniversity Hospital, LMU MunichMunichGermany
| | - Michael Bergwelt‐Baildon
- COVID‐19 Registry of the LMU Munich (CORKUM)University Hospital, LMU MunichMunichGermany
- Department of Medicine IIIUniversity Hospital, LMU MunichMunichGermany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Munich Cluster of Systems Neurology (Synergy)MunichGermany
- Institute of Neuronal Cell BiologyTechnical University of MunichMunichGermany
| | - Tobias Straub
- Core facility BioinformaticsBiomedical Center (BMC)Faculty of MedicineLMU MunichMunichGermany
| | - Anne B. Krug
- Institute for ImmunologyBiomedical Center (BMC), Faculty of Medicine, LMU MunichMunichGermany
| | - Jan Kranich
- Institute for ImmunologyBiomedical Center (BMC), Faculty of Medicine, LMU MunichMunichGermany
| | - Thomas Brocker
- Institute for ImmunologyBiomedical Center (BMC), Faculty of Medicine, LMU MunichMunichGermany
| |
Collapse
|
15
|
Davidson BP, Hodovan J, Layoun ME, Golwala H, Zahr F, Lindner JR. Echocardiographic Ischemic Memory Molecular Imaging for Point-of-Care Detection of Myocardial Ischemia. J Am Coll Cardiol 2021; 78:1990-2000. [PMID: 34763776 DOI: 10.1016/j.jacc.2021.08.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Noninvasive molecular imaging of recent ischemia can potentially be used to diagnose acute coronary syndrome (ACS) with high accuracy. OBJECTIVES The authors hypothesized that bedside myocardial contrast echocardiography (MCE) ischemic memory imaging could be achieved with phosphatidylserine microbubbles (MBPS) that are retained in the microcirculation via ischemia-associated endothelial activation. METHODS A dose-finding study was performed in healthy volunteers (n = 17) to establish optimal MBPS dosing. Stable patients with ACS (n = 30) and confirmed antecedent but resolved myocardial ischemia were studied within 2 hours of coronary angiography and percutaneous coronary intervention (PCI) when indicated. MCE molecular imaging was performed 8 minutes after intravenous administration of MBPS. MCE perfusion imaging was used to assess the status of the postischemic microcirculation. RESULTS Based on dose-finding studies, 0.10 or 0.15 mL of MBPS based on body mass was selected. In patients with ACS, all but 2 underwent primary PCI. MCE molecular imaging signal intensity was greater in the postischemic risk area vs remote territory (median [95% CI]: 56 [33-66] vs 8 [2-17] IU; P < 0.001) with a receiver-operating characteristic curve C-statistic of 0.94 to differentiate post-ischemic from remote territory. Molecular imaging signal in the risk area was not related to type of ACS (unstable angina: 3; non-ST-segment elevation myocardial infarction: 14; ST-segment elevation myocardial infarction: 13), peak troponin, time to PCI, post-PCI myocardial perfusion, GRACE (Global Registry of Acute Coronary Events) score, or HEART score. CONCLUSIONS Molecular imaging with point-of-care echocardiography and MBPS can detect recent but resolved myocardial ischemia. This bedside technique requires only minutes to perform and appears independent of the degree of ischemia. (Ischemic Memory Imaging With Myocardial Contrast Echocardiography; NCT03009266).
Collapse
Affiliation(s)
- Brian P Davidson
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - James Hodovan
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Michael E Layoun
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Harsh Golwala
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Firas Zahr
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA; Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA.
| |
Collapse
|
16
|
Hebbel RP, Vercellotti GM. Multiple inducers of endothelial NOS (eNOS) dysfunction in sickle cell disease. Am J Hematol 2021; 96:1505-1517. [PMID: 34331722 PMCID: PMC9292023 DOI: 10.1002/ajh.26308] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/26/2022]
Abstract
A characteristic aspect of the robust, systemic inflammatory state in sickle cell disease is dysfunction of endothelial nitric oxide synthase (eNOS). We identify 10 aberrant endothelial cell inputs, present in the specific sickle context, that are known to have the ability to cause eNOS dysfunction. These are: endothelial arginase depletion, asymmetric dimethylarginine, complement activation, endothelial glycocalyx degradation, free fatty acids, inflammatory mediators, microparticles, oxidized low density lipoproteins, reactive oxygen species, and Toll‐like receptor 4 signaling ligands. The effect of true eNOS dysfunction on clinical testing using flow‐mediated dilation can be simulated by two known examples of endothelial dysfunction mimicry (hemoglobin consumption of NO; and oxidation of smooth muscle cell soluble guanylate cyclase). This lends ambiguity to interpretation of such clinical testing. The presence of these multiple perturbing factors argues that a therapeutic approach targeting only a single injurious endothelial input (or either example of mimicry) would not be sufficiently efficacious. This would seem to argue for identifying therapeutics that directly protect eNOS function or application of multiple therapeutic approaches.
Collapse
Affiliation(s)
- Robert P. Hebbel
- Division of Hematology‐Oncology‐Transplantation, Department of Medicine University of Minnesota Medical School Minneapolis Minnesota USA
| | - Gregory M. Vercellotti
- Division of Hematology‐Oncology‐Transplantation, Department of Medicine University of Minnesota Medical School Minneapolis Minnesota USA
| |
Collapse
|
17
|
Abstract
The complement cascade is an evolutionary ancient innate immune defense system, playing a major role in the defense against infections. Its function in maintaining host homeostasis on activated cells has been emphasized by the crucial role of its overactivation in ever growing number of diseases, such as atypical hemolytic uremic syndrome (aHUS), autoimmune diseases as systemic lupus erythematosus (SLE), C3 glomerulopathies (C3GN), age-related macular degeneration (AMD), graft rejection, Alzheimer disease, and cancer, to name just a few. The last decade of research on complement has extended its implication in many pathological processes, offering new insights to potential therapeutic targets and asserting the necessity of reliable, sensitive, specific, accurate, and reproducible biomarkers to decipher complement role in pathology. We need to evaluate accurately which pathway or role should be targeted pharmacologically, and optimize treatment efficacy versus toxicity. This chapter is an introduction to the role of complement in human diseases and the use of complement-related biomarkers in the clinical practice. It is a part of a book intending to give reliable and standardized methods to evaluate complement according to nowadays needs and knowledge.
Collapse
|
18
|
Cannas G, Dubreuil L, Fichez A, Gerfaud-Valentin M, Debard AL, Hot A. Delayed Severe Hemolytic Transfusion Reaction During Pregnancy in a Woman with β-Thalassemia Intermediate: Successful Outcome After Eculizumab Administration. AMERICAN JOURNAL OF CASE REPORTS 2021; 22:e931107. [PMID: 33983909 PMCID: PMC8130975 DOI: 10.12659/ajcr.931107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 04/13/2021] [Accepted: 03/25/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Delayed hemolytic transfusion reactions (DHTR) are life-threatening complications mostly triggered by red blood cell (RBC) transfusions in patients with hemoglobinopathy. CASE REPORT We present a case of DHTR and hyperhemolysis syndrome in a 39-year-old pregnant woman with a history of ß-thalassemia intermediate in whom the hemoglobin (Hb) level fell to 27 g/L after transfusion of 2 units of crossmatch-compatible packed RBCs. No allo- or auto-antibody formation was detected. Administration of intravenous immunoglobulins and methylprednisolone followed by anti-CD20 rituximab was tried, but was unsuccessful. Infusions of eculizumab (900 mg twice at a 7-day interval) followed by another course of intravenous immunoglobulins (2 g/kg/day for 5 days) and combined with repeated erythropoietin injections (darbepoetin alpha 300 µg/week) finally allowed biological and clinical improvement. Blood counts remained controlled until delivery. Despite signs of intrauterine growth retardation, she gave birth by cesarean section at 31 weeks of pregnancy to a 1.15-kg infant. CONCLUSIONS Eculizumab seems to be of benefit in DHTR associated with hyperhemolysis and should be used early in the treatment of this pathology. Despite premature birth, our case report showed an acceptable outcome for the infant when eculizumab treatment was used during pregnancy.
Collapse
Affiliation(s)
- Giovanna Cannas
- Department of Internal Medicine, Lyon Civil Hospices, Edouard Herriot Hospital, Lyon, France
| | - Léa Dubreuil
- French Blood Establishment Auvergne-Rhône-Alpes, Croix-Rousse Hospital, Lyon, France
| | - Axel Fichez
- Department of Pregnancy Pathology, Lyon Civil Hospices, Croix-Rousse Hospital, Lyon, France
| | | | - Anne-Lise Debard
- French Blood Establishment Auvergne-Rhône-Alpes, Croix-Rousse Hospital, Lyon, France
| | - Arnaud Hot
- Department of Internal Medicine, Lyon Civil Hospices, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
19
|
Varelas C, Tampaki A, Sakellari I, Anagnostopoulos Α, Gavriilaki E, Vlachaki E. Complement in Sickle Cell Disease: Are We Ready for Prime Time? J Blood Med 2021; 12:177-187. [PMID: 33790681 PMCID: PMC8001680 DOI: 10.2147/jbm.s287301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Sickle cell disease (SCD) is a widely spread inherited hemoglobinopathy that includes a group of congenital hemolytic anemias, all characterized by the predominance of sickle hemoglobin (HbS). Its features are anemia, predisposal to bacterial infections and complications such as vaso-occlusive crisis (VOC) or delayed hemolytic transfusion reaction (DHTR), which lead to increased rate of morbidity and mortality even in the era of hydroxyurea. The interaction between sickle cells, neutrophils, platelets or endothelial cells in small vessels results in hemolysis and has been considered the disease’s main pathophysiological mechanism. Complement activation has been reported in small cohorts of SCD patients, but the governing mechanism has not been fully elucidated. This will be important to predict the patient group that would benefit from complement inhibition. Until now, eculizumab-mediated complement inhibition has shown beneficial effects in DHTR, with limited reports in patients with VOC. In the meantime, several innovative agents are under clinical development Our state-of-the-art review summarizes current data on 1) complement activation in SCD both in steady state and crisis, 2) underlying mechanisms of complement over-activation for the clinician in the context of SCD, 3) actions of hydroxyurea and new therapeutic approaches including indirect involvement in complement activation, and 4) novel paradigms in complement inhibition.
Collapse
Affiliation(s)
- Christos Varelas
- Hematology Department - BMT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| | - Athina Tampaki
- Adults Thalassemia Unit, 2nd Department of Internal Medicine, Hippokration Hospital, Thessaloniki, Greece
| | - Ioanna Sakellari
- Hematology Department - BMT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| | | | - Eleni Gavriilaki
- Hematology Department - BMT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| | - Efthymia Vlachaki
- Adults Thalassemia Unit, 2nd Department of Internal Medicine, Hippokration Hospital, Thessaloniki, Greece
| |
Collapse
|
20
|
Complement in sickle cell disease and targeted therapy: I know one thing, that I know nothing. Blood Rev 2021; 48:100805. [PMID: 33504459 DOI: 10.1016/j.blre.2021.100805] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 11/10/2020] [Accepted: 01/13/2021] [Indexed: 12/19/2022]
Abstract
Sickle cell disease (SCD) is a common inherited clinical syndrome, characterized by the presence of hemoglobin S. Anemia, susceptibility to infections and episodes of vaso-occlusive crisis (VOC) are among its features. Since SCD complications (VOC or delayed hemolytic transfusion reaction/DHTR) lead to significant morbidity and mortality, a number of studies have addressed their pathophysiology Although SCD pathophysiology has been mainly attributed to the interaction between sickle cells and neutrophils, platelets or endothelial cells in small vessels leading to hemolysis, the role of complement activation has been increasingly investigated. Importantly, complement inhibition with eculizumab has shown beneficial effects in DHTR. Given the unmet clinical need of novel therapeutics in SCD, our review summarizes current understanding of (a) complement system for the clinician, (b) complement activation in SCD both in asymptomatic state and severe clinical manifestations, (c) probable underlying mechanisms of complement activation in SCD, and (d) new therapeutic perspective of complement inhibition.
Collapse
|
21
|
Chonat S, Graciaa S, Shin HS, Newton JG, Quarmyne MO, Boudreaux J, Tang A, Zerra PE, Rollins MR, Josephson CD, Brown C, Joiner CH, Fasano RM, Stowell SR. Eculizumab for complement mediated thrombotic microangiopathy in sickle cell disease. Haematologica 2020; 105:2887-2891. [PMID: 33256394 PMCID: PMC7716365 DOI: 10.3324/haematol.2020.262006] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Satheesh Chonat
- Department of Pediatrics, Emory University School of Medicine
- Aflac Cancer and Blood Disorders Center
| | | | - H. Stella Shin
- Department of Pediatrics, Emory University School of Medicine
- Division of Pediatric Nephrology, Children’s Healthcare of Atlanta
| | - Joanna G. Newton
- Department of Pediatrics, Emory University School of Medicine
- Aflac Cancer and Blood Disorders Center
| | - Maa-Ohui Quarmyne
- Department of Pediatrics, Emory University School of Medicine
- Aflac Cancer and Blood Disorders Center
| | - Jeanne Boudreaux
- Department of Pediatrics, Emory University School of Medicine
- Aflac Cancer and Blood Disorders Center
| | - Amy Tang
- Department of Pediatrics, Emory University School of Medicine
- Aflac Cancer and Blood Disorders Center
| | - Patricia E. Zerra
- Aflac Cancer and Blood Disorders Center
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Margo R. Rollins
- Aflac Cancer and Blood Disorders Center
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Cassandra D. Josephson
- Aflac Cancer and Blood Disorders Center
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Clark Brown
- Department of Pediatrics, Emory University School of Medicine
- Aflac Cancer and Blood Disorders Center
| | - Clinton H. Joiner
- Department of Pediatrics, Emory University School of Medicine
- Aflac Cancer and Blood Disorders Center
| | - Ross M. Fasano
- Aflac Cancer and Blood Disorders Center
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Sean R. Stowell
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
22
|
Hendrickson JE. Red blood cell alloimmunization and sickle cell disease: a narrative review on antibody induction. ANNALS OF BLOOD 2020; 5:33. [PMID: 33554044 PMCID: PMC7861514 DOI: 10.21037/aob-2020-scd-01] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The high prevalence of red blood cell (RBC) alloantibodies in people with sickle cell disease (SCD) cannot be debated. Why people with SCD are so likely to form RBC alloantibodies, however, remains poorly understood. Over the past decade, a better understanding of non-ABO blood group antigen variants has emerged; RH genetic diversity and the role this diversity plays in RBC alloimmunization is discussed elsewhere. Outside of antigen variants, the immune systems of people with SCD are known to be different than those of people without SCD. Some of these differences are due to effects of free heme, whereas others are impacted by hyposplenism. Descriptive studies of differences in white blood cell (WBC) subsets, platelet counts and function, and complement activation between people with SCD and race-matched controls exist. Studies comparing the immune systems of alloimmunized people with SCD to non-alloimmunized people with SCD to race-matched controls without SCD have uncovered differences in T-cell subsets, monocytes, Fcγ receptor polymorphisms, and responses to free heme. Studies in murine models have documented the role that recipient inflammation plays in RBC alloantibody formation, with human studies reporting a similar association. Murine studies have also reported the importance of type 1 interferon (IFNα/β), known to play a pivotal role in autoimmunity, in RBC alloantibody formation. The goal of this manuscript is to review existing data on factors influencing RBC alloantibody induction in people with SCD with a focus on inflammation and other immune system considerations, from the bench to the bedside.
Collapse
Affiliation(s)
- Jeanne E. Hendrickson
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
23
|
Role of the coagulation system in the pathogenesis of sickle cell disease. Blood Adv 2020; 3:3170-3180. [PMID: 31648337 DOI: 10.1182/bloodadvances.2019000193] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 09/16/2019] [Indexed: 01/12/2023] Open
Abstract
Sickle cell disease (SCD) is an inherited monogenic red blood cell disorder affecting millions worldwide. SCD causes vascular occlusions, chronic hemolytic anemia, and cumulative organ damage such as nephropathy, pulmonary hypertension, pathologic heart remodeling, and liver necrosis. Coagulation system activation, a conspicuous feature of SCD that causes chronic inflammation, is an important component of SCD pathophysiology. The key coagulation factor, thrombin (factor IIa [FIIa]), is both a central protease in hemostasis and thrombosis and a key modifier of inflammation. Pharmacologic or genetic reduction of circulating prothrombin in Berkeley sickle mice significantly improves survival, ameliorates vascular inflammation, and results in markedly reduced end-organ damage. Accordingly, factors both upstream and downstream of thrombin, such as the tissue factor-FX complex, fibrinogen, platelets, von Willebrand factor, FXII, high-molecular-weight kininogen, etc, also play important roles in SCD pathogenesis. In this review, we discuss the various aspects of coagulation system activation and their roles in the pathophysiology of SCD.
Collapse
|
24
|
The cause and pathogenesis of hemolytic transfusion reactions in sickle-cell disease. Curr Opin Hematol 2020; 26:488-494. [PMID: 31589171 DOI: 10.1097/moh.0000000000000546] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The current review aims to summarize the epidemiology, cause, pathophysiology, and management of hemolytic transfusion reactions in sickle-cell disease (SCD). RECENT FINDINGS Patients undergoing occasional, isolated transfusions have been shown to have a higher risk of developing this condition. Despite the identification of well known risk factors, including alloimmunization, the pathophysiology of this syndrome remains unclear, as very severe forms with hyperhemolysis may develop in the absence of detectable antibodies, or with antibodies that are not considered to be clinically significant. Complement plays a crucial role in this reaction, particularly in cases of intravascular hemolysis. Complement triggers the reaction, but it also amplifies the inflammatory response and aggravates tissue damage. Free heme and hemoglobin are released and interact with complement, causing tissue damage. SUMMARY Hemolytic transfusion reactions are the most feared complications of blood transfusion in patients with SCD. This reaction is underdiagnosed because it mimics a vaso-occlusive crisis. Alloimmunization against red blood cell antigens is known to be a major trigger of this reaction, but abnormal complement activation and the underlying condition in patients with chronic hemolysis, may amplify the reaction. There is an urgent need to develop evidence-based approaches for preventing and treating this reaction.
Collapse
|
25
|
Roumenina LT, Chadebech P, Bodivit G, Vieira‐Martins P, Grunenwald A, Boudhabhay I, Poillerat V, Pakdaman S, Kiger L, Jouard A, Audureau E, Pirenne F, Galactéros F, Frémeaux‐Bacchi V, Bartolucci P. Complement activation in sickle cell disease: Dependence on cell density, hemolysis and modulation by hydroxyurea therapy. Am J Hematol 2020; 95:456-464. [PMID: 31990387 DOI: 10.1002/ajh.25742] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/14/2020] [Accepted: 01/21/2020] [Indexed: 12/15/2022]
Abstract
The complement system is an innate immune defense cascade that can cause tissue damage when inappropriately activated. Evidence for complement over activation has been reported in small cohorts of patients with sickle cell disease (SCD). However, the mechanism governing complement activation in SCD has not been elucidated. Here, we observe that the plasma concentration of sC5b-9, a reliable marker for terminal complement activation, is increased at steady state in 61% of untreated SCD patients. We show that greater complement activation in vitro is promoted by SCD erythrocytes compared to normal ones, although no significant differences were observed in the regulatory proteins CD35, CD55, and CD59 in whole blood. Complement activation is positively correlated with the percentage of dense sickle cells (DRBCs). The expression levels of CD35, CD55, and CD59 are reduced in DRBCs, suggesting inefficient regulation when cell density increases. Moreover, the surface expression of the complement regulator CD46 on granulocytes was inversely correlated with the plasma sC5b-9. We also show increased complement deposition in cultured human endothelial cells incubated with SCD serum, which is diminished by the addition of the heme scavenger hemopexin. Treatment of SCD patients with hydroxyurea produces substantial reductions in complement activation, measured by sC5b-9 concentration and upregulation of CD46, as well as decreased complement activation on RBCs in vitro. In conclusion, complement over activation is a common pathogenic event in SCD that is associated with formation of DRBCs and hemolysis. And, it affects red cells, leukocytes and endothelial cells. This complement over activation is partly alleviated by hydroxyurea therapy.
Collapse
Affiliation(s)
- Lubka T. Roumenina
- Centre de Recherche des CordeliersINSERM, Sorbonne Université, Université de Paris Paris France
| | - Philippe Chadebech
- Etablissement Français du SangIle‐de‐France, Hôpital Henri‐Mondor, AP‐HP Créteil France
- INSERM U955 équipe 2, Institut Mondor de Recherche Biomédicale (IMRB)Université Paris‐Est‐Créteil (UPEC), Créteil, France; and Laboratoire d'excellence GR‐Ex Paris France
| | - Gwellaouen Bodivit
- Etablissement Français du SangIle‐de‐France, Hôpital Henri‐Mondor, AP‐HP Créteil France
- INSERM U955 équipe 2, Institut Mondor de Recherche Biomédicale (IMRB)Université Paris‐Est‐Créteil (UPEC), Créteil, France; and Laboratoire d'excellence GR‐Ex Paris France
| | - Paula Vieira‐Martins
- Service d'Immunologie Biologique, Hôpital Européen Georges‐PompidouAssistance Publique—Hôpitaux de Paris, AP‐HP Paris France
| | - Anne Grunenwald
- Centre de Recherche des CordeliersINSERM, Sorbonne Université, Université de Paris Paris France
| | - Idris Boudhabhay
- Centre de Recherche des CordeliersINSERM, Sorbonne Université, Université de Paris Paris France
| | - Victoria Poillerat
- Centre de Recherche des CordeliersINSERM, Sorbonne Université, Université de Paris Paris France
| | - Sadaf Pakdaman
- Etablissement Français du SangIle‐de‐France, Hôpital Henri‐Mondor, AP‐HP Créteil France
- INSERM U955 équipe 2, Institut Mondor de Recherche Biomédicale (IMRB)Université Paris‐Est‐Créteil (UPEC), Créteil, France; and Laboratoire d'excellence GR‐Ex Paris France
| | - Laurent Kiger
- INSERM U955 équipe 2, Institut Mondor de Recherche Biomédicale (IMRB)Université Paris‐Est‐Créteil (UPEC), Créteil, France; and Laboratoire d'excellence GR‐Ex Paris France
| | - Alicia Jouard
- Etablissement Français du SangIle‐de‐France, Hôpital Henri‐Mondor, AP‐HP Créteil France
- INSERM U955 équipe 2, Institut Mondor de Recherche Biomédicale (IMRB)Université Paris‐Est‐Créteil (UPEC), Créteil, France; and Laboratoire d'excellence GR‐Ex Paris France
| | - Etienne Audureau
- Sickle Cell Referral Center, Service de Médecine InterneHôpital Henri‐Mondor, AP‐HP Créteil France
| | - France Pirenne
- Etablissement Français du SangIle‐de‐France, Hôpital Henri‐Mondor, AP‐HP Créteil France
- INSERM U955 équipe 2, Institut Mondor de Recherche Biomédicale (IMRB)Université Paris‐Est‐Créteil (UPEC), Créteil, France; and Laboratoire d'excellence GR‐Ex Paris France
| | - Frédéric Galactéros
- INSERM U955 équipe 2, Institut Mondor de Recherche Biomédicale (IMRB)Université Paris‐Est‐Créteil (UPEC), Créteil, France; and Laboratoire d'excellence GR‐Ex Paris France
- Service de Santé PubliqueHôpital Henri‐Mondor, AP‐HP Créteil France
| | - Véronique Frémeaux‐Bacchi
- Centre de Recherche des CordeliersINSERM, Sorbonne Université, Université de Paris Paris France
- Service d'Immunologie Biologique, Hôpital Européen Georges‐PompidouAssistance Publique—Hôpitaux de Paris, AP‐HP Paris France
| | - Pablo Bartolucci
- INSERM U955 équipe 2, Institut Mondor de Recherche Biomédicale (IMRB)Université Paris‐Est‐Créteil (UPEC), Créteil, France; and Laboratoire d'excellence GR‐Ex Paris France
- Service de Santé PubliqueHôpital Henri‐Mondor, AP‐HP Créteil France
| |
Collapse
|
26
|
Nader E, Romana M, Connes P. The Red Blood Cell-Inflammation Vicious Circle in Sickle Cell Disease. Front Immunol 2020; 11:454. [PMID: 32231672 PMCID: PMC7082402 DOI: 10.3389/fimmu.2020.00454] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/27/2020] [Indexed: 12/31/2022] Open
Abstract
Sickle cell disease (SCD) is a genetic disease caused by a single mutation in the β-globin gene, leading to the production of an abnormal hemoglobin called hemoglobin S (HbS), which polymerizes under deoxygenation, and induces the sickling of red blood cells (RBCs). Sickled RBCs are very fragile and rigid, and patients consequently become anemic and develop frequent and recurrent vaso-occlusive crises. However, it is now evident that SCD is not only a RBC rheological disease. Accumulating evidence shows that SCD is also characterized by the presence of chronic inflammation and oxidative stress, participating in the development of chronic vasculopathy and several chronic complications. The accumulation of hemoglobin and heme in the plasma, as a consequence of enhanced intravascular hemolysis, decreases nitric oxide bioavailability and enhances the production of reactive oxygen species (ROS). Heme and hemoglobin also represent erythrocytic danger-associated molecular pattern molecules (eDAMPs), which may activate endothelial inflammation through TLR-4 signaling and promote the development of complications, such as acute chest syndrome. It is also suspected that heme may activate the innate immune complement system and stimulate neutrophils to release neutrophil extracellular traps. A large amount of microparticles (MPs) from various cellular origins (platelets, RBCs, white blood cells, endothelial cells) is also released into the plasma of SCD patients and participate in the inflammation and oxidative stress in SCD. In turn, this pro-inflammatory and oxidative stress environment further alters the RBC properties. Increased pro-inflammatory cytokine concentrations promote the activation of RBC NADPH oxidase and, thus, raise the production of intra-erythrocyte ROS. Such enhanced oxidative stress causes deleterious damage to the RBC membrane and further alters the deformability of the cells, modifying their aggregation properties. These RBC rheological alterations have been shown to be associated to specific SCD complications, such as leg ulcers, priapism, and glomerulopathy. Moreover, RBCs positive for the Duffy antigen receptor for chemokines may be very sensitive to various inflammatory molecules that promote RBC dehydration and increase RBC adhesiveness to the vascular wall. In summary, SCD is characterized by a vicious circle between abnormal RBC rheology and inflammation, which modulates the clinical severity of patients.
Collapse
Affiliation(s)
- Elie Nader
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team Vascular Biology and Red Blood Cell, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| | - Marc Romana
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France.,Université des Antilles, UMR_S1134, BIGR, Pointe-à-Pitre, France.,Université de Paris, UMR_S1134, BIGR, INSERM, Paris, France
| | - Philippe Connes
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team Vascular Biology and Red Blood Cell, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| |
Collapse
|
27
|
Complement Signals Determine Opposite Effects of B Cells in Chemotherapy-Induced Immunity. Cell 2020; 180:1081-1097.e24. [PMID: 32142650 DOI: 10.1016/j.cell.2020.02.015] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/16/2019] [Accepted: 02/05/2020] [Indexed: 12/21/2022]
Abstract
Understanding molecular mechanisms that dictate B cell diversity is important for targeting B cells as anti-cancer treatment. Through the single-cell dissection of B cell heterogeneity in longitudinal samples of patients with breast cancer before and after neoadjuvant chemotherapy, we revealed that an ICOSL+ B cell subset emerges after chemotherapy. Using three immunocompetent mouse models, we recapitulated the subset switch of human tumor-infiltrating B cells during chemotherapy. By employing B-cell-specific deletion mice, we showed that ICOSL in B cells boosts anti-tumor immunity by enhancing the effector to regulatory T cell ratio. The signature of ICOSL+ B cells is imprinted by complement-CR2 signaling, which is triggered by immunogenic cell death. Moreover, we identified that CD55, a complement inhibitory protein, determines the opposite roles of B cells in chemotherapy. Collectively, we demonstrated a critical role of the B cell subset switch in chemotherapy response, which has implications in designing novel anti-cancer therapies. VIDEO ABSTRACT.
Collapse
|
28
|
Matte A, Cappellini MD, Iolascon A, Enrica F, De Franceschi L. Emerging drugs in randomized controlled trials for sickle cell disease: are we on the brink of a new era in research and treatment? Expert Opin Investig Drugs 2019; 29:23-31. [PMID: 31847604 DOI: 10.1080/13543784.2020.1703947] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Sickle cell disease (SCD) is caused by a mutation in the HBB gene which is key for making a component of hemoglobin. The mutation leads to the formation of an abnormal hemoglobin molecule called sickle hemoglobin (HbS). SCD is a chronic, complex disease with a multiplicity of pathophysiological targets; it has high morbidity and mortality.Hydroxyurea has for many years been the only approved drug for SCD; hence, the development of new therapeutics is critical.Areas covered: This article offers an overview of the key studies of new therapeutic options for SCD. We searched the PubMed database and Cochrane Database of Systemic Reviews for agents in early phase clinic trials and preclinical development.Expert opinion: Although knowledge of SCD has progressed, patient survival and quality of life must be improved. Phase II and phase III clinical trials investigating pathophysiology-based novel agents show promising results in the clinical management of SCD acute events. The design of long-term clinical studies is necessary to fully understand the clinical impact of these new therapeutics on the natural history of the disease. Furthermore, the building of global collaborations will enhance the clinical management of SCD and the design of primary outcomes of future clinical trials.
Collapse
Affiliation(s)
- Alessandro Matte
- Department of Medicine, University of Verona and AOUI Verona, Policlinico GB Rossi, Verona, Italy
| | - Maria Domenica Cappellini
- Ca Granda Foundation IRCCS, Dept of Clinical Science and Community, University of Milan, Milan, Italy
| | - Achille Iolascon
- Dept of Chemical Sciences, University Federico II, Naples, Italy
| | - Federti Enrica
- Department of Medicine, University of Verona and AOUI Verona, Policlinico GB Rossi, Verona, Italy
| | - Lucia De Franceschi
- Department of Medicine, University of Verona and AOUI Verona, Policlinico GB Rossi, Verona, Italy
| |
Collapse
|
29
|
Roumenina LT, Bartolucci P, Pirenne F. The role of Complement in Post-Transfusion Hemolysis and Hyperhemolysis Reaction. Transfus Med Rev 2019; 33:225-230. [DOI: 10.1016/j.tmrv.2019.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/12/2019] [Accepted: 09/12/2019] [Indexed: 02/08/2023]
|
30
|
Van Avondt K, Nur E, Zeerleder S. Mechanisms of haemolysis-induced kidney injury. Nat Rev Nephrol 2019; 15:671-692. [PMID: 31455889 DOI: 10.1038/s41581-019-0181-0] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2019] [Indexed: 12/16/2022]
Abstract
Intravascular haemolysis is a fundamental feature of chronic hereditary and acquired haemolytic anaemias, including those associated with haemoglobinopathies, complement disorders and infectious diseases such as malaria. Destabilization of red blood cells (RBCs) within the vasculature results in systemic inflammation, vasomotor dysfunction, thrombophilia and proliferative vasculopathy. The haemoprotein scavengers haptoglobin and haemopexin act to limit circulating levels of free haemoglobin, haem and iron - potentially toxic species that are released from injured RBCs. However, these adaptive defence systems can fail owing to ongoing intravascular disintegration of RBCs. Induction of the haem-degrading enzyme haem oxygenase 1 (HO1) - and potentially HO2 - represents a response to, and endogenous defence against, large amounts of cellular haem; however, this system can also become saturated. A frequent adverse consequence of massive and/or chronic haemolysis is kidney injury, which contributes to the morbidity and mortality of chronic haemolytic diseases. Intravascular destruction of RBCs and the resulting accumulation of haemoproteins can induce kidney injury via a number of mechanisms, including oxidative stress and cytotoxicity pathways, through the formation of intratubular casts and through direct as well as indirect proinflammatory effects, the latter via the activation of neutrophils and monocytes. Understanding of the detailed pathophysiology of haemolysis-induced kidney injury offers opportunities for the design and implementation of new therapeutic strategies to counteract the unfavourable and potentially fatal effects of haemolysis on the kidney.
Collapse
Affiliation(s)
- Kristof Van Avondt
- Department of Immunopathology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands. .,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Munich, Germany.
| | - Erfan Nur
- Department of Haematology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Sacha Zeerleder
- Department of Immunopathology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands. .,Department of Haematology and Central Haematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland. .,Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
31
|
Clinical promise of next-generation complement therapeutics. Nat Rev Drug Discov 2019; 18:707-729. [PMID: 31324874 DOI: 10.1038/s41573-019-0031-6] [Citation(s) in RCA: 240] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2019] [Indexed: 02/07/2023]
Abstract
The complement system plays a key role in pathogen immunosurveillance and tissue homeostasis. However, subversion of its tight regulatory control can fuel a vicious cycle of inflammatory damage that exacerbates pathology. The clinical merit of targeting the complement system has been established for rare clinical disorders such as paroxysmal nocturnal haemoglobinuria and atypical haemolytic uraemic syndrome. Evidence from preclinical studies and human genome-wide analyses, supported by new molecular and structural insights, has revealed new pathomechanisms and unmet clinical needs that have thrust a new generation of complement inhibitors into clinical development for a variety of indications. This review critically discusses recent clinical milestones in complement drug discovery, providing an updated translational perspective that may guide optimal target selection and disease-tailored complement intervention.
Collapse
|
32
|
Merrill SA, Brodsky RA, Lanzkron SM, Naik R. A case-control analysis of hyperhemolysis syndrome in adults and laboratory correlates of complement involvement. Transfusion 2019; 59:3129-3139. [PMID: 31292968 DOI: 10.1111/trf.15445] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/10/2019] [Accepted: 06/18/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Hyperhemolysis syndrome (HS) is a poorly understood, severe hemolytic anemia provoked by transfusion. Both host and donor RBCs are destroyed in HS; thus, transfusion paradoxically worsens anemia. Risk factors and mechanism of HS are unknown. STUDY DESIGN AND METHODS A retrospective case-control analysis was performed on adults with HS. Patients with HS were matched 1:1 with matched, transfused controls, and HS risk factors were analyzed with multivariable logistic regression. HS samples were analyzed for complement deposition by flow cytometry, and an in vitro model of bystander hemolysis was developed. RESULTS Forty-one patients with 54 episodes of HS were identified in a 26-year period from 1992 to 2018. Of the HS episodes, only 18.5% were associated with a new alloantibody, and such patients were more tolerant of additional transfusion in the acute episode (p = 0.005). Thirteen percent of episodes were fatal, and HS recurred in 52.6%. Alloimmunization (odds ratio [OR], 17.3), non-B blood type (OR, 9.8), D antigen (OR, 9.1), and infection (OR, 5.5) were associated with HS on multivariable analysis. Hyperbilirubinemia was predictive of fatal HS (OR, 33.6). Increased complement was observed on RBCs during HS episodes, and the in vitro model of bystander hemolysis recapitulated complement decoration of sickled RBCs. CONCLUSIONS HS is associated with significant morbidity, mortality, and recurrence. Risk factors such as known alloimmunization, blood group, and infection predispose to HS. Bystander complement activation may drive HS. These factors may help physicians refine risk-benefit assessments for transfusion and guide further therapeutic development.
Collapse
Affiliation(s)
- Samuel A Merrill
- Division of Hematology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Robert A Brodsky
- Division of Hematology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sophie M Lanzkron
- Division of Hematology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Rakhi Naik
- Division of Hematology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
33
|
Nemer WE, Koehl B. Factor H: a novel modulator in sickle cell disease. Haematologica 2019; 104:857-859. [PMID: 31040228 DOI: 10.3324/haematol.2018.214668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Wassim El Nemer
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Univ. Paris Diderot, Sorbonne Paris Cité, Univ. de la Réunion, Univ. des Antilles.,Institut National de la Transfusion Sanguine, F-75015.,Laboratoire d'Excellence GR-Ex
| | - Bérengère Koehl
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Univ. Paris Diderot, Sorbonne Paris Cité, Univ. de la Réunion, Univ. des Antilles.,Institut National de la Transfusion Sanguine, F-75015.,Laboratoire d'Excellence GR-Ex.,Hematology Unit, Sickle Cell Disease Center, Robert Debré Hospital, AP-HP, Paris, France
| |
Collapse
|
34
|
Vercellotti GM, Dalmasso AP, Schaid TR, Nguyen J, Chen C, Ericson ME, Abdulla F, Killeen T, Lindorfer MA, Taylor RP, Belcher JD. Critical role of C5a in sickle cell disease. Am J Hematol 2019; 94:327-337. [PMID: 30569594 DOI: 10.1002/ajh.25384] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/16/2018] [Accepted: 12/10/2018] [Indexed: 12/31/2022]
Abstract
Innate immune complement activation may contribute to sickle cell disease (SCD) pathogenesis. Ischemia-reperfusion physiology is a key component of the inflammatory and vaso-occlusive milieu in SCD and is associated with complement activation. C5a is an anaphylatoxin, a potent pro-inflammatory mediator that can activate leukocytes, platelets, and endothelial cells, all of which play a role in vaso-occlusion. We hypothesize that hypoxia-reoxygenation (H/R) in SCD mice activates complement, promoting inflammation and vaso-occlusion. At baseline and after H/R, sickle Townes-SS mice had increased C3 activation fragments and C5b-9 deposition in kidneys, livers and lungs and alternative pathway Bb fragments in plasma compared to control AA-mice. Activated complement promoted vaso-occlusion (microvascular stasis) in SS-mice; infusion of zymosan-activated, but not heat-inactivated serum, induced substantial vaso-occlusion in the skin venules of SS-mice. Infusion of recombinant C5a induced stasis in SS, but not AA-mice that was blocked by anti-C5a receptor (C5aR) IgG. C5a-mediated stasis was accompanied by inflammatory responses in SS-mice including NF-κB activation and increased expression of TLR4 and adhesion molecules VCAM-1, ICAM-1, and E-selectin in the liver. Anti-C5aR IgG blocked these inflammatory responses. Also, C5a rapidly up-regulated Weibel-Palade body P-selectin and von Willebrand factor on the surface of human umbilical vein endothelial cells in vitro and on vascular endothelium in vivo. In SS-mice, a blocking antibody to P-selectin inhibited C5a-induced stasis. Similarly, an antibody to C5 that blocks murine C5 cleavage or an antibody that blocks C5aR inhibited H/R-induced stasis in SS-mice. These results suggest that inhibition of C5a may be beneficial in SCD.
Collapse
Affiliation(s)
- Gregory M. Vercellotti
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | | | - Terry R. Schaid
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Julia Nguyen
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Chunsheng Chen
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Marna E. Ericson
- Department of Dermatology; University of Minnesota; Minneapolis Minnesota
| | - Fuad Abdulla
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Trevor Killeen
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Margaret A. Lindorfer
- Department of Biochemistry and Molecular Genetics; University of Virginia School of Medicine; Charlottesville Virginia
| | - Ronald P. Taylor
- Department of Biochemistry and Molecular Genetics; University of Virginia School of Medicine; Charlottesville Virginia
| | - John D. Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| |
Collapse
|
35
|
Balbuena-Merle R, Hendrickson JE. Red blood cell alloimmunization and delayed hemolytic transfusion reactions in patients with sickle cell disease. Transfus Clin Biol 2019; 26:112-115. [PMID: 30857806 DOI: 10.1016/j.tracli.2019.02.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Red blood cell (RBC) alloimmunization is more common in patients with sickle cell disease (SCD) than in any other studied patient population. The high prevalence of RBC alloimmunization is multi-factorial, likely involving the chronic hemolysis and inflammatory status of SCD itself, the transfusion burden of patients, and the RH genetic diversity of patients and blood donors, among other reasons. Antibody evanescence, or the decrease of RBC alloantibodies below levels detectable by blood bank testing, occurs frequently with fewer than 30% of alloantibodies estimated to be detected by current screening practices. Evanescence increases the likelihood that a patient with SCD will have a delayed hemolytic transfusion reaction upon future RBC exposure, with previously undetected alloantibodies coming roaring back in an anamnestic manner after exposure to the cognate RBC antigen. A subset of patients having delayed hemolytic transfusion reactions go on to experience hyperhemolysis; some but not all cases of hyperhemolysis are associated with previously evanescent RBC alloantibodies. There is an increasing appreciation of the association between RBC alloantibodies and RBC autoantibodies, as well as involvement of the alternative complement pathway in some instances of hyperhemolysis. A case report in this manuscript describes a highly alloimmunized patient with SCD who experiences a delayed hemolytic transfusion reaction with bystander hemolysis due to a previously evanescent, complement binding anti-M RBC alloantibody. Additional studies, including those involving multiple centers and countries, are needed to further understand RBC alloimmunization in patients with SCD and to develop strategies to prevent or mitigate potentially life-threatening hemolytic transfusion reactions.
Collapse
Affiliation(s)
- R Balbuena-Merle
- Department of Laboratory Medicine, 330 Cedar Street, CB 405, New Haven, CT 06520-8035, United States
| | - J E Hendrickson
- Department of Laboratory Medicine, 330 Cedar Street, CB 405, New Haven, CT 06520-8035, United States.
| |
Collapse
|
36
|
Complement activation during intravascular hemolysis: Implication for sickle cell disease and hemolytic transfusion reactions. Transfus Clin Biol 2019; 26:116-124. [PMID: 30879901 DOI: 10.1016/j.tracli.2019.02.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Intravascular hemolysis is a hallmark of a large spectrum of diseases, including the sickle cell disease (SCD), and is characterized by liberation of red blood cell (RBC) degradation products in the circulation. Released Hb, heme, RBC fragments and microvesicles (MVs) exert pro-inflammatory, pro-oxidative and cytotoxic effects and contribute to vascular and tissue damage. The innate immune complement system not only contributes to the RBC lysis, but it is also itself activated by heme, RBC MVs and the hypoxia-altered endothelium, amplifying thus the cell and tissue damage. This review focuses on the implication of the complement system in hemolysis and hemolysis-mediated injuries in SCD and in cases of delayed hemolytic transfusion reactions (DHTR). We summarize the evidences for presence of biomarkers of complement activation in patients with SCD and the mechanisms of complement activation in DHTR. We discuss the role of antibodies-dependent activation of the classical complement pathway as well as the heme-dependent activation of the alternative pathway. Finally, we describe the available evidences for the efficacy of therapeutic blockade of complement in cases of DHTR. In conclusion, complement blockade is holding promises but future prospective studies are required to introduce Eculizumab or another upcoming complement therapeutic for DHTR and even in SCD.
Collapse
|
37
|
Telen MJ, Malik P, Vercellotti GM. Therapeutic strategies for sickle cell disease: towards a multi-agent approach. Nat Rev Drug Discov 2019; 18:139-158. [PMID: 30514970 PMCID: PMC6645400 DOI: 10.1038/s41573-018-0003-2] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
For over 100 years, clinicians and scientists have been unravelling the consequences of the A to T substitution in the β-globin gene that produces haemoglobin S, which leads to the systemic manifestations of sickle cell disease (SCD), including vaso-occlusion, anaemia, haemolysis, organ injury and pain. However, despite growing understanding of the mechanisms of haemoglobin S polymerization and its effects on red blood cells, only two therapies for SCD - hydroxyurea and L-glutamine - are approved by the US Food and Drug Administration. Moreover, these treatment options do not fully address the manifestations of SCD, which arise from a complex network of interdependent pathophysiological processes. In this article, we review efforts to develop new drugs targeting these processes, including agents that reactivate fetal haemoglobin, anti-sickling agents, anti-adhesion agents, modulators of ischaemia-reperfusion and oxidative stress, agents that counteract free haemoglobin and haem, anti-inflammatory agents, anti-thrombotic agents and anti-platelet agents. We also discuss gene therapy, which holds promise of a cure, although its widespread application is currently limited by technical challenges and the expense of treatment. We thus propose that developing systems-oriented multi-agent strategies on the basis of SCD pathophysiology is needed to improve the quality of life and survival of people with SCD.
Collapse
Affiliation(s)
- Marilyn J Telen
- Division of Hematology, Department of Medicine and Duke Comprehensive Sickle Cell Center, Duke University, Durham, NC, USA.
| | - Punam Malik
- Division of Experimental Hematology and Cancer Biology and the Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Gregory M Vercellotti
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
38
|
Zanette DL, Santiago RP, Leite IPR, Santana SS, da Guarda C, Maffili VV, Ferreira JRD, Adanho CSA, Yahouedehou SCMA, Menezes IL, Goncalves MS. Differential gene expression analysis of sickle cell anemia in steady and crisis state. Ann Hum Genet 2019; 83:310-317. [PMID: 30698275 DOI: 10.1111/ahg.12290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 09/21/2018] [Accepted: 09/25/2018] [Indexed: 11/27/2022]
Abstract
Sickle cell anemia is one of the most prevalent genetic diseases worldwide, showing great clinical heterogeneity. This study compared the gene expression patterns between sickle cell anemia pediatric patients in steady state and in crisis state, as compared to age-paired, healthy individuals. RNA sequencing was performed from these groups of patients/controls using Illumina HiSeq 2500 equipment. The resulting differentially expressed genes were loaded into QIAGEN's ingenuity pathway analysis. The results showed that EIF2 pathway and NRF2-mediated oxidative stress-response pathways were more highly activated both in steady state and in crisis patients, as compared to healthy individuals. In addition, we found increased activation of eIF4 and p70S6K signaling pathways in crisis state compared to healthy individuals. The transcription factor GATA-1 was found exclusively in steady state while SPI was found exclusively in crisis state. IL6 and VEGFA were found only in crisis state, while IL-1B was found exclusively in steady state. The regulator effects analysis revealed IgG1 as an upstream regulator in steady state compared to healthy individuals, resulting in invasion of prostate cancer cell lines as the disease/function outcome. For crisis-state patients versus healthy individuals, two networks of regulator effects revealed STAT1, CD40LG, TGM2, IRF7, IRF4, and IRF1 acting as upstream regulators, resulting in disease/function outcomes, including engulfment of cells and aggregation of blood cells and inflammation of joints. Our results indicated genes and pathways that can provide clues on the molecular events involved in the severity of sickle cell disease.
Collapse
Affiliation(s)
- Dalila L Zanette
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - Fiocruz/BA, 40296710, Salvador, Bahia, Brazil
| | - Rayra P Santiago
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - Fiocruz/BA, 40296710, Salvador, Bahia, Brazil
| | - Ivana Paula Ribeiro Leite
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - Fiocruz/BA, 40296710, Salvador, Bahia, Brazil.,Hospital Pediátrico Professor Hosannah de Oliveira, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Sanzio S Santana
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - Fiocruz/BA, 40296710, Salvador, Bahia, Brazil
| | - Caroline da Guarda
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - Fiocruz/BA, 40296710, Salvador, Bahia, Brazil
| | - Vitor V Maffili
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - Fiocruz/BA, 40296710, Salvador, Bahia, Brazil
| | | | | | | | - Isa Lyra Menezes
- Hospital Pediátrico Professor Hosannah de Oliveira, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Marilda Souza Goncalves
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - Fiocruz/BA, 40296710, Salvador, Bahia, Brazil.,Faculdade de Farmácia, Universidade Federal da Bahia, 40170115, Salvador, Bahia, Brasil
| |
Collapse
|
39
|
Lombardi E, Matte A, Risitano AM, Ricklin D, Lambris JD, De Zanet D, Jokiranta ST, Martinelli N, Scambi C, Salvagno G, Bisoffi Z, Colato C, Siciliano A, Bortolami O, Mazzuccato M, Zorzi F, De Marco L, De Franceschi L. Factor H interferes with the adhesion of sickle red cells to vascular endothelium: a novel disease-modulating molecule. Haematologica 2019; 104:919-928. [PMID: 30630982 PMCID: PMC6518911 DOI: 10.3324/haematol.2018.198622] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 01/08/2019] [Indexed: 12/27/2022] Open
Abstract
Sickle cell disease is an autosomal recessive genetic red cell disorder with a worldwide distribution. Growing evidence suggests a possible involvement of complement activation in the severity of clinical complications of sickle cell disease. In this study we found activation of the alternative complement pathway with microvascular deposition of C5b-9 on skin biopsies from patients with sickle cell disease. There was also deposition of C3b on sickle red cell membranes, which is promoted locally by the exposure of phosphatidylserine. In addition, we showed for the first time a peculiar “stop-and-go” motion of sickle cell red blood cells on tumor factor-α–activated vascular endothelial surfaces. Using the C3b/iC3b binding plasma protein factor Has an inhibitor of C3b cell-cell interactions, we found that factor H and its domains 19-20 prevent the adhesion of sickle red cells to the endothelium, normalizing speed transition times of red cells. We documented that factor H acts by preventing the adhesion of sickle red cells to P-selectin and/or the Mac-1 receptor (CD11b/CD18), supporting the activation of the alternative pathway of complement as an additional mechanism in the pathogenesis of acute sickle cell related vaso-occlusive crises. Our data provide a rationale for further investigation of the potential contribution of factor H and other modulators of the alternative complement pathway with potential implications for the treatment of sickle cell disease.
Collapse
Affiliation(s)
| | | | - Antonio M Risitano
- Hematology, Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Daniel Ricklin
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, University of Basel, Switzerland
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; USA
| | - Denise De Zanet
- Department of Translational Research, National Cancer Center, Aviano, Italy.,Polytechnic Department of Engineering and Architecture, University of Udine, Italy
| | - Sakari T Jokiranta
- Research Programs Unit, Immunobiology, University of Helsinki and United Medix Laboratories, Helsinki, Finland
| | | | - Cinzia Scambi
- Department of Medicine, University of Verona-AOUI Verona; Italy
| | - Gianluca Salvagno
- Laboratory of Clinical Biochemistry, Department of Life and Reproduction Sciences, University of Verona, Italy
| | - Zeno Bisoffi
- Centre of Tropical Diseases, Sacro Cuore-Don Calabria Hospital Negrar, Verona, Italy.,Department of Diagnostics and Public Health, University of Verona-AOUI Verona, Italy
| | - Chiara Colato
- Department of Diagnostics and Public Health, University of Verona-AOUI Verona, Italy
| | | | - Oscar Bortolami
- Unit of Epidemiology and Medical Statistics, Department of Diagnostic & Public Health, University of Verona
| | - Mario Mazzuccato
- Department of Translational Research, National Cancer Center, Aviano, Italy
| | - Francesco Zorzi
- Department of Medicine, University of Verona-AOUI Verona; Italy
| | - Luigi De Marco
- Department of Translational Research, National Cancer Center, Aviano, Italy.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | |
Collapse
|
40
|
Merle NS, Grunenwald A, Rajaratnam H, Gnemmi V, Frimat M, Figueres ML, Knockaert S, Bouzekri S, Charue D, Noe R, Robe-Rybkine T, Le-Hoang M, Brinkman N, Gentinetta T, Edler M, Petrillo S, Tolosano E, Miescher S, Le Jeune S, Houillier P, Chauvet S, Rabant M, Dimitrov JD, Fremeaux-Bacchi V, Blanc-Brude OP, Roumenina LT. Intravascular hemolysis activates complement via cell-free heme and heme-loaded microvesicles. JCI Insight 2018; 3:96910. [PMID: 29925688 DOI: 10.1172/jci.insight.96910] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 05/08/2018] [Indexed: 01/08/2023] Open
Abstract
In hemolytic diseases, such as sickle cell disease (SCD), intravascular hemolysis results in the release of hemoglobin, heme, and heme-loaded membrane microvesicles in the bloodstream. Intravascular hemolysis is thus associated with inflammation and organ injury. Complement system can be activated by heme in vitro. We investigated the mechanisms by which hemolysis and red blood cell (RBC) degradation products trigger complement activation in vivo. In kidney biopsies of SCD nephropathy patients and a mouse model with SCD, we detected tissue deposits of complement C3 and C5b-9. Moreover, drug-induced intravascular hemolysis or injection of heme or hemoglobin in mice triggered C3 deposition, primarily in kidneys. Renal injury markers (Kim-1, NGAL) were attenuated in C3-/- hemolytic mice. RBC degradation products, such as heme-loaded microvesicles and heme, induced alternative and terminal complement pathway activation in sera and on endothelial surfaces, in contrast to hemoglobin. Heme triggered rapid P selectin, C3aR, and C5aR expression and downregulated CD46 on endothelial cells. Importantly, complement deposition was attenuated in vivo and in vitro by heme scavenger hemopexin. In conclusion, we demonstrate that intravascular hemolysis triggers complement activation in vivo, encouraging further studies on its role in SCD nephropathy. Conversely, heme inhibition using hemopexin may provide a novel therapeutic opportunity to limit complement activation in hemolytic diseases.
Collapse
Affiliation(s)
- Nicolas S Merle
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie - Paris 06, Paris France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne Grunenwald
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Université Lille, INSERM, CHRU Lille, Service de pathologie, UMRS 1172, Jean-Pierre Aubert Research Center, Lille, France
| | - Helena Rajaratnam
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.,SupBiotech Paris, Villejuif, France
| | - Viviane Gnemmi
- Université Lille, INSERM, CHRU Lille, Service de pathologie, UMRS 1172, Jean-Pierre Aubert Research Center, Lille, France
| | - Marie Frimat
- INSERM, UMR 995, Lille, France.,CHRU Lille, Service de néphrologie, Lille, France
| | - Marie-Lucile Figueres
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie - Paris 06, Paris France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Samantha Knockaert
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie - Paris 06, Paris France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sanah Bouzekri
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Dominique Charue
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Paris Center for Cardiovascular Research, INSERM UMRS 970, Paris, France
| | - Remi Noe
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.,Ecole Pratique des Hautes Études, Paris, France
| | - Tania Robe-Rybkine
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie - Paris 06, Paris France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marie Le-Hoang
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Paris Center for Cardiovascular Research, INSERM UMRS 970, Paris, France
| | | | | | | | - Sara Petrillo
- Department Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Emanuela Tolosano
- Department Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | | | - Sylvain Le Jeune
- Assistance Publique - Hôpitaux de Paris, Service de Médecine Interne, Hôpital Avicenne, Bobigny, France
| | - Pascal Houillier
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie - Paris 06, Paris France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sophie Chauvet
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie - Paris 06, Paris France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service de Néphrologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Marion Rabant
- Assistance Publique - Hôpitaux de Paris, Service de Pathologie, Hôpital Necker Enfants Malades, Paris, France
| | - Jordan D Dimitrov
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie - Paris 06, Paris France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Veronique Fremeaux-Bacchi
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie - Paris 06, Paris France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou, Paris, France
| | - Olivier P Blanc-Brude
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Paris Center for Cardiovascular Research, INSERM UMRS 970, Paris, France
| | - Lubka T Roumenina
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie - Paris 06, Paris France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
41
|
Abstract
The primary β-globin gene mutation that causes sickle cell disease (SCD) has significant pathophysiological consequences that result in hemolytic events and the induction of the inflammatory processes that ultimately lead to vaso-occlusion. In addition to their role in the initiation of the acute painful vaso-occlusive episodes that are characteristic of SCD, inflammatory processes are also key components of many of the complications of the disease including autosplenectomy, acute chest syndrome, pulmonary hypertension, leg ulcers, nephropathy and stroke. We, herein, discuss the events that trigger inflammation in the disease, as well as the mechanisms, inflammatory molecules and cells that propagate these inflammatory processes. Given the central role that inflammation plays in SCD pathophysiology, many of the therapeutic approaches currently under pre-clinical and clinical development for the treatment of SCD endeavor to counter aspects or specific molecules of these inflammatory processes and it is possible that, in the future, we will see anti-inflammatory drugs being used either together with, or in place of, hydroxyurea in those SCD patients for whom hematopoietic stem cell transplants and evolving gene therapies are not a viable option.
Collapse
Affiliation(s)
- Nicola Conran
- Hematology Center, University of Campinas - UNICAMP, Cidade Universitária, Campinas-SP, Brazil
| | - John D Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
42
|
Abstract
Non-adherence and deformability are the key intrinsic biomechanical features of the red blood cell (RBC), which allow it to tightly squeeze and pass through even the narrowest of microcirculatory networks. Blockage of microcirculatory flow, also known as vaso-occlusion, is a consequence of abnormal cellular adhesion to the vascular endothelium. In sickle cell disease (SCD), an inherited anaemia, even though RBCs have been shown to be heterogeneous in adhesiveness and deformability, this has not been studied in the context of physiologically relevant dynamic shear gradients at the microscale. We developed a microfluidic system that simulates physiologically relevant shear gradients of microcirculatory blood flow at a constant single volumetric flow rate. Using this system, shear dependent adhesion of RBCs from 28 subjects with SCD and from 11 healthy subjects was investigated using vascular endothelial protein functionalized microchannels. We defined a new term, RBC Shear Gradient Microfluidic Adhesion (SiGMA) index to assess shear dependent RBC adhesion in a subject-specific manner. We have shown for the first time that shear dependent adhesion of RBCs is heterogeneous in a microfluidic flow model, which correlates clinically with inflammatory markers and iron overload in subjects with SCD. This study reveals the complex dynamic interactions between RBC-mediated microcirculatory occlusion and clinical outcomes in SCD. These interactions may also be relevant to other microcirculatory disorders and microvascular diseases.
Collapse
Affiliation(s)
- Erdem Kucukal
- Department of Mechanical and Aerospace Engineering, Case Biomanufacturing and Microfabrication Laboratory, Case Western Reserve University, Glennan 616B, 10900 Euclid Ave., Cleveland, OH, USA.
| | - Jane A Little
- Department of Hematology and Oncology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA and Seidman Cancer Center at University Hospitals, Case Medical Center, Cleveland, OH, USA
| | - Umut A Gurkan
- Department of Mechanical and Aerospace Engineering, Case Biomanufacturing and Microfabrication Laboratory, Case Western Reserve University, Glennan 616B, 10900 Euclid Ave., Cleveland, OH, USA. and Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA and Department of Orthopaedics, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
43
|
Taylor RP, Lindorfer MA. Mechanisms of Complement-Mediated Damage in Hematological Disorders. Semin Hematol 2018; 55:118-123. [PMID: 30032747 DOI: 10.1053/j.seminhematol.2018.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 02/09/2018] [Indexed: 12/27/2022]
Abstract
The complement cascade is an ancient defense system that destroys and eliminates threats to normal homeostasis in the bloodstream and tissues. Although multiple controls keep complement in check to minimize innocent bystander injury to normal cells and tissues, defects in complement regulation due to mutations in, or autoantibodies to, complement control proteins underlie the pathogenesis of several hemolytic diseases including paroxysmal nocturnal hemoglobinuria, and atypical hemolytic uremic syndrome. In autoimmune hemolytic anemias complement plays an important role in erythrocyte destruction mediated by antierythrocyte antibodies. The pathogenic mechanisms of these hemolytic diseases are discussed, with an emphasis on pivotal steps in complement activation.
Collapse
Affiliation(s)
- Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA.
| | - Margaret A Lindorfer
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA
| |
Collapse
|
44
|
Red Blood Cell Transfusion and Surgical Site Infection After Colon Resection Surgery: A Cohort Study. Anesth Analg 2017; 125:1316-1321. [PMID: 28504993 DOI: 10.1213/ane.0000000000002099] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Surgical site infections (SSIs) after colon surgery remain a critical safety issue. Patients with an SSI have an increased risk of death, prolonged hospitalization, and increased costs of care. Red blood cell (RBC) transfusion is given during the perioperative period to increase blood oxygen delivery, but it is associated with complications, including infection. We hypothesized that RBC transfusion would be associated with increased SSI risk in patients undergoing colon resection surgery. METHODS A retrospective cohort study was performed using the 2014 National Surgical Quality Improvement Program participant use file. Patients who had colon resection surgery were identified using current procedural terminology codes. The association between perioperative RBC transfusion and superficial and deep incisional SSIs, organ space SSIs, and postoperative septic shock was modeled using logistic regression with propensity score analysis. RESULTS Of 23,388 patients who had colon resection surgery, 1845 (7.9%) received perioperative RBC transfusion. After controlling for confounders with propensity score analysis and inverse probability of treatment weighting, RBC transfusion had no apparent association with superficial incisional SSI (odds ratio [OR], 1.18; 99% confidence interval [CI], 0.48-2.88) or deep incisional SSI (OR, 1.47; 99% CI, 0.23-9.43). However, RBC transfusion appeared to be associated with increased risk of organ space SSI (OR, 2.93; 99% CI, 1.43-6.01) and septic shock (OR, 9.23; 99% CI, 3.53-24.09). CONCLUSIONS RBC transfusion has no apparent association with increased risk for incisional SSIs, but may be associated with increased risk for organ space SSI and septic shock after colon resection surgery.
Collapse
|
45
|
Gavriilaki E, Mainou M, Christodoulou I, Koravou EE, Paleta A, Touloumenidou T, Papalexandri A, Athanasiadou A, Apostolou C, Klonizakis P, Anagnostopoulos A, Vlachaki E. In vitro evidence of complement activation in patients with sickle cell disease. Haematologica 2017; 102:e481-e482. [PMID: 28912175 DOI: 10.3324/haematol.2017.174201] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Eleni Gavriilaki
- Haematology Department BMT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| | - Maria Mainou
- Adults Thalassaemia Unit, 2 Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, Greece
| | - Ioanna Christodoulou
- Adults Thalassaemia Unit, 2 Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, Greece
| | | | - Aggeliki Paleta
- Haematology Department BMT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| | | | | | | | - Chrysa Apostolou
- Adults Thalassaemia Unit, 2 Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, Greece
| | - Philippos Klonizakis
- Adults Thalassaemia Unit, 2 Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, Greece
| | | | - Efthymia Vlachaki
- Adults Thalassaemia Unit, 2 Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, Greece
| |
Collapse
|
46
|
Larson MC, Karafin MS, Hillery CA, Hogg N. Phosphatidylethanolamine is progressively exposed in RBCs during storage. Transfus Med 2017; 27:136-141. [PMID: 28134466 DOI: 10.1111/tme.12382] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/15/2016] [Accepted: 11/26/2016] [Indexed: 01/28/2023]
Abstract
BACKGROUND It is well established that as a blood unit ages, fewer of the unit's red blood cells (RBCs) remain in circulation post-transfusion. The mechanism for clearance is not well defined. Phosphatidylethanolamine (PE) is a phospholipid that is primarily found on the inner leaflet of healthy cells, and is an important ligand for phagocytosis of dead cells when exposed. OBJECTIVES The objective of the present study was to measure the change in PE exposure in donor RBCs over increasing storage ages using the novel PE-specific probe, duramycin. METHODS Five adsol (AS-1) preserved RBC units were sampled weekly for 6 weeks and were labelled with duramycin. The percentage of PE exposed on red cells in each sample was determined using flow cytometry. Surface phosphatidylserine (PS) was evaluated for comparison. RESULTS We found that RBCs in AS-preserved donor units increasingly exposed PE, from less than 1% in freshly processed RBCs, to nearly 20% at 42 days of storage and correlated with increased relative vesiculation or microparticle concentration and release of cell-free haemoglobin. By comparison, only 5% of cells exposed PS at 42 days. CONCLUSION We conclude that exposure of PE in the RBC outer membrane was higher than that of PS during 42 days of storage and correlated significantly with increased vesiculation and release of haemoglobin.
Collapse
Affiliation(s)
- M C Larson
- Medical Imaging Department, University of Arizona, Tucson, Arizona, USA
| | - M S Karafin
- Blood Research Institute, Blood Center of Wisconsin, Wisconsin, USA.,Pathology Department, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - C A Hillery
- Pediatric Hematology Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - N Hogg
- Biophysics Department, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
47
|
Barretto OCDO, Soeiro Prestes K, Figueiredo Fonseca LK, Achucarro Silveira PA. Functional alterations of rabbit erythrocytes induced by Loxosceles gaucho venom. Hum Exp Toxicol 2016; 26:817-21. [DOI: 10.1177/0960327107084070] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Loxoscelism is the syndrome caused by the brown spider Loxosceles gaucho bite in humans. Its effect on erythrocytes has been studied in humans, rabbits, pigs and guinea pigs. In this study, the damage that the L gaucho spider venom causes to the structure and function of erythrocytes in vivo was investigated in rabbits. Before and after the rabbits were envenomed, membrane proteins were studied through polyacrylamide gel electrophoresis and membrane function was ascertained using the osmotic fragility test, together with the highly sensitive technique of ektacytometry. Six New Zealand rabbits were inoculated by intradermal injection into the dorsal region (10 μg of venom/kg of body weight in 0.2 mL of saline). Blood was collected at 24, 48, 72 and 120 h after inoculation. The membrane protein study did not reveal any alteration in their relative band concentrations, but the osmotic fragility test showed increased hemolysis in slightly hypotonic sodium chloride solutions (at 0.6 and 0.55%). In addition, the ektacytometer study revealed greater deformability to increasing shear stress on the order of 3—30 Pascals when compared with controls, showing that the L gaucho venom does in fact alter red cell function. Human & Experimental Toxicology (2007) 26 , 817— 821
Collapse
|
48
|
Chapin J, Terry HS, Kleinert D, Laurence J. The role of complement activation in thrombosis and hemolytic anemias. Transfus Apher Sci 2016; 54:191-8. [PMID: 27156108 DOI: 10.1016/j.transci.2016.04.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The objective of this study was to describe complement activation in hemostatic and pathologic states of coagulation and in the acquired and congenital hemolytic anemias. METHODS AND RESULTS We review published and emerging data on the involvement of the classic, alternative and lectin-based complement pathways in coagulation and the hemolytic anemias. The alternative pathway in particular is always "on," at low levels, and is particularly sensitive to hyper-activation in a variety of physiologic and pathologic states including infection, autoimmune disorders, thrombosis and pregnancy, requiring tight control predicated on a variety of soluble and membrane bound regulatory proteins. In acquired hemolytic anemias such as paroxysmal nocturnal hemoglobinuria (PNH) and cold agglutinin disease (CAD), the complement system directly induces red blood cell injury, resulting in intravascular and extravascular hemolysis. In congenital hemolytic anemias such as sickle cell disease and β-thalassemia, the complement system may also contribute to thrombosis and vascular disease. Complement activation may also lead to a storage lesion in red blood cells prior to transfusion. CONCLUSION Complement pathways are activated in hemolytic anemias and are closely linked with thrombosis. In acquired disorders such as PNH and possibly CAD, inhibition of the alternative complement pathway improves clinical outcomes and reduces thrombosis risk. Whether complement inhibition has a similar role in congenital hemolytic anemias apart from the atypical hemolytic-uremic (aHUS)-type thrombotic microangiopathies remains to be determined.
Collapse
Affiliation(s)
- John Chapin
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine-New York Presbyterian, New York, NY, USA.
| | - Hunter S Terry
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine-New York Presbyterian, New York, NY, USA
| | - Dorothy Kleinert
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine-New York Presbyterian, New York, NY, USA
| | - Jeffrey Laurence
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine-New York Presbyterian, New York, NY, USA
| |
Collapse
|
49
|
Eculizumab salvage therapy for delayed hemolysis transfusion reaction in sickle cell disease patients. Blood 2016; 127:1062-4. [DOI: 10.1182/blood-2015-09-669770] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
50
|
Olsen I, Yilmaz Ö. Modulation of inflammasome activity by Porphyromonas gingivalis in periodontitis and associated systemic diseases. J Oral Microbiol 2016; 8:30385. [PMID: 26850450 PMCID: PMC4744328 DOI: 10.3402/jom.v8.30385] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 01/06/2016] [Accepted: 01/06/2016] [Indexed: 12/19/2022] Open
Abstract
Inflammasomes are large multiprotein complexes localized in the cytoplasm of the cell. They are responsible for the maturation of pro-inflammatory cytokines such as interleukin-1β (IL-1β) and IL-18 as well as for the activation of inflammatory cell death, the so-called pyroptosis. Inflammasomes assemble in response to cellular infection, cellular stress, or tissue damage; promote inflammatory responses and are of great importance in regulating the innate immune system in chronic inflammatory diseases such as periodontitis and several chronic systemic diseases. In addition to sensing cellular integrity, inflammasomes are involved in the homeostatic mutualism between the indigenous microbiota and the host. There are several types of inflammasomes of which NLRP3 is best characterized in microbial pathogenesis. Many opportunistic bacteria try to evade the innate immune system in order to survive in the host cells. One of these is the periodontopathogen Porphyromonas gingivalis which has been shown to have several mechanisms of modulating innate immunity by limiting the activation of the NLRP3 inflammasome. Among them, ATP-/P2X7- signaling is recently associated not only with periodontitis but also with development of several systemic diseases. The present paper reviews multiple mechanisms through which P. gingivalis can modify innate immunity by affecting inflammasome activity.
Collapse
Affiliation(s)
- Ingar Olsen
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway;
| | - Özlem Yilmaz
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|