1
|
Tkachenko A, Havranek O. Erythronecroptosis: an overview of necroptosis or programmed necrosis in red blood cells. Mol Cell Biochem 2024; 479:3273-3291. [PMID: 38427167 DOI: 10.1007/s11010-024-04948-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/20/2024] [Indexed: 03/02/2024]
Abstract
Necroptosis is considered a programmed necrosis that requires receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3), and pore-forming mixed lineage kinase domain-like protein (MLKL) to trigger a regulated cell membrane lysis. Membrane rupture in necroptosis has been shown to fuel innate immune response due to release of damage-associated molecular patterns (DAMPs). Recently published studies indicate that mature erythrocytes can undergo necroptosis as well. In this review, we provide an outline of multiple cell death modes occurring in erythrocytes, discuss possible immunological aspects of diverse erythrocyte cell deaths, summarize available evidence related to the ability of erythrocytes to undergo necroptosis, outline key involved molecular mechanisms, and discuss the potential implication of erythrocyte necroptosis in the physiology and pathophysiology. Furthermore, we aim to highlight the interplay between necroptosis and eryptosis signaling in erythrocytes, emphasizing specific characteristics of these pathways distinct from their counterparts in nucleated cells. Thus, our review provides a comprehensive summary of the current knowledge of necroptosis in erythrocytes. To reflect critical differences between necroptosis of nucleated cells and necroptosis of erythrocytes, we suggest a term erythronecroptosis for necroptosis of enucleated cells.
Collapse
Affiliation(s)
- Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 25250, Vestec, Czech Republic.
| | - Ondrej Havranek
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 25250, Vestec, Czech Republic
- First Department of Internal Medicine-Hematology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
2
|
Cao Z, Jiang X, He Y, Zheng X. Metabolic landscape in venous thrombosis: insights into molecular biology and therapeutic implications. Ann Med 2024; 56:2401112. [PMID: 39297312 DOI: 10.1080/07853890.2024.2401112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 03/20/2024] [Accepted: 05/12/2024] [Indexed: 09/21/2024] Open
Abstract
The findings of the last decade suggest a complex link between inflammatory cells, coagulation, and the activation of platelets and their synergistic interaction to promote venous thrombosis. Inflammation is present throughout the process of venous thrombosis, and various metabolic pathways of erythrocytes, endothelial cells, and immune cells involved in venous thrombosis, including glucose metabolism, lipid metabolism, homocysteine metabolism, and oxidative stress, are associated with inflammation. While the metabolic microenvironment has been identified as a marker of malignancy, recent studies have revealed that for cancer thrombosis, alterations in the metabolic microenvironment appear to also be a potential risk. In this review, we discuss how the synergy between metabolism and thrombosis drives thrombotic disease. We also explore the great potential of anti-inflammatory strategies targeting venous thrombosis and the complex link between anti-inflammation and metabolism. Furthermore, we suggest how we can use our existing knowledge to reduce the risk of venous thrombosis.
Collapse
Affiliation(s)
- Zheng Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yiyu He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaoxin Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
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
|
Argueta DA, Tran H, Goel Y, Nguyen A, Nguyen J, Kiven SB, Chen C, Abdulla F, Vercellotti GM, Belcher JD, Gupta K. Mast cell extracellular trap formation underlies vascular and neural injury and hyperalgesia in sickle cell disease. Life Sci Alliance 2024; 7:e202402788. [PMID: 39242155 PMCID: PMC11381676 DOI: 10.26508/lsa.202402788] [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: 04/22/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024] Open
Abstract
Sickle cell disease (SCD) is the most common inherited monogenetic disorder. Chronic and acute pain are hallmark features of SCD involving neural and vascular injury and inflammation. Mast cells reside in the vicinity of nerve fibers and vasculature, but how they influence these structures remains unknown. We therefore examined the mechanism of mast cell activation in a sickle microenvironment replete with cell-free heme and inflammation. Mast cells exposed to this environment showed an explosion of nuclear contents with the release of citrullinated histones, suggestive of mast cell extracellular trap (MCET) release. MCETs interacted directly with the vasculature and nerve fibers, a cause of vascular and neural injury in sickle cell mice. MCET formation was dependent upon peptidylarginine deiminase 4 (PAD4). Inhibition of PAD4 ameliorated vasoocclusion, chronic and acute hyperalgesia, and inflammation in sickle mice. PAD4 activation may also underlie neutrophil trap formation in SCD, thus providing a novel target to treat the sequelae of vascular and neural injury in SCD.
Collapse
Affiliation(s)
- Donovan A Argueta
- https://ror.org/04gyf1771 Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Huy Tran
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Yugal Goel
- https://ror.org/04gyf1771 Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Aithanh Nguyen
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Julia Nguyen
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Stacy B Kiven
- https://ror.org/04gyf1771 Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Chunsheng Chen
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Fuad Abdulla
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Gregory M Vercellotti
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - John D Belcher
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Kalpna Gupta
- https://ror.org/04gyf1771 Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| |
Collapse
|
5
|
Huang Y, Jiang W, Zhou R. DAMP sensing and sterile inflammation: intracellular, intercellular and inter-organ pathways. Nat Rev Immunol 2024; 24:703-719. [PMID: 38684933 DOI: 10.1038/s41577-024-01027-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2024] [Indexed: 05/02/2024]
Abstract
Damage-associated molecular patterns (DAMPs) are endogenous molecules that are released from host cells as a result of cell death or damage. The release of DAMPs in tissues is associated with loss of tissue homeostasis. Sensing of DAMPs by innate immune receptors triggers inflammation, which can be beneficial in initiating the processes that restore tissue homeostasis but can also drive inflammatory diseases. In recent years, the sensing of intracellular DAMPs has received extensive attention in the field of sterile inflammation. However, emerging studies have shown that DAMPs that originate from neighbouring cells, and even from distal tissues or organs, also mediate sterile inflammatory responses. This multi-level sensing of DAMPs is crucial for intercellular, trans-tissue and trans-organ communication. Here, we summarize how DAMP-sensing receptors detect DAMPs from intracellular, intercellular or distal tissue and organ sources to mediate sterile inflammation. We also discuss the possibility of targeting DAMPs or their corresponding receptors to treat inflammatory diseases.
Collapse
Affiliation(s)
- Yi Huang
- Key Laboratory of Immune Response and Immunotherapy, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Wei Jiang
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Rongbin Zhou
- Key Laboratory of Immune Response and Immunotherapy, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China.
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
6
|
Haroun E, Lim SH, Dutta D. GBT1118, a Voxelotor Analog, Ameliorates Hepatopathy in Sickle Cell Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1581. [PMID: 39459368 PMCID: PMC11509622 DOI: 10.3390/medicina60101581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: In sickle cell disease (SCD), hepatopathy is a cumulative consequence of ischemia/reperfusion (I/R) injury from a vaso-occlusive crisis, tissue inflammation, and iron overload due to blood transfusion. Hepatopathy is a major contributing factor of shortened life span in SCD patients. We hypothesized that the voxelotor, a hemoglobin allosteric modifier, ameliorates sickle hepatopathy. Materials and Methods: Townes SCD mice and their controls were treated with either chow containing GBT1118, a voxelotor analog, or normal chow. We evaluated inflammation, fibrosis, apoptosis and ferroptosis in their livers using qPCR, ELISA, histology, and immunohistochemistry. Results: GBT1118 treatment resulted in reduced hemolysis, iron overload and inflammation in the liver of SCD mice. There were significant reductions in the liver enzyme levels and bile acids. Furthermore, GBT1118-treated mice exhibited reduced apoptosis, necrosis, and fibrosis. Increased ferroptosis as evident from elevated 4-hydroxynonenal (4-HNE) staining, malondialdehyde (MDA) levels, and expression of Ptgs2 and Slc7a11 mRNAs, were also significantly reduced after GBT1118 treatment. To explain the increased ferroptosis, we evaluated iron homeostasis markers in livers. SCD mice showed decreased expression of heme oxygenase-1, ferritin, hepcidin, and ferroportin mRNA levels. GBT1118 treatment significantly increased expressions of these genes. Conclusions: Our results suggest GBT1118 treatment in SCD confers the amelioration of sickle hepatopathy by reducing inflammation, fibrosis, apoptosis, iron overload and ferroptosis.
Collapse
Affiliation(s)
| | | | - Dibyendu Dutta
- Division of Hematology and Oncology, Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY 13210, USA; (E.H.); (S.H.L.)
| |
Collapse
|
7
|
Grujcic M, Milovanovic M, Nedeljkovic J, Jovanovic D, Arsenijevic D, Solovjova N, Stankovic V, Tanaskovic I, Arsenijevic A, Milovanovic J. The Possible Effects of Galectin-3 on Mechanisms of Renal and Hepatocellular Injury Induced by Intravascular Hemolysis. Int J Mol Sci 2024; 25:8129. [PMID: 39125698 PMCID: PMC11311984 DOI: 10.3390/ijms25158129] [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: 06/05/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Intravascular hemolysis is a central feature of congenital and acquired hemolytic anemias, complement disorders, infectious diseases, and toxemias. Massive and/or chronic hemolysis is followed by the induction of inflammation, very often with severe damage of organs, which enhances the morbidity and mortality of hemolytic diseases. Galectin-3 (Gal-3) is a β-galactoside-binding lectin that modulates the functions of many immune cells, thus affecting inflammatory processes. Gal-3 is also one of the main regulators of fibrosis. The role of Gal-3 in the development of different kidney and liver diseases and the potential of therapeutic Gal-3 inhibition have been demonstrated. Therefore, the objective of this review is to discuss the possible effects of Gal-3 on the process of kidney and liver damage induced by intravascular hemolysis, as well as to shed light on the potential therapeutic targeting of Gal-3 in intravascular hemolysis.
Collapse
Affiliation(s)
- Mirjana Grujcic
- Institute for Transfusiology and Hemobiology of Military Medical Academy, 11000 Belgrade, Serbia;
| | - Marija Milovanovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Jelena Nedeljkovic
- Department of Medical Statistics and Informatics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Danijela Jovanovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Dragana Arsenijevic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Natalija Solovjova
- Academy of Applied Studies Belgrade, The College of Health Science, Cara Dušana 254, 11080 Belgrade, Serbia;
| | - Vesna Stankovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Irena Tanaskovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Aleksandar Arsenijevic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Jelena Milovanovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
8
|
Stuart CM, Jacob C, Varatharaj A, Howard S, Chouhan JK, Teeling JL, Galea I. Mild Systemic Inflammation Increases Erythrocyte Fragility. Int J Mol Sci 2024; 25:7027. [PMID: 39000133 PMCID: PMC11241827 DOI: 10.3390/ijms25137027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/15/2024] [Accepted: 06/23/2024] [Indexed: 07/16/2024] Open
Abstract
There is growing evidence that inflammation impairs erythrocyte structure and function. We assessed the impact of mild systemic inflammation on erythrocyte fragility in three different settings. In order to investigate causation, erythrocyte osmotic fragility was measured in mice challenged with a live attenuated bacterial strain to induce low-grade systemic inflammation; a significant increase in erythrocyte osmotic fragility was observed. To gather evidence that systemic inflammation is associated with erythrocyte fragility in humans, two observational studies were conducted. First, using a retrospective study design, the relationship between reticulocyte-based surrogate markers of haemolysis and high-sensitivity C-reactive protein was investigated in 9292 healthy participants of the UK Biobank project. Secondly, we prospectively assessed the relationship between systemic inflammation (measured by the urinary neopterin/creatinine ratio) and erythrocyte osmotic fragility in a mixed population (n = 54) of healthy volunteers and individuals with long-term medical conditions. Both human studies were in keeping with a relationship between inflammation and erythrocyte fragility. Taken together, we conclude that mild systemic inflammation increases erythrocyte fragility and may contribute to haemolysis. Further research is needed to assess the molecular underpinnings of this pathway and the clinical implications in inflammatory conditions.
Collapse
Affiliation(s)
- Charlotte M. Stuart
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Carmen Jacob
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Aravinthan Varatharaj
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Sarah Howard
- Biological Sciences, Faculty of Life Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Joe K. Chouhan
- Biological Sciences, Faculty of Life Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Jessica L. Teeling
- Biological Sciences, Faculty of Life Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| |
Collapse
|
9
|
Ramadas N, Lowder K, Dutton J, Trebak F, Faes C, Griffin JH, Pawlinski R, Mosnier LO, Sparkenbaugh E. Biased agonism of protease-activated receptor-1 regulates thromboinflammation in murine sickle cell disease. Blood Adv 2024; 8:3272-3283. [PMID: 38640339 PMCID: PMC11226968 DOI: 10.1182/bloodadvances.2023011907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/21/2024] [Accepted: 04/08/2024] [Indexed: 04/21/2024] Open
Abstract
ABSTRACT Sickle cell disease (SCD) is a hereditary hemoglobinopathy marked by hemolytic anemia and vaso-occlusive events (VOEs). Chronic endothelial activation, inflammation, and coagulation activation contribute to vascular congestion, VOEs, and end-organ damage. Coagulation proteases such as thrombin and activated protein C (APC) modulate inflammation and endothelial dysfunction by activating protease-activated receptor 1 (PAR1), a G-protein-coupled receptor. Thrombin cleaves PAR1 at Arg41, while APC cleaves PAR1 at Arg46, initiating either proinflammatory or cytoprotective signaling, respectively, a signaling conundrum known as biased agonism. Our prior research established the role of thrombin and PAR1 in vascular stasis in an SCD mouse model. However, the role of APC and APC-biased PAR1 signaling in thrombin generation, inflammation, and endothelial activation in SCD remains unexplored. Inhibition of APC in SCD mice increased thrombin generation, inflammation, and endothelial activation during both steady state and tumor necrosis factor α challenge. To dissect the individual contributions of thrombin-PAR1 and APC-PAR1 signaling, we used transgenic mice with point mutations at 2 PAR1 cleavage sites, ArgR41Gln (R41Q) imparting insensitivity to thrombin and Arg46Gln (R46Q) imparting insensitivity to APC. Sickle bone marrow chimeras expressing PAR1-R41Q exhibited reduced thrombo-inflammatory responses compared with wild type PAR1 or PAR1-R46Q mice. These findings highlight the potential benefit of reducing thrombin-dependent PAR1 activation while preserving APC-PAR1 signaling in SCD thromboinflammation. These results also suggest that pharmacological strategies promoting biased PAR1 signaling could effectively mitigate vascular complications associated with SCD.
Collapse
Affiliation(s)
- Nirupama Ramadas
- Division of Hematology, Department of Medicine, Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Kailyn Lowder
- Division of Hematology, Department of Medicine, Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Joshua Dutton
- Division of Hematology, Department of Medicine, Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Fatima Trebak
- Division of Hematology, Department of Medicine, Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Camille Faes
- Division of Hematology, Department of Medicine, Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Rafal Pawlinski
- Division of Hematology, Department of Medicine, Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Laurent O. Mosnier
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Erica Sparkenbaugh
- Division of Hematology, Department of Medicine, Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
10
|
Rogers K, Alsawas M, Chapman J, Schlueter AJ, Knudson CM. Using the daily rate of rise in hemoglobin S to manage RBC depletion/exchange treatment in sickle cell disease. Transfusion 2024; 64:685-692. [PMID: 38506484 DOI: 10.1111/trf.17797] [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: 01/29/2024] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Red blood cell exchange is often used prophylactically in patients with sickle cell disease, with the goal to maintain hemoglobin S (HbS) below a target threshold level. We reviewed whether the daily "rate of rise" (RoR) in HbS that occurs between procedures can be used for patient management. For some patients not achieving their HbS goals despite efficient exchanges, the post-procedure hematocrit (Hct) target is increased to potentially suppress HbS production. This case series explores the utility of this approach, other clinical uses of the daily RoR in HbS, and the factors that influence it. STUDY DESIGN AND METHODS A total of 660 procedures from 24 patients undergoing prophylactic RBC depletion/exchange procedures were included. Laboratory values and clinical parameters were collected and used to calculate the daily RoR in HbS. Factors such as Hct or medications that might influence the RoR in HbS were evaluated. RESULTS The RoR in HbS varied widely between patients but remained relatively stable within individuals. Surprisingly, this value was not significantly influenced by changes in post-procedure Hct or concurrent hydroxyurea use. A patient's average RoR in HbS effectively predicted the pre-procedure HbS at the following visit (R2 = 0.65). DISCUSSION The RoR in HbS is a relatively consistent parameter for individual patients that is unaffected by medication use or procedural Hct targets and may be useful in determining intervals between procedures.
Collapse
Affiliation(s)
- Kai Rogers
- Department of Pathology, University of Iowa Health Care, Iowa City, Iowa, USA
| | - Mouaz Alsawas
- Department of Pathology, University of Iowa Health Care, Iowa City, Iowa, USA
| | - James Chapman
- Department of Pathology, University of Iowa Health Care, Iowa City, Iowa, USA
| | - Annette J Schlueter
- Department of Pathology, University of Iowa Health Care, Iowa City, Iowa, USA
| | | |
Collapse
|
11
|
Ross JT, Robles AJ, Mazer MB, Studer AC, Remy KE, Callcut RA. Cell-Free Hemoglobin in the Pathophysiology of Trauma: A Scoping Review. Crit Care Explor 2024; 6:e1052. [PMID: 38352942 PMCID: PMC10863949 DOI: 10.1097/cce.0000000000001052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Abstract
OBJECTIVES Cell-free hemoglobin (CFH) is a potent mediator of endothelial dysfunction, organ injury, coagulopathy, and immunomodulation in hemolysis. These mechanisms have been demonstrated in patients with sepsis, hemoglobinopathies, and those receiving transfusions. However, less is known about the role of CFH in the pathophysiology of trauma, despite the release of equivalent levels of free hemoglobin. DATA SOURCES Ovid MEDLINE, Embase, Web of Science Core Collection, and BIOSIS Previews were searched up to January 21, 2023, using key terms related to free hemoglobin and trauma. DATA EXTRACTION Two independent reviewers selected studies focused on hemolysis in trauma patients, hemoglobin breakdown products, hemoglobin-mediated injury in trauma, transfusion, sepsis, or therapeutics. DATA SYNTHESIS Data from the selected studies and their references were synthesized into a narrative review. CONCLUSIONS Free hemoglobin likely plays a role in endothelial dysfunction, organ injury, coagulopathy, and immune dysfunction in polytrauma. This is a compelling area of investigation as multiple existing therapeutics effectively block these pathways.
Collapse
Affiliation(s)
- James T Ross
- Department of Surgery, University of California Davis, Sacramento, CA
- The Blood, Heart, Lung, and Immunology Research Center, Case Western Reserve University, University Hospitals Cleveland, Cleveland, OH
| | - Anamaria J Robles
- Department of Surgery, University of California Davis, Sacramento, CA
| | - Monty B Mazer
- The Blood, Heart, Lung, and Immunology Research Center, Case Western Reserve University, University Hospitals Cleveland, Cleveland, OH
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, UH Rainbow Babies and Children's Hospital, Cleveland, OH
| | - Amy C Studer
- Blaisdell Medical Library, University of California Davis, Sacramento, CA
| | - Kenneth E Remy
- The Blood, Heart, Lung, and Immunology Research Center, Case Western Reserve University, University Hospitals Cleveland, Cleveland, OH
- Division of Pulmonary Critical Care Medicine, Department of Medicine, University Hospitals of Cleveland, Case Western Reserve School of Medicine, Cleveland, OH
| | - Rachael A Callcut
- Department of Surgery, University of California Davis, Sacramento, CA
| |
Collapse
|
12
|
Kamimura S, Smith M, Vogel S, Almeida LEF, Thein SL, Quezado ZMN. Mouse models of sickle cell disease: Imperfect and yet very informative. Blood Cells Mol Dis 2024; 104:102776. [PMID: 37391346 PMCID: PMC10725515 DOI: 10.1016/j.bcmd.2023.102776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/16/2023] [Indexed: 07/02/2023]
Abstract
The root cause of sickle cell disease (SCD) has been known for nearly a century, however, few therapies to treat the disease are available. Over several decades of work, with advances in gene editing technology and after several iterations of mice with differing genotype/phenotype relationships, researchers have developed humanized SCD mouse models. However, while a large body of preclinical studies has led to huge gains in basic science knowledge about SCD in mice, this knowledge has not led to the development of effective therapies to treat SCD-related complications in humans, thus leading to frustration with the paucity of translational progress in the SCD field. The use of mouse models to study human diseases is based on the genetic and phenotypic similarities between mouse and humans (face validity). The Berkeley and Townes SCD mice express only human globin chains and no mouse hemoglobin. With this genetic composition, these models present many phenotypic similarities, but also significant discrepancies that should be considered when interpreting preclinical studies results. Reviewing genetic and phenotypic similarities and discrepancies and examining studies that have translated to humans and those that have not, offer a better perspective of construct, face, and predictive validities of humanized SCD mouse models.
Collapse
Affiliation(s)
- Sayuri Kamimura
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Meghann Smith
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sebastian Vogel
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Luis E F Almeida
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zenaide M N Quezado
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
13
|
Nappi F. To Gain Insights into the Pathophysiological Mechanisms of the Thrombo-Inflammatory Process in the Atherosclerotic Plaque. Int J Mol Sci 2023; 25:47. [PMID: 38203218 PMCID: PMC10778759 DOI: 10.3390/ijms25010047] [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: 11/17/2023] [Revised: 12/17/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024] Open
Abstract
Thromboinflammation, the interplay between thrombosis and inflammation, is a significant pathway that drives cardiovascular and autoimmune diseases, as well as COVID-19. SARS-CoV-2 causes inflammation and blood clotting issues. Innate immune cells have emerged as key modulators of this process. Neutrophils, the most predominant white blood cells in humans, are strategically positioned to promote thromboinflammation. By releasing decondensed chromatin structures called neutrophil extracellular traps (NETs), neutrophils can initiate an organised cell death pathway. These structures are adorned with histones, cytoplasmic and granular proteins, and have cytotoxic, immunogenic, and prothrombotic effects that can hasten disease progression. Protein arginine deiminase 4 (PAD4) catalyses the citrullination of histones and is involved in the release of extracellular DNA (NETosis). The neutrophil inflammasome is also required for this process. Understanding the link between the immunological function of neutrophils and the procoagulant and proinflammatory activities of monocytes and platelets is important in understanding thromboinflammation. This text discusses how vascular blockages occur in thromboinflammation due to the interaction between neutrophil extracellular traps and ultra-large VWF (von Willebrand Factor). The activity of PAD4 is important for understanding the processes that drive thromboinflammation by linking the immunological function of neutrophils with the procoagulant and proinflammatory activities of monocytes and platelets. This article reviews how vaso-occlusive events in thrombo-inflammation occur through the interaction of neutrophil extracellular traps with von Willebrand factor. It highlights the relevance of PAD4 in neutrophil inflammasome assembly and neutrophil extracellular traps in thrombo-inflammatory diseases such as atherosclerosis and cardiovascular disease. Interaction between platelets, VWF, NETs and inflammasomes is critical for the progression of thromboinflammation in several diseases and was recently shown to be active in COVID-19.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| |
Collapse
|
14
|
Liu J, Zhang S, Jing Y, Zou W. Neutrophil extracellular traps in intracerebral hemorrhage: implications for pathogenesis and therapeutic targets. Metab Brain Dis 2023; 38:2505-2520. [PMID: 37486436 DOI: 10.1007/s11011-023-01268-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 07/19/2023] [Indexed: 07/25/2023]
Abstract
Intracerebral hemorrhage is a common neurological disease, and its pathological mechanism is complex. As the first recruited leukocyte subtype after intracerebral hemorrhage, neutrophils play an important role in tissue damage. In the past, it was considered that neutrophils performed their functions through phagocytosis, chemotaxis, and degranulation. In recent years, studies have found that neutrophils also have the function of secreting extracellular traps. Extracellular traps are fibrous structure composed of chromatin and granular proteins, which plays an important role in innate immunity. Studies have shown a large number of neutrophil extracellular traps in hematoma samples, plasma samples, and drainage samples after intracerebral hemorrhage. In this paper, we summarized the related mechanisms of neutrophil external traps and injury after intracerebral hemorrhage. Neutrophil extracellular traps are involved in the process of brain injury after intracerebral hemorrhage. The application of related inhibitors to inhibit the formation of neutrophil external traps or promote their dissolution can effectively alleviate the pathological damage caused by intracerebral hemorrhage.
Collapse
Affiliation(s)
- Jiawei Liu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Shuang Zhang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yunnan Jing
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Wei Zou
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| |
Collapse
|
15
|
Silva M, Faustino P. From Stress to Sick(le) and Back Again-Oxidative/Antioxidant Mechanisms, Genetic Modulation, and Cerebrovascular Disease in Children with Sickle Cell Anemia. Antioxidants (Basel) 2023; 12:1977. [PMID: 38001830 PMCID: PMC10669666 DOI: 10.3390/antiox12111977] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Sickle cell anemia (SCA) is a genetic disease caused by the homozygosity of the HBB:c.20A>T mutation, which results in the production of hemoglobin S (HbS). In hypoxic conditions, HbS suffers autoxidation and polymerizes inside red blood cells, altering their morphology into a sickle shape, with increased rigidity and fragility. This triggers complex pathophysiological mechanisms, including inflammation, cell adhesion, oxidative stress, and vaso-occlusion, along with metabolic alterations and endocrine complications. SCA is phenotypically heterogeneous due to the modulation of both environmental and genetic factors. Pediatric cerebrovascular disease (CVD), namely ischemic stroke and silent cerebral infarctions, is one of the most impactful manifestations. In this review, we highlight the role of oxidative stress in the pathophysiology of pediatric CVD. Since oxidative stress is an interdependent mechanism in vasculopathy, occurring alongside (or as result of) endothelial dysfunction, cell adhesion, inflammation, chronic hemolysis, ischemia-reperfusion injury, and vaso-occlusion, a brief overview of the main mechanisms involved is included. Moreover, the genetic modulation of CVD in SCA is discussed. The knowledge of the intricate network of altered mechanisms in SCA, and how it is affected by different genetic factors, is fundamental for the identification of potential therapeutic targets, drug development, and patient-specific treatment alternatives.
Collapse
Affiliation(s)
- Marisa Silva
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge (INSA), Av. Padre Cruz, 1649-016 Lisboa, Portugal;
| | - Paula Faustino
- Departamento de Genética Humana, Instituto Nacional de Saúde Doutor Ricardo Jorge (INSA), Av. Padre Cruz, 1649-016 Lisboa, Portugal;
- Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental (ISAMB), Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
- Laboratório Associado TERRA, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
16
|
Hamali HA. Hypercoagulability in Sickle Cell Disease: A Thrombo-Inflammatory Mechanism. Hemoglobin 2023; 47:205-214. [PMID: 38189099 DOI: 10.1080/03630269.2023.2301026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 12/28/2023] [Indexed: 01/09/2024]
Abstract
Sickle cell disease (SCD) is a group of inherited disorders characterized by the presence of abnormal hemoglobin S. Patients with SCD suffer from frequent episodes of anemia, chronic hemolysis, pain crisis, and vaso-occlusion. Additionally, SCD is associated with diverse and serious clinical complications, including thrombosis, which can lead to organ failure, increased morbidity, and eventually, mortality. SCD is known to be a hypercoagulable condition, and the cause of hypercoagulability is multifactorial, with the molecular basis of hemoglobin S being the main driver. The presence of hemoglobin S induces sickling of the RBCs and their subsequent hemolysis, as well as oxidative stress. Both of these processes can alter the hemostatic system, through the activation of platelets, coagulation system, and fibrinolysis, as well as depletion of coagulation inhibitors. These changes can also induce the formation of microvesicles and expression of tissue factor, leading to activation of WBCs, endothelial cell damage, and inflammatory response. Understanding the various factors that drive hypercoagulability as a thrombo-inflammatory mechanism in SCD can help provide explanations for the pathogenesis and other complications of the disease.
Collapse
Affiliation(s)
- Hassan A Hamali
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| |
Collapse
|
17
|
Ku TH, Ram-Mohan N, Zudock EJ, Abe R, Yang S. Neutrophil Extracellular Traps have DNAzyme activity that drives bactericidal potential. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563618. [PMID: 37961380 PMCID: PMC10634746 DOI: 10.1101/2023.10.23.563618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The mechanisms of bacterial killing by neutrophil extracellular traps (NETs) are unclear. DNA, the largest component of NETs is believed to merely be a scaffold with minimal antimicrobial activity through the charge of the backbone. Here, we report that NETs DNA is beyond a scaffold and produces hydroxyl free radicals through the spatially concentrated G-quadruplex/hemin DNAzyme complexes, driving bactericidal effects. Immunofluorescence staining showed colocalization of G-quadruplex and hemin in extruded NETs DNA, and Amplex UltraRed assay portrayed its peroxidase activity. Proximity labeling of bacteria revealed localized concentration of radicals resulting from NETs bacterial trapping. Ex vivo bactericidal assays revealed that G-quadruplex/hemin DNAzyme is the primary driver of bactericidal activity in NETs. NETs are DNAzymes that may have important biological consequences.
Collapse
Affiliation(s)
- Ti-Hsuan Ku
- Department of Emergency Medicine, Stanford University, Stanford, CA 94305, United States
| | - Nikhil Ram-Mohan
- Department of Emergency Medicine, Stanford University, Stanford, CA 94305, United States
| | - Elizabeth J Zudock
- Department of Emergency Medicine, Stanford University, Stanford, CA 94305, United States
| | - Ryuichiro Abe
- Department of Emergency Medicine, Stanford University, Stanford, CA 94305, United States
| | - Samuel Yang
- Department of Emergency Medicine, Stanford University, Stanford, CA 94305, United States
| |
Collapse
|
18
|
Cheng X, Yin X, Song J. Integrated analysis of NET-DNA receptor CCDC25 in malignant tumors: Pan-cancer analysis. Health Sci Rep 2023; 6:e1621. [PMID: 37841944 PMCID: PMC10568976 DOI: 10.1002/hsr2.1621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/17/2023] Open
Abstract
Background Previously, it was reported that the coiled-coil domain containing 25 (CCDC25) plays a role in the formation of neutrophil extracellular traps (NETs). This study systematically analyzed the expression profiles of CCDC25 in 30 different types of cancer and one type of blood cancer, acute myeloid leukemia. Methods The GTEx and CCLE databases were used to evaluate the distribution of CCDC25 expression in both normal tissue and cancer cell lines. A comparison was performed between normal tissue and tumor tissue to analyze the differential expression of CCDC25. We assessed the impact of CCDC25 on the clinical outlook in the TCGA pan-cancer data set by analyzing the Kaplan-Meier survival plot and conducting COX regression analysis. Moreover, the association between the expression levels of CCDC25 and the tumor microenvironment in multiple cancers was conducted. Additionally, the investigation also examined the link between CCDC25 and immune neoantigen, tumor mutational burden, microsatellite instability, mismatch repair genes (MMRs), HLA-related genes, and DNA methyltransferase (DNMT). Results CCDC25 was expressed in nearly all of the 31 normal tissues while exhibiting a moderate to low level of expression in cancer cell lines. While abnormal expression was detected in the majority of malignancies, there was no link found between elevated CCDC25 levels and overall survival, disease-free survival, recurrence-free survival, and disease-free interval in the TCGA comprehensive cancer data set. Nevertheless, the expression of CCDC25 exhibited a notable link with the infiltration levels of activated CD4 memory T cells, quiescent mast cells, dendritic cells in an activated state, T cells that assist in follicle development, M2 macrophages, and neutrophils in various tumors. Conclusions In most cancers, the results indicate that there is no link between CCDC25 and prognosis. However, CCDC25 can be targeted for therapeutic purposes concerning metastasis and immune infiltration.
Collapse
Affiliation(s)
- Xianlin Cheng
- Department of Oral and Maxillofacial SurgeryGuizhou Provincial People's HospitalGuizhouChina
| | - Xinhai Yin
- Department of Oral and Maxillofacial SurgeryGuizhou Provincial People's HospitalGuizhouChina
| | - Jukun Song
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Guizhou Medical UniversityGuiyangChina
| |
Collapse
|
19
|
Orrico F, Laurance S, Lopez AC, Lefevre SD, Thomson L, Möller MN, Ostuni MA. Oxidative Stress in Healthy and Pathological Red Blood Cells. Biomolecules 2023; 13:1262. [PMID: 37627327 PMCID: PMC10452114 DOI: 10.3390/biom13081262] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Red cell diseases encompass a group of inherited or acquired erythrocyte disorders that affect the structure, function, or production of red blood cells (RBCs). These disorders can lead to various clinical manifestations, including anemia, hemolysis, inflammation, and impaired oxygen-carrying capacity. Oxidative stress, characterized by an imbalance between the production of reactive oxygen species (ROS) and the antioxidant defense mechanisms, plays a significant role in the pathophysiology of red cell diseases. In this review, we discuss the most relevant oxidant species involved in RBC damage, the enzymatic and low molecular weight antioxidant systems that protect RBCs against oxidative injury, and finally, the role of oxidative stress in different red cell diseases, including sickle cell disease, glucose 6-phosphate dehydrogenase deficiency, and pyruvate kinase deficiency, highlighting the underlying mechanisms leading to pathological RBC phenotypes.
Collapse
Affiliation(s)
- Florencia Orrico
- Laboratorio de Fisicoquímica Biológica, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay; (F.O.); (A.C.L.); (M.N.M.)
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay;
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Sandrine Laurance
- Université Paris Cité and Université des Antilles, UMR_S1134, BIGR, Inserm, F-75014 Paris, France; (S.L.); (S.D.L.)
| | - Ana C. Lopez
- Laboratorio de Fisicoquímica Biológica, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay; (F.O.); (A.C.L.); (M.N.M.)
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay;
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Sophie D. Lefevre
- Université Paris Cité and Université des Antilles, UMR_S1134, BIGR, Inserm, F-75014 Paris, France; (S.L.); (S.D.L.)
| | - Leonor Thomson
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay;
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Matias N. Möller
- Laboratorio de Fisicoquímica Biológica, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay; (F.O.); (A.C.L.); (M.N.M.)
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Mariano A. Ostuni
- Université Paris Cité and Université des Antilles, UMR_S1134, BIGR, Inserm, F-75014 Paris, France; (S.L.); (S.D.L.)
| |
Collapse
|
20
|
Van Avondt K, Schimmel M, Bulder I, van Mierlo G, Nur E, van Bruggen R, Biemond BJ, Luken BM, Zeerleder S. Circulating Iron in Patients with Sickle Cell Disease Mediates the Release of Neutrophil Extracellular Traps. Transfus Med Hemother 2023; 50:321-329. [PMID: 37767280 PMCID: PMC10521246 DOI: 10.1159/000526760] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 08/24/2022] [Indexed: 09/29/2023] Open
Abstract
Introduction Neutrophils promote chronic inflammation and release neutrophil extracellular traps (NETs) that can drive inflammatory responses. Inflammation influences progression of sickle cell disease (SCD), and a role for NETs has been suggested in the onset of vaso-occlusive crisis (VOC). We aimed to identify factors in the circulation of these patients that provoke NET release, with a focus on triggers associated with hemolysis. Methods Paired serum and plasma samples during VOC and steady state of 18 SCD patients (HbSS/HbSβ0-thal and HbSC/HbSβ+-thal) were collected. Cell-free heme, hemopexin, and labile plasma iron have been measured in the plasma samples of the SCD patients. NETs formation by human neutrophils from healthy donors induced by serum of SCD patients was studied using confocal microscopy and staining for extracellular DNA using Sytox, followed by quantification of surface coverage using ImageJ. Results Eighteen patients paired samples obtained during VOC and steady state were available (11 HbSS/HbSβ0-thal and 7 HbSC/HbSβ+-thal). We observed high levels of systemic heme and iron, concomitant with low levels of the heme-scavenger hemopexin in sera of patients with SCD, both during VOC and in steady state. In our in vitro experiments, neutrophils released NETs when exposed to sera from SCD patients. The release of NETs was associated with high levels of circulating iron in these sera. Although hemin triggered NET formation in vitro, addition of hemopexin to scavenge heme did not suppress NET release in SCD sera. By contrast, the iron scavengers deferoxamine and apotransferrin attenuated NET formation in a significant proportion of SCD sera. Discussion Our results suggest that redox-active iron in the circulation of non-transfusion-dependent SCD patients activates neutrophils to release NETs, and hence, exerts a direct pro-inflammatory effect. Thus, we propose that chelation of iron requires further investigation as a therapeutic strategy in SCD.
Collapse
Affiliation(s)
- Kristof Van Avondt
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
- Institute of Experimental Pathology, Center for Molecular Biology of Inflammation, University Medical Center Münster, University of Münster, Münster, Germany
| | - Marein Schimmel
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Ingrid Bulder
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Gerard van Mierlo
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Erfan Nur
- Department of Hematology, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Robin van Bruggen
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Bart J. Biemond
- Department of Hematology, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Brenda M. Luken
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Sacha Zeerleder
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Division of Internal Medicine, Kantonsspital Lucerne, Lucerne and University of Berne, Berne, Switzerland
| |
Collapse
|
21
|
de Lima F, Hounkpe BW, de Moraes CRP, Borba-Junior IT, Costa FF, De Paula EV. Safety and feasibility of the gene transfer of hemopexin for conditions with increased free heme. Exp Biol Med (Maywood) 2023; 248:1103-1111. [PMID: 37452705 PMCID: PMC10583756 DOI: 10.1177/15353702231182199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/16/2023] [Indexed: 07/18/2023] Open
Abstract
Heme is a fundamental molecule for several biological processes, but when released in the extracellular space such as in hemolytic diseases, it can be toxic to cells and tissues. Hemopexin (HPX) is a circulating protein responsible for removing free heme from the circulation, whose levels can be severely depleted in conditions such as sickle cell diseases. Accordingly, increasing HPX levels represents an attractive strategy to mitigate the deleterious effects of heme in these conditions. Gene transfer of liver-produced proteins with adeno-associated virus (AAV) has been shown to be an effective and safety strategy in animal and human studies mainly in hemophilia. Here, we report the feasibility of increasing HPX levels using an AAV8 vector expressing human HPX (hHPX). C57Bl mice were injected with escalating doses of our vector, and expression was assessed by enzyme immunoassay (ELISA), Western blot, and quantitative polymerase chain reaction (qPCR). In addition, the biological activity of transgenic hHPX was confirmed using two different models of heme challenge consisting of serial heme injections or phenylhydrazine-induced hemolysis. Sustained expression of hHPX was confirmed for up to 26 weeks in plasma. Expression was dose-dependent and not associated with clinical signs of toxicity. hHPX levels were significantly reduced by heme infusions and phenylhydrazine-induced hemolysis. No clinical toxicity or laboratory signs of liver damage were observed in preliminary short-term heme challenge studies. Our results confirm that long-term expression of hHPX is feasible and safe in mice, even in the presence of heme overload. Additional studies are needed to explore the effect of transgenic HPX protein in animal models of chronic hemolysis.
Collapse
Affiliation(s)
- Franciele de Lima
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | | | | | | | - Fernando Ferreira Costa
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
- Hematology and Hemotherapy Center, University of Campinas, Campinas 13083-878, Brazil
| | - Erich V De Paula
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
- Hematology and Hemotherapy Center, University of Campinas, Campinas 13083-878, Brazil
| |
Collapse
|
22
|
Chai CZ, Ho UC, Kuo LT. Systemic Inflammation after Aneurysmal Subarachnoid Hemorrhage. Int J Mol Sci 2023; 24:10943. [PMID: 37446118 DOI: 10.3390/ijms241310943] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/16/2023] [Accepted: 06/18/2023] [Indexed: 07/15/2023] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is one of the most severe neurological disorders, with a high mortality rate and severe disabling functional sequelae. Systemic inflammation following hemorrhagic stroke may play an important role in mediating intracranial and extracranial tissue damage. Previous studies showed that various systemic inflammatory biomarkers might be useful in predicting clinical outcomes. Anti-inflammatory treatment might be a promising therapeutic approach for improving the prognosis of patients with aSAH. This review summarizes the complicated interactions between the nervous system and the immune system.
Collapse
Affiliation(s)
- Chang-Zhang Chai
- Department of Medical Education, National Taiwan University, School of Medicine, Taipei 100, Taiwan
| | - Ue-Cheung Ho
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital Yunlin Branch, Yunlin 640, Taiwan
| | - Lu-Ting Kuo
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital Yunlin Branch, Yunlin 640, Taiwan
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan
| |
Collapse
|
23
|
Gour A, Kour D, Pandian R, Bhardwaj M, Sawant SD, Kumar A, Nandi U. Ellagic Acid Exerts Dual Action to Curb the Pathophysiological Manifestations of Sickle Cell Disease and Attenuate the Hydroxyurea-Induced Myelosuppression in Berkeley Mice. ACS Pharmacol Transl Sci 2023; 6:868-877. [PMID: 37325443 PMCID: PMC10262317 DOI: 10.1021/acsptsci.3c00026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Indexed: 06/17/2023]
Abstract
The use of adjuvant therapy is an attractive approach to manage sickle cell disease (SCD) symptomatically. The present study aimed to investigate the potential of ellagic acid as an adjuvant therapy with hydroxyurea (HU), a key drug for SCD with myelosuppressive toxic effects. A panel of experiments was performed using SCD patient's blood (ex vivo) and transgenic mice model of SCD (in vivo). Ellagic acid exhibited the following beneficial pharmacological actions: (a) potent anti-sickling, polymerization inhibitory, and inherent non-hemolytic activity; (b) pronounced action to abrogate HU-induced neutropenia and to improve key hematological parameters during SCD (RBC, Hb, platelet levels); (c) considerable action to foster vascular tone (L-proline); (d) marked attenuating effect against oxidative stress (nitrotyrosine, hypoxanthine, MDA, GSH); (e) substantial inhibitory role against inflammation (analgesic activity and regulation of hemin, TNF-α, IL-1β, NF-κB/IκBα); (f) remarkable outcome of declining vaso-occlusive crisis (P-selectin, ERK1/2); (g) notable shielding deed against elevated biochemical marker for organ toxicity (creatinine); (h) noticeably prevented histopathological alterations of the spleen. Additionally, the pharmacokinetic study results of HU in the presence and absence of ellagic acid using a mouse model demonstrate that ellagic acid could be safely co-administered with HU. Overall findings suggest that ellagic acid is a promising candidate for adjuvant therapy in SCD based on its own significant ability against SCD and potentiating capability of HU action via targeting improvement at the various stages of pathophysiological complications during SCD and minimizing HU-induced toxicological manifestations.
Collapse
Affiliation(s)
- Abhishek Gour
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Dilpreet Kour
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ramajayan Pandian
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mahir Bhardwaj
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sanghapal D. Sawant
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Natural
Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Ajay Kumar
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Utpal Nandi
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
24
|
Nasir NJM, Heemskerk H, Jenkins J, Hamadee NH, Bunte R, Tucker-Kellogg L. Myoglobin-derived iron causes wound enlargement and impaired regeneration in pressure injuries of muscle. eLife 2023; 12:85633. [PMID: 37267120 DOI: 10.7554/elife.85633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/25/2023] [Indexed: 06/04/2023] Open
Abstract
The reasons for poor healing of pressure injuries are poorly understood. Vascular ulcers are worsened by extracellular release of hemoglobin, so we examined the impact of myoglobin (Mb) iron in murine muscle pressure injuries (mPI). Tests used Mb-knockout or treatment with deferoxamine iron chelator (DFO). Unlike acute injuries from cardiotoxin, mPI regenerated poorly with a lack of viable immune cells, persistence of dead tissue (necro-slough), and abnormal deposition of iron. However, Mb-knockout or DFO-treated mPI displayed a reversal of the pathology: decreased tissue death, decreased iron deposition, decrease in markers of oxidative damage, and higher numbers of intact immune cells. Subsequently, DFO treatment improved myofiber regeneration and morphology. We conclude that myoglobin iron contributes to tissue death in mPI. Remarkably, a large fraction of muscle death in untreated mPI occurred later than, and was preventable by, DFO treatment, even though treatment started 12 hr after pressure was removed. This demonstrates an opportunity for post-pressure prevention to salvage tissue viability.
Collapse
Affiliation(s)
- Nurul Jannah Mohamed Nasir
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Hans Heemskerk
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- BioSyM and CAMP Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, CREATE, Singapore, Singapore
| | - Julia Jenkins
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | | | - Ralph Bunte
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Lisa Tucker-Kellogg
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore
- BioSyM and CAMP Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, CREATE, Singapore, Singapore
| |
Collapse
|
25
|
Papadakis E, Brenner B. Thrombosis in Pregnant Women with Hemolytic Anemia. Semin Thromb Hemost 2023; 49:364-370. [PMID: 36410400 DOI: 10.1055/s-0042-1758819] [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: 11/23/2022]
Abstract
Hemolytic anemias are a group of uncommon disorders affecting both genders, frequently occurring at the reproductive age. While a link between hemolysis and hypercoagulability has been suggested based on the elucidation of certain involved pathophysiological mechanisms, the extent of thrombotic risk in pregnant women with hemolytic anemia remains debatable. Due to the paucity of pregnancy-related data, risk assessment of gestations in women with hemolytic anemia is complicated. This review will highlight the latest advances in the diagnosis and management of these challenging disorders in pregnancy.
Collapse
Affiliation(s)
| | - Benjamin Brenner
- Department of Hematology, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
26
|
Sparkenbaugh EM, Henderson MW, Miller-Awe M, Abrams C, Ilich A, Trebak F, Ramadas N, Vital S, Bohinc D, Bane KL, Chen C, Patel M, Wallisch M, Renné T, Gruber A, Cooley B, Gailani D, Kasztan M, Vercellotti GM, Belcher JD, Gavins FE, Stavrou EX, Key NS, Pawlinski R. Factor XII contributes to thrombotic complications and vaso-occlusion in sickle cell disease. Blood 2023; 141:1871-1883. [PMID: 36706361 PMCID: PMC10122107 DOI: 10.1182/blood.2022017074] [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: 05/20/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/28/2023] Open
Abstract
A hypercoagulable state, chronic inflammation, and increased risk of venous thrombosis and stroke are prominent features in patients with sickle cell disease (SCD). Coagulation factor XII (FXII) triggers activation of the contact system that is known to be involved in both thrombosis and inflammation, but not in physiological hemostasis. Therefore, we investigated whether FXII contributes to the prothrombotic and inflammatory complications associated with SCD. We found that when compared with healthy controls, patients with SCD exhibit increased circulating biomarkers of FXII activation that are associated with increased activation of the contact pathway. We also found that FXII, but not tissue factor, contributes to enhanced thrombin generation and systemic inflammation observed in sickle cell mice challenged with tumor necrosis factor α. In addition, FXII inhibition significantly reduced experimental venous thrombosis, congestion, and microvascular stasis in a mouse model of SCD. Moreover, inhibition of FXII attenuated brain damage and reduced neutrophil adhesion to the brain vasculature of sickle cell mice after ischemia/reperfusion induced by transient middle cerebral artery occlusion. Finally, we found higher FXII, urokinase plasminogen activator receptor, and αMβ2 integrin expression in neutrophils of patients with SCD compared with healthy controls. Our data indicate that targeting FXII effectively reduces experimental thromboinflammation and vascular complications in a mouse model of SCD, suggesting that FXII inhibition may provide a safe approach for interference with inflammation, thrombotic complications, and vaso-occlusion in patients with SCD.
Collapse
Affiliation(s)
- Erica M. Sparkenbaugh
- Division of Hematology and Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Michael W. Henderson
- Division of Hematology and Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Megan Miller-Awe
- Division of Hematology and Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Christina Abrams
- Division of Hematology and Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Anton Ilich
- Division of Hematology and Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Fatima Trebak
- Division of Hematology and Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Nirupama Ramadas
- Division of Hematology and Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Shantel Vital
- Louisiana State University Health Sciences Center, Shreveport, LA
| | - Dillon Bohinc
- Hematology and Oncology Division, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Kara L. Bane
- Hematology and Oncology Division, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Chunsheng Chen
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Margi Patel
- Division of Hematology-Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL
| | | | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Brian Cooley
- Division of Hematology and Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | - Malgorzata Kasztan
- Division of Hematology-Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL
| | - Gregory M. Vercellotti
- 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
| | - Felicity E. Gavins
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine, Brunel University London, London, United Kingdom
| | - Evi X. Stavrou
- Hematology and Oncology Division, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
- Department of Medicine, Section of Hematology-Oncology, Louis Stokes Veterans Administration Medical Center, Cleveland, OH
| | - Nigel S. Key
- Division of Hematology and Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Rafal Pawlinski
- Division of Hematology and Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
27
|
Hamzaoui A, Louhaichi S, Hamdi B. [Lung manifestations of sickle-cell disease]. Rev Mal Respir 2023:S0761-8425(23)00107-9. [PMID: 37059617 DOI: 10.1016/j.rmr.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 03/04/2023] [Indexed: 04/16/2023]
Abstract
Sickle-cell disease is an autosomal recessive genetic disorder of hemoglobin that causes systemic damage. Hypoxia is the main actor of sickle-cell disease. It initiates acutely the pathogenic cascade leading to tissue damages that in turn induce chronic hypoxia. Lung lesions represent the major risk of morbidity and mortality. Management of sickle-cell disease requires a tight collaboration between hematologists, intensivists and chest physicians. Recurrent episodes of thrombosis and hemolysis characterize the disease. New therapeutic protocols, associating hydroxyurea, transfusion program and stem cell transplantation in severe cases allow a prolonged survival until the fifth decade. However, recurrent pain, crisis, frequent hospital admissions due to infection, anemia or acute chest syndrome and chronic complications leading to organ deficiencies degrade the patients' quality of life. In low-income countries where the majority of sickle-cell patients are living, the disease is still associated with a high mortality in childhood. This paper focuses on acute chest syndrome and chronic lung manifestations.
Collapse
Affiliation(s)
- A Hamzaoui
- Pavillon B/LR19SP02, hôpital Abderrahmen-Mami, 2080 Ariana, Tunisie; Faculté de médecine de Tunis, 1006 Tunis, Tunisie.
| | - S Louhaichi
- Pavillon B/LR19SP02, hôpital Abderrahmen-Mami, 2080 Ariana, Tunisie; Faculté de médecine de Tunis, 1006 Tunis, Tunisie
| | - B Hamdi
- Pavillon B/LR19SP02, hôpital Abderrahmen-Mami, 2080 Ariana, Tunisie; Faculté de médecine de Tunis, 1006 Tunis, Tunisie
| |
Collapse
|
28
|
Saxena P, Muthu J. COVID-19 and Sickle Cell Disease: Two Independent Risk Factors for Venous Thromboembolism. Cureus 2023; 15:e37226. [PMID: 37159776 PMCID: PMC10163976 DOI: 10.7759/cureus.37226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been widely documented as a multi-systemic illness and associated with an increased incidence of thromboses. Likewise, sickle cell disease (SCD) is a hematologic disease responsible for widespread effects on the vasculature and is also associated with elevated thrombotic risk. In this review, we examine the incidence rates of venous thromboembolism (VTE) in SCD and COVID-19 independently and review the mechanisms of coagulopathy associated with both diseases. We describe the possible associations and commonalities between VTE mechanisms, as both diseases cause widespread inflammation that influences each tenet of Virchow's triad. We also discuss current anticoagulation guideline recommendations for the prevention of VTE events in each of these diseases. We report on current literature to date describing rates of VTE in SCD-COVID-19 patients and outline prospective areas of research to further understand the possible synergistic influence of coagulopathy in these patients. The association between SCD and COVID-19 remains a largely under-researched area of coagulopathy in current hematology and thrombotic literature, and our report lays out potential future prospects in the field.
Collapse
|
29
|
Sesti-Costa R, Costa FF, Conran N. Role of Macrophages in Sickle Cell Disease Erythrophagocytosis and Erythropoiesis. Int J Mol Sci 2023; 24:ijms24076333. [PMID: 37047304 PMCID: PMC10094208 DOI: 10.3390/ijms24076333] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Sickle cell disease (SCD) is an inherited blood disorder caused by a β-globin gene point mutation that results in the production of sickle hemoglobin that polymerizes upon deoxygenation, causing the sickling of red blood cells (RBCs). RBC deformation initiates a sequence of events leading to multiple complications, such as hemolytic anemia, vaso-occlusion, chronic inflammation, and tissue damage. Macrophages participate in extravascular hemolysis by removing damaged RBCs, hence preventing the release of free hemoglobin and heme, and triggering inflammation. Upon erythrophagocytosis, macrophages metabolize RBC-derived hemoglobin, activating mechanisms responsible for recycling iron, which is then used for the generation of new RBCs to try to compensate for anemia. In the bone marrow, macrophages can create specialized niches, known as erythroblastic islands (EBIs), which regulate erythropoiesis. Anemia and inflammation present in SCD may trigger mechanisms of stress erythropoiesis, intensifying RBC generation by expanding the number of EBIs in the bone marrow and creating new ones in extramedullary sites. In the current review, we discuss the distinct mechanisms that could induce stress erythropoiesis in SCD, potentially shifting the macrophage phenotype to an inflammatory profile, and changing their supporting role necessary for the proliferation and differentiation of erythroid cells in the disease. The knowledge of the soluble factors, cell surface and intracellular molecules expressed by EBI macrophages that contribute to begin and end the RBC’s lifespan, as well as the understanding of their signaling pathways in SCD, may reveal potential targets to control the pathophysiology of the disease.
Collapse
|
30
|
Platelet activation and ferroptosis mediated NETosis drives heme induced pulmonary thrombosis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166688. [PMID: 36925054 DOI: 10.1016/j.bbadis.2023.166688] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023]
Abstract
Cell-free heme (CFH) is a product of hemoglobin, myoglobin and hemoprotein degradation, which is a hallmark of pathologies associated with extensive hemolysis and tissue damage. CHF and iron collectively induce cytokine storm, lung injury, respiratory distress and infection susceptibility in the lungs suggesting their key role in the progression of lung disease pathology. We have previously demonstrated that heme-mediated reactive oxygen species (ROS) induces platelet activation and ferroptosis. However, interaction of ferroptotic platelets and neutrophils, the mechanism of action and associated complications remain unclear. In this study, we demonstrate that heme-induced P-selectin expression and Phosphatidylserine (PS) externalization in platelets via ASK-1-inflammasome axis increases platelet-neutrophil aggregates in circulation, resulting in Neutrophil extracellular traps (NET) formation in vitro and in vivo. Further, heme-induced platelet activation in mice increased platelet-neutrophil aggregates and accumulation of NETs in the lungs causing pulmonary damage. Thus, connecting CFH-mediated platelet activation to NETosis and pulmonary thrombosis. As lung infections induce acute respiratory stress, thrombosis and NETosis, we propose that heme -mediated platelet activation and ferroptosis might be crucial in such clinical manifestations. Further, considering the ability of redox modulators and ferroptosis inhibitors like FS-1, Lpx-1 and DFO to inhibit heme-induced ferroptotic platelets-mediated NETosis and pulmonary thrombosis. They could be potential adjuvant therapy to regulate respiratory distress-associated clinical complications.
Collapse
|
31
|
Recchiuti A, Federti E, Matte A, Mazzi F, Ceolan J, Porreca A, Di Nicola M, Menotti S, Alivernini S, De Franceschi L. Impaired pro-resolving mechanisms promote abnormal NETosis, fueling autoimmunity in sickle cell disease. Am J Hematol 2023; 98:E45-E48. [PMID: 36540948 DOI: 10.1002/ajh.26797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/21/2022] [Accepted: 11/26/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Antonio Recchiuti
- Department of Medical, Oral and Biotechnological Sciences, Laboratory of Molecular Medicine, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Enrica Federti
- Department of Medicine, University of Verona & AOUI Verona, Verona, Italy
| | - Alessandro Matte
- Department of Medicine, University of Verona & AOUI Verona, Verona, Italy
| | - Filippo Mazzi
- Department of Medicine, University of Verona & AOUI Verona, Verona, Italy
| | - Jacopo Ceolan
- Department of Medicine, University of Verona & AOUI Verona, Verona, Italy
| | - Annamaria Porreca
- Department of Medical, Oral and Biotechnological Sciences, Laboratory of Biostatistics, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Marta Di Nicola
- Department of Medical, Oral and Biotechnological Sciences, Laboratory of Biostatistics, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Sofia Menotti
- Department of Medicine, University of Verona & AOUI Verona, Verona, Italy
| | - Stefano Alivernini
- Institute of Rheumatology, Università Cattolica del Sacro Cuore & Fondazione Policlinico Gemelli, Rome, Italy
| | | |
Collapse
|
32
|
Abstract
While neutrophils are the main effectors of protective innate immune responses, they are also key players in inflammatory pathologies. Sickle cell disease (SCD) is a genetic blood disorder in which red blood cells (RBCs) are constantly destroyed in the circulation which generates a highly inflammatory environment that culminates in vascular occlusions. Vaso-occlusion is the hallmark of SCD and a predictor of disease severity. Neutrophils initiate and propagate SCD-related vaso-occlusion through adhesive interactions with the activated and dysfunctional endothelium, sickle RBCs, and platelets, leading to acute and chronic complications that progress to irreversible organ damage and ultimately death. The use of SCD humanized mouse models, in combination with in vivo imaging techniques, has emerged as a fundamental tool to understand the dynamics of neutrophils under complex inflammatory contexts and their contribution to vascular injury in SCD. In this review, we discuss the various mechanisms by which circulating neutrophils sense and respond to the wide range of stimuli present in the blood of SCD patients and mice. We argue that the central role of neutrophils in SCD can be rationalized to develop targets for the management of clinical complications in SCD patients.
Collapse
Affiliation(s)
- Lidiane S Torres
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Andrés Hidalgo
- Area of Cell and Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
33
|
Gbotosho OT, Gollamudi J, Hyacinth HI. The Role of Inflammation in The Cellular and Molecular Mechanisms of Cardiopulmonary Complications of Sickle Cell Disease. Biomolecules 2023; 13:381. [PMID: 36830749 PMCID: PMC9953727 DOI: 10.3390/biom13020381] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Cardiopulmonary complications remain the major cause of mortality despite newer therapies and improvements in the lifespan of patients with sickle cell disease (SCD). Inflammation has been identified as a major risk modifier in the pathogenesis of SCD-associated cardiopulmonary complications in recent mechanistic and observational studies. In this review, we discuss recent cellular and molecular mechanisms of cardiopulmonary complications in SCD and summarize the most recent evidence from clinical and laboratory studies. We emphasize the role of inflammation in the onset and progression of these complications to better understand the underlying pathobiological processes. We also discuss future basic and translational research in addressing questions about the complex role of inflammation in the development of SCD cardiopulmonary complications, which may lead to promising therapies and reduce morbidity and mortality in this vulnerable population.
Collapse
Affiliation(s)
- Oluwabukola T. Gbotosho
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267-0525, USA
| | - Jahnavi Gollamudi
- Division of Hematology & Oncology, Department of Internal Medicine, 3125 Eden Avenue, ML 0562, Cincinnati, OH 45219-0562, USA
| | - Hyacinth I. Hyacinth
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267-0525, USA
| |
Collapse
|
34
|
Planchais C, Noe R, Gilbert M, Lecerf M, Kaveri SV, Lacroix-Desmazes S, Roumenina LT, Dimitrov JD. Oxidized hemoglobin triggers polyreactivity and autoreactivity of human IgG via transfer of heme. Commun Biol 2023; 6:168. [PMID: 36774392 PMCID: PMC9922299 DOI: 10.1038/s42003-023-04535-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/26/2023] [Indexed: 02/13/2023] Open
Abstract
Intravascular hemolysis occurs in diverse pathological conditions. Extracellular hemoglobin and heme have strong pro-oxidative and pro-inflammatory potentials that can contribute to the pathology of hemolytic diseases. However, many of the effects of extracellular hemoglobin and heme in hemolytic diseases are still not well understood. Here we demonstrate that oxidized hemoglobin (methemoglobin) can modify the antigen-binding characteristics of human immunoglobulins. Thus, incubation of polyclonal or some monoclonal human IgG in the presence of methemoglobin results in an appearance of binding reactivities towards distinct unrelated self-proteins, including the protein constituent of hemoglobin i.e., globin. We demonstrate that a transfer of heme from methemoglobin to IgG is indispensable for this acquisition of antibody polyreactivity. Our data also show that only oxidized form of hemoglobin have the capacity to induce polyreactivity of antibodies. Site-directed mutagenesis of a heme-sensitive human monoclonal IgG1 reveals details about the mechanism of methemoglobin-induced antigen-binding polyreactivity. Further here we assess the kinetics and thermodynamics of interaction of a heme-induced polyreactive human antibody with hemoglobin and myoglobin. Taken together presented data contribute to a better understanding of the functions of extracellular hemoglobin in the context of hemolytic diseases.
Collapse
Affiliation(s)
- Cyril Planchais
- Laboratory of Humoral Immunology, Institut Pasteur, Université Paris Cité, INSERM U1222, 75015 Paris, France
| | - Remi Noe
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Marie Gilbert
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Maxime Lecerf
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Srini V. Kaveri
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Sébastien Lacroix-Desmazes
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Lubka T. Roumenina
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Jordan D. Dimitrov
- grid.4444.00000 0001 2112 9282Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| |
Collapse
|
35
|
Wiatr M, Hadzhieva M, Lecerf M, Noé R, Justesen S, Lacroix-Desmazes S, Dragon-Durey MA, Dimitrov JD. Hyperoxidized Species of Heme Have a Potent Capacity to Induce Autoreactivity of Human IgG Antibodies. Int J Mol Sci 2023; 24:ijms24043416. [PMID: 36834827 PMCID: PMC9960230 DOI: 10.3390/ijms24043416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
The interaction of some human antibodies with heme results in posttranslational acquisition of binding to various self- and pathogen-derived antigens. The previous studies on this phenomenon were performed with oxidized heme (Fe3+). In the present study, we elucidated the effect of other pathologically relevant species of heme, i.e., species that were formed after contact of heme with oxidizing agents such as hydrogen peroxide, situations in which heme's iron could acquire higher oxidation states. Our data reveal that hyperoxidized species of heme have a superior capacity to heme (Fe3+) in triggering the autoreactivity of human IgG. Mechanistic studies demonstrated that oxidation status of iron was of critical importance for the heme's effect on antibodies. We also demonstrated that hyperoxidized heme species interacted at higher affinities with IgG and that this binding occurred through a different mechanism as compared to heme (Fe3+). Regardless of their profound functional impact on the antigen-binding properties of antibodies, hyperoxidized species of heme did not affect Fc-mediated functions of IgG, such as binding to the neonatal Fc receptor. The obtained data contribute to a better understanding of the pathophysiological mechanism of hemolytic diseases and of the origin of elevated antibody autoreactivity in patients with some hemolytic disorders.
Collapse
Affiliation(s)
- Marie Wiatr
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Maya Hadzhieva
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Maxime Lecerf
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Rémi Noé
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Sune Justesen
- Immunitrack Aps, Lersoe Park Alle 42, 2100 Copenhagen, Denmark
| | - Sébastien Lacroix-Desmazes
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
| | - Marie-Agnès Dragon-Durey
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Service d’Immunologie Biologique, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 75610 Paris, France
| | - Jordan D. Dimitrov
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Correspondence: ; Tel.: +33-144-278206
| |
Collapse
|
36
|
Salgar S, Bolívar BE, Flanagan JM, Anum SJ, Bouchier-Hayes L. The NLRP3 inflammasome fires up heme-induced inflammation in hemolytic conditions. Transl Res 2023; 252:34-44. [PMID: 36041706 DOI: 10.1016/j.trsl.2022.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/29/2022] [Accepted: 08/21/2022] [Indexed: 01/14/2023]
Abstract
Overactive inflammatory responses are central to the pathophysiology of many hemolytic conditions including sickle cell disease. Excessive hemolysis leads to elevated serum levels of heme due to saturation of heme scavenging mechanisms. Extracellular heme has been shown to activate the NLRP3 inflammasome, leading to activation of caspase-1 and release of pro-inflammatory cytokines IL-1β and IL-18. Heme also activates the non-canonical inflammasome pathway, which may contribute to NLRP3 inflammasome formation and leads to pyroptosis, a type of inflammatory cell death. Some clinical studies indicate there is a benefit to blocking the NLRP3 inflammasome pathway in patients with sickle cell disease and other hemolytic conditions. However, a thorough understanding of the mechanisms of heme-induced inflammasome activation is needed to fully leverage this pathway for clinical benefit. This review will explore the mechanisms of heme-induced NLRP3 inflammasome activation and the role of this pathway in hemolytic conditions including sickle cell disease.
Collapse
Affiliation(s)
- Suruchi Salgar
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas
| | - Beatriz E Bolívar
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jonathan M Flanagan
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas
| | - Shaniqua J Anum
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas
| | - Lisa Bouchier-Hayes
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, Texas; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
37
|
Chiang KC, Gupta A, Sundd P, Krishnamurti L. Thrombo-Inflammation in COVID-19 and Sickle Cell Disease: Two Faces of the Same Coin. Biomedicines 2023; 11:338. [PMID: 36830874 PMCID: PMC9953430 DOI: 10.3390/biomedicines11020338] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 01/26/2023] Open
Abstract
People with sickle cell disease (SCD) are at greater risk of severe illness and death from respiratory infections, including COVID-19, than people without SCD (Centers for Disease Control and Prevention, USA). Vaso-occlusive crises (VOC) in SCD and severe SARS-CoV-2 infection are both characterized by thrombo-inflammation mediated by endothelial injury, complement activation, inflammatory lipid storm, platelet activation, platelet-leukocyte adhesion, and activation of the coagulation cascade. Notably, lipid mediators, including thromboxane A2, significantly increase in severe COVID-19 and SCD. In addition, the release of thromboxane A2 from endothelial cells and macrophages stimulates platelets to release microvesicles, which are harbingers of multicellular adhesion and thrombo-inflammation. Currently, there are limited therapeutic strategies targeting platelet-neutrophil activation and thrombo-inflammation in either SCD or COVID-19 during acute crisis. However, due to many similarities between the pathobiology of thrombo-inflammation in SCD and COVID-19, therapies targeting one disease may likely be effective in the other. Therefore, the preclinical and clinical research spurred by the COVID-19 pandemic, including clinical trials of anti-thrombotic agents, are potentially applicable to VOC. Here, we first outline the parallels between SCD and COVID-19; second, review the role of lipid mediators in the pathogenesis of these diseases; and lastly, examine the therapeutic targets and potential treatments for the two diseases.
Collapse
Affiliation(s)
| | - Ajay Gupta
- KARE Biosciences, Orange, CA 89128, USA
- Division of Nephrology, Hypertension and Kidney Transplantation, Department of Medicine, University of California Irvine (UCI) School of Medicine, Irvine, CA 92868, USA
| | - Prithu Sundd
- Vascular Medicine Institute and Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Lakshmanan Krishnamurti
- Division of Pediatric Hematology-Oncology, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
38
|
Changes in Heme Levels During Acute Vaso-occlusive Crisis in Sickle Cell Anemia. Hematol Oncol Stem Cell Ther 2023; 16:124-132. [PMID: 34450106 DOI: 10.1016/j.hemonc.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/20/2021] [Accepted: 08/09/2021] [Indexed: 01/31/2023] Open
Abstract
OBJECTIVE/BACKGROUND Sickle cell anemia (SCA) is associated with increased levels of extracellular heme, which is a key mediator of inflammation in this condition. Despite abundant evidence supporting this concept in cell and animal models, few studies addressed the association between heme levels and the development and severity of acute vasoocclusive crises (VOC) in humans. METHODS A cross-sectional study was conducted in patients with acute VOC. Total extracellular heme levels were measured in both plasma and serum at admission and after convalescence, and correlated with other clinical and laboratory markers of SCA severity. RESULTS A total of 28 episodes of VOC in 25 patients were included. Heme levels were similar between admission and convalescence, and correlated with the difference between pre and post hemoglobin, and SCA severity estimated by a composite score of clinical and laboratory markers. Heme levels were neither associated with VOC severity nor with markers of hemostasis activation, and were similar to those reported in an independent population of SCA patients at steady state. DISCUSSION Acute VOC are not characterized by significant increases in total extracellular heme levels. Studies measuring the fraction of free extracellular heme unbound to proteins are warranted to further refine our understanding of the role of heme in acute VOC.
Collapse
|
39
|
Cherukury HM, Argueta DA, Garcia N, Fouda R, Kiven S, Lei J, Sagi V, Velasco GJ, Avalos B, DiPatrizio NV, Gupta K. Cannabidiol attenuates hyperalgesia in a mouse model of sickle cell disease. Blood 2023; 141:203-208. [PMID: 36122382 PMCID: PMC9936305 DOI: 10.1182/blood.2022016382] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 08/17/2022] [Accepted: 09/04/2022] [Indexed: 01/17/2023] Open
Affiliation(s)
- Hemanth M. Cherukury
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA
| | - Donovan A. Argueta
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA
| | - Natalie Garcia
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA
| | - Raghda Fouda
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA
| | - Stacy Kiven
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA
| | - Jianxun Lei
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Varun Sagi
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | | | - Bryant Avalos
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA
| | - Nicholas V. DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA
| | - Kalpna Gupta
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
- Southern California Institute for Research and Education, VA Medical Center, Long Beach, CA
| |
Collapse
|
40
|
Ivnitsky JJ, Schäfer TV, Rejniuk VL, Golovko AI. Endogenous humoral determinants of vascular endothelial dysfunction as triggers of acute poisoning complications. J Appl Toxicol 2023; 43:47-65. [PMID: 35258106 DOI: 10.1002/jat.4312] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/13/2022] [Accepted: 02/26/2022] [Indexed: 12/16/2022]
Abstract
The vascular endothelium is not only the semipermeable membrane that separates tissue from blood but also an organ that regulates inflammation, vascular tone, blood clotting, angiogenesis and synthesis of connective tissue proteins. It is susceptible to the direct cytotoxic action of numerous xenobiotics and to the acute hypoxia that accompanies acute poisoning. This damage is superimposed on the preformed state of the vascular endothelium, which, in turn, depends on many humoral factors. The probability that an exogenous toxicant will cause life-threatening dysfunction of the vascular endothelium, thereby complicating the course of acute poisoning, increases with an increase in the content of endogenous substances in the blood that disrupt endothelial function. These include ammonia, bacterial endotoxin, indoxyl sulfate, para-cresyl sulfate, trimethylamine N-oxide, asymmetric dimethylarginine, glucose, homocysteine, low-density and very-low-density lipoproteins, free fatty acids and products of intravascular haemolysis. Some other endogenous substances (albumin, haptoglobin, haemopexin, biliverdin, bilirubin, tetrahydrobiopterin) or food-derived compounds (ascorbic acid, rutin, omega-3 polyunsaturated fatty acids, etc.) reduce the risk of lethal vascular endothelial dysfunction. The individual variability of the content of these substances in the blood contributes to the stochasticity of the complications of acute poisoning and is a promising target for the risk reduction measures. Another feasible option may be the repositioning of drugs that affect the function of the vascular endothelium while being currently used for other indications.
Collapse
Affiliation(s)
- Jury Ju Ivnitsky
- Golikov Research Clinical Center of Toxicology under the Federal Medical Biological Agency, Saint Petersburg, Russia
| | - Timur V Schäfer
- State Scientific Research Test Institute of the Military Medicine of Defense Ministry of the Russian Federation, Saint Petersburg, Russia
| | - Vladimir L Rejniuk
- Golikov Research Clinical Center of Toxicology under the Federal Medical Biological Agency, Saint Petersburg, Russia
| | - Alexandr I Golovko
- Golikov Research Clinical Center of Toxicology under the Federal Medical Biological Agency, Saint Petersburg, Russia
| |
Collapse
|
41
|
Ansari J, Vital SA, Yadav S, Gavins FNE. Regulating Neutrophil PAD4/NOX-Dependent Cerebrovasular Thromboinflammation. Int J Biol Sci 2023; 19:852-864. [PMID: 36778112 PMCID: PMC9910005 DOI: 10.7150/ijbs.77434] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/21/2022] [Indexed: 01/11/2023] Open
Abstract
Background: Neutrophil extracellular trap (NET) production has been implicated in the pathogenesis of thromboinflammatory conditions such as Sickle Cell Disease (SCD), contributing to heightened risk for ischemic stroke. NETs are catalyzed by the enzyme Peptidyl Arginine Deiminase 4 (PAD4) and neutrophil derived reactive oxygen species (ROS), especially NADPH oxidase (NOX) which interacts with PAD4 and is therefore critical for neutrophil function. However, the role that NOX-dependent ROS and NETs play in the accelerated cerebral microvascular thrombosis associated with thromboinflammatory conditions, such as SCD, has not been fully elucidated and is the aim of this study. Methods: The in-vitro effects of targeting PAD4 and NOX were examined using physiologically relevant NET assays with neutrophils isolated from healthy volunteers (control) and SCD patients. In addition, in-vivo intravascular effects of targeting PAD4 and NOX in the cerebral microcirculation of C57BL/6 and sickle transgenic mice (STM) were assessed using a photoactivation thrombosis model (light/dye) coupled with real-time fluorescence intravital microscopy. Results: We found that targeting PAD4 and NOX in human neutrophils significantly inhibited ionomycin dependent H3cit+ neutrophils. Targeting PAD4 and NOX in-vivo resulted in prolonged blood flow cessation in cerebrovascular arterioles as well as venules. Moreover, we were able to replicate the effects of PAD4 and NOX targeting in a clinical model of accelerated thromboinflammation by increasing blood flow cessation times in cerebral microvessels in STM. These findings concurred with the clinical setting i.e. neutrophils isolated from SCD patients, which possessed an attenuation of H3cit+ neutrophil production on targeting PAD4 and NOX. Conclusions: Taken together, our compelling data suggests that PAD4 and NOX play a significant role in neutrophil driven thromboinflammation. Targeting PAD4 and NOX limits pathological H3cit+ neutrophils, which may further explain attenuation of cerebral thrombosis. Overall, this study presents a viable pre-clinical model of prevention and management of thromboinflammatory complications such as ischemic stroke.
Collapse
Affiliation(s)
- Junaid Ansari
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Shantel A Vital
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71130, USA
| | - Shreya Yadav
- Department of Life Sciences, Centre of Inflammation Research and Translational Medicine (CIRTM), Brunel University London, London. UB8 3PH, UK
| | - Felicity N E Gavins
- Department of Life Sciences, Centre of Inflammation Research and Translational Medicine (CIRTM), Brunel University London, London. UB8 3PH, UK
| |
Collapse
|
42
|
Ellsworth P, Sparkenbaugh EM. Targeting the von Willebrand Factor-ADAMTS-13 axis in sickle cell disease. J Thromb Haemost 2023; 21:2-6. [PMID: 36695390 PMCID: PMC10413208 DOI: 10.1016/j.jtha.2022.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 01/09/2023]
Affiliation(s)
- Patrick Ellsworth
- Department of Medicine, Division of Hematology and Blood Research Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Erica M Sparkenbaugh
- Department of Medicine, Division of Hematology and Blood Research Center, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA.
| |
Collapse
|
43
|
Gonzales J, Fraidenburg DR. Caught in a vicious cycle: Cell-free hemoglobin and the pathogenesis of pulmonary hypertension. Pulm Circ 2023; 13:e12194. [PMID: 36714058 PMCID: PMC9872046 DOI: 10.1002/pul2.12194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Affiliation(s)
- Janae Gonzales
- Department of MedicineUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | | |
Collapse
|
44
|
Kour D, Ali M, Khajuria P, Sharma K, Ghosh P, Kaur S, Mahajan S, Ramajayan P, Bharate SS, Bhardwaj S, Sawant SD, Reddy DS, Kumar A. Flurbiprofen inhibits heme induced NLRP3 inflammasome in Berkeley sickle cell disease mice. Front Pharmacol 2023; 14:1123734. [PMID: 37180702 PMCID: PMC10171431 DOI: 10.3389/fphar.2023.1123734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/03/2023] [Indexed: 05/16/2023] Open
Abstract
Sickle cell disease (SCD) is accompanied by several complications, which emanate from the sickling of erythrocytes due to a point mutation in the β-globin chain of hemoglobin. Sickled erythrocytes are unable to move smoothly through small blood capillaries and therefore, cause vaso occlusion and severe pain. Apart from pain, continuous lysis of fragile sickled erythrocytes leads to the release of heme, which is a strong activator of the NLRP3 inflammasome, thus producing chronic inflammation in sickle cell disease. In this study, we identified flurbiprofen among other COX-2 inhibitors to be a potent inhibitor of heme-induced NLRP3 inflammasome. We found that apart from being a nociceptive agent, flurbiprofen exerts a strong anti-inflammatory effect by suppressing NF-κB signaling, which was evidenced by reduced levels of TNF-α and IL-6 in wild-type and sickle cell disease Berkeley mice models. Our data further demonstrated the protective effect of flurbiprofen on liver, lungs, and spleen in Berkeley mice. The current sickle cell disease pain management regime relies mainly on opiate drugs, which is accompanied by several side effects without modifying the sickle cell disease-related pathology. Considering the potent role of flurbiprofen in inhibiting NLRP3 inflammasome and other inflammatory cytokines in sickle cell disease, our data suggests that it can be explored further for better sickle cell disease pain management along with the possibility of disease modification.
Collapse
Affiliation(s)
- Dilpreet Kour
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Mehboob Ali
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Parul Khajuria
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Kuhu Sharma
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Palash Ghosh
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Sukhleen Kaur
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Surbhi Mahajan
- Department of Pathology, Government Medical College, Jammu, India
| | - P. Ramajayan
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
| | - Sonali S. Bharate
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’s NMIMS, Mumbai, India
| | - Subhash Bhardwaj
- Department of Pathology, Government Medical College, Jammu, India
| | - Sanghapal D. Sawant
- Academy of Scientific and Innovative Research, Ghaziabaad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India
| | - D. Srinivasa Reddy
- Academy of Scientific and Innovative Research, Ghaziabaad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir, India
- *Correspondence: D. Srinivasa Reddy, ; Ajay Kumar,
| | - Ajay Kumar
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research, Ghaziabaad, India
- *Correspondence: D. Srinivasa Reddy, ; Ajay Kumar,
| |
Collapse
|
45
|
Investigation of Neutrophil Extracellular Traps as Potential Mediators in the Pathogenesis of Non-Acute Subdural Hematomas: A Pilot Study. Diagnostics (Basel) 2022; 12:diagnostics12122934. [PMID: 36552941 PMCID: PMC9776444 DOI: 10.3390/diagnostics12122934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Non-acute subdural hematomas (NASHs) are a cause of significant morbidity and mortality, particularly with recurrences. Although recurrence is believed to involve a disordered neuroinflammatory cascade involving vascular endothelial growth factor (VEGF), this pathway has yet to be completely elucidated. Neutrophil extracellular traps (NETs) are key factors that promote inflammation/apoptosis and can be induced by VEGF. We investigated whether NETs are present in NASH membranes, quantified NET concentrations, and examined whether NET and VEGF levels are correlated in NASHs. Samples from patients undergoing NASH evacuation were collected during surgery and postoperatively at 24 and 48 h. Fluid samples and NASH membranes were analyzed for levels of VEGF, NETs, and platelet activation. NASH samples contained numerous neutrophils positive for NET formation. Myeloperoxidase-DNA complexes (a marker of NETs) remained elevated 48 h postoperatively (1.06 ± 0.22 day 0, 0.72 ± 0.23 day 1, and 0.83 ± 0.33 day 2). VEGF was also elevated in NASHs (7.08 ± 0.98 ng/mL day 0, 3.40 ± 0.68 ng/mL day 1, and 6.05 ± 1.8 ng/mL day 2). VEGF levels were significantly correlated with myeloperoxidase-DNA levels. These results show that NETs are increasing in NASH, a finding that was previously unknown. The strong correlation between NET and VEGF levels indicates that VEGF may be an important mediator of NET-related inflammation in NASH.
Collapse
|
46
|
HbS promotes TLR4-mediated monocyte activation and proinflammatory cytokine production in sickle cell disease. Blood 2022; 140:1972-1982. [PMID: 35984906 DOI: 10.1182/blood.2021014894] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 08/04/2022] [Indexed: 11/20/2022] Open
Abstract
Monocytes are considered crucial actors of inflammation in sickle cell disease (SCD), being responsible for an increased production of proinflammatory cytokines such as tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), and IL-6. Although a role of free heme released by intravascular hemolysis has been suspected, the mechanisms underlying monocyte activation in patients with SCD remain unknown. Using purified human hemoglobin (Hb), we demonstrate herein, that cell-free HbS, unlike HbA or heme, is responsible for a major enhancement in the expression of proinflammatory cytokines by human monocytes. This effect was found mediated by direct interaction with the Toll-like receptor 4 (TLR4)/myeloid differentiation factor 2 (MD-2) complex, resulting in the activation of both the nuclear factor-κB (NF-κB) and type I interferon pathways. In Townes SCD mice, injection of HbS, unlike HbA, was responsible for an increased production of proinflammatory cytokines, which was prevented by the TLR4 inhibitor, TAK-242. Our results reveal a novel mechanism of monocyte activation and systemic inflammation in SCD, which opens new promising therapeutic perspectives targeting the HbS-TLR4 interaction.
Collapse
|
47
|
Protoporphyrin IX derived from dual-species anaerobic biofilms of Fusobacterium necrophorum and Porphyromonas levii attenuates bovine neutrophil function. Biofilm 2022; 4:100095. [DOI: 10.1016/j.bioflm.2022.100095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022] Open
|
48
|
Feugray G, Kasonga F, Grall M, Dumesnil C, Benhamou Y, Brunel V, Le Cam Duchez V, Lahary A, Billoir P. Investigation of thrombin generation assay to predict vaso-occlusive crisis in adulthood with sickle cell disease. Front Cardiovasc Med 2022; 9:883812. [PMID: 36277754 PMCID: PMC9579298 DOI: 10.3389/fcvm.2022.883812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Sickle cell disease (SCD) is an inherited hemoglobinopathy disorder. The main consequence is synthesis of hemoglobin S leading to chronic hemolysis associated with morbidity. The aim of this study was to investigate Thrombin Generation Assay (TGA) to assess hypercoagulability in SCD and TGA parameters as biomarkers of vaso-occlusive crisis (VOC) risk and hospitalization within 1 year. Materials and methods We performed TGA in platelet poor plasma (PPP) with 1 pM of tissue factor and 4 μM of phospholipid-standardized concentration, in duplicate for patients and controls. We measured thrombomodulin (TM), soluble endothelial Protein C Receptor and Tissue Factor Pathway Inhibitor (TFPI). Results A total of 113 adult patients with SCD, 83 at steady state and 30 during VOC, and 25 healthy controls matched on age and gender were included. Among the 83 patients at steady state, (36 S/S-1 S/β0, 20 S/Sα3.7, and 19 S/C-7 S/β+) 28 developed a VOC within 1 year (median: 4 months [2.25–6]). We observed an increase of peak and velocity associated with a shortening of lagtime and time to peak (TTP) and no difference of endogenous thrombin potential (ETP) in patients compared to controls. TFPI (p < 0.001) and TM (p = 0.006) were significantly decreased. TGA confirmed hypercoagulability in all SCD genotypes and clinical status. The association of ETP > 1,207 nM.min and peak >228.5 nM presented a sensitivity of 73.5% and a specificity of 93.9% to predict VOC development within 1 year. Conclusion We have demonstrated a hypercoagulable state in SCD associated with chronic hemolysis. These preliminary findings suggest that TGA parameters, as ETP and peak, could be used to predict VOC development within 1 year.
Collapse
Affiliation(s)
- Guillaume Feugray
- Vascular Hemostasis Unit, CHU Rouen, Normandie Université, UNIROUEN, INSERM U1096, Rouen, France
| | | | | | - Cécile Dumesnil
- Department of Pediatric Onco-Hematology, CHU Rouen, Rouen, France
| | - Ygal Benhamou
- Department of Internal Medicine, CHU Rouen, Normandie Université, UNIROUEN, INSERM U1096, Rouen, France
| | - Valery Brunel
- Department of General Biochemistry, CHU Rouen, Rouen, France
| | - Véronique Le Cam Duchez
- Vascular Hemostasis Unit, CHU Rouen, Normandie Université, UNIROUEN, INSERM U1096, Rouen, France
| | | | - Paul Billoir
- Vascular Hemostasis Unit, CHU Rouen, Normandie Université, UNIROUEN, INSERM U1096, Rouen, France,*Correspondence: Paul Billoir, ; orcid.org/0000-0001-5632-7713
| |
Collapse
|
49
|
Ngo ATP, Gollomp K. Building a better
NET
: Neutrophil extracellular trap targeted therapeutics in the treatment of infectious and inflammatory disorders. Res Pract Thromb Haemost 2022. [DOI: 10.1002/rth2.12808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Anh T. P. Ngo
- Division of Hematology Children's Hospital of Philadelphia Philadelphia Pennsylvania USA
| | - Kandace Gollomp
- Division of Hematology Children's Hospital of Philadelphia Philadelphia Pennsylvania USA
- Department of Pediatrics, Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| |
Collapse
|
50
|
Ni S, Yuan Y, Song S, Li X. A double-edged sword with a therapeutic target: iron and ferroptosis in immune regulation. Nutr Rev 2022; 81:587-596. [PMID: 36130411 DOI: 10.1093/nutrit/nuac071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cellular activities such as DNA synthesis, adenosine triphosphate production, and mitochondrial respiration are affected by iron metabolism. Disturbance of iron homeostasis usually leads to damage in cells and organs in the context of iron overload or deficiency. Thus, iron, a key regulator in nutritional immunity, was shown to be critical in innate and adaptive immunity. Unlike apoptosis, ferroptosis, a feature of iron-dependent lipid peroxidation, is thought to be associated with immune regulation because of its immunogenic nature. In this review, we summarize the role of iron and ferroptosis in immune regulation and discuss their therapeutic potential in the treatment of arthropathies like osteoarthritis and rheumatoid arthritis.
Collapse
Affiliation(s)
- Shuo Ni
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yin Yuan
- the State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shangdao Song
- the Department of Rehabilitation Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xiaolin Li
- the Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|