1
|
Wang WX, Spiegelman D, Rao PK, Ford AL, Apte RS. Crizanlizumab for retinal vasculopathy with cerebral leukoencephalopathy in a phase II clinical study. J Clin Invest 2024; 134:e180916. [PMID: 38950286 PMCID: PMC11178534 DOI: 10.1172/jci180916] [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: 03/06/2024] [Accepted: 05/01/2024] [Indexed: 07/03/2024] Open
Abstract
BackgroundRetinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations (RVCL-S) is a rare, autosomal dominant, universally fatal disease without effective treatment options. This study explores the safety and preliminary efficacy of crizanlizumab, a humanized monoclonal antibody against P-selectin approved for the prevention of sickle cell crises, in slowing retinal nonperfusion and preserving vision in patients with RVCL-S.METHODSEleven patients with RVCL-S with confirmed exonuclease 3 prime repair exonuclease 1 (TREX1) mutations received monthly crizanlizumab infusions over 2 years. The study measured the nonperfusion index within 3 retinal zones and the total retina with fluorescein angiography, visual acuity, intraocular pressure (IOP), and optical coherence tomography central subfield thickness (CST) at baseline, 1 year, and 2 years. A mixed repeated-measures analysis was performed to assess the progression rates and changes from baseline.RESULTSEleven participants received crizanlizumab infusions. All of the participants tolerated crizanlizumab well, with 8 of 11 (72.7%) reporting mild adverse effects such as nausea, fatigue, and gastrointestinal symptoms. The change in total retinal nonperfusion was 7.22% [4.47, 9.97] in year 1 and -0.69% [-4.06, 2.68] in year 2 (P < 0.001). In the mid periphery, the change in nonperfusion was 10.6% [5.1, 16.1] in year 1 and -0.68% [-3.98, 5.35] in year 2 (P < 0.01), demonstrating a reduction in progression of nonperfusion in the second year of treatment. Visual acuity, IOP, and CST remained stable.CONCLUSIONCrizanlizumab has an acceptable safety profile. These results show promising potential for examining crizanlizumab in larger studies of RVCL-S and similar small-vessel diseases and for using the retina as a biomarker for systemic disease.Trial registrationClinicalTrials.gov NCT04611880.FUNDINGThe Clayco Foundation; DeNardo Education and Research Foundation Grant; Jeffrey T. Fort Innovation Fund; Siteman Retina Research Fund; unrestricted grant from Research to Prevent Blindness Inc.; National Heart,Lung, and Blood Institute (NHLBI), NIH (R01HL129241); National Institute of Neurological Disorders and Stroke (NINDS), NIH (RF1NS116565).
Collapse
Affiliation(s)
- Wilson X. Wang
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, and
| | - Dan Spiegelman
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, and
| | - P. Kumar Rao
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, and
| | - Andria L. Ford
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rajendra S. Apte
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, and
- Department of Medicine, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
2
|
Oliveira JMF, Arcanjo GS, Domingos IF, Batista JVGF, Pereira-Martins DA, Batista THC, Hatzlhofer BLD, Falcão DA, Diniz MV, Silva AP, Pires BCV, Dos Anjos AC, Costa FF, Araujo AS, Lucena-Araujo AR, Bezerra MA. A-296G variant of THBS1 gene (rs1478605) is associated with a lower frequency of stroke in a Brazilian population with sickle cell anemia. J Stroke Cerebrovasc Dis 2024; 33:107474. [PMID: 38006767 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107474] [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: 06/25/2023] [Revised: 10/24/2023] [Accepted: 11/05/2023] [Indexed: 11/27/2023] Open
Abstract
OBJECTIVES Stroke is a devastating clinical outcome that significantly contributes to the morbidity and mortality of sickle cell anemia (SCA) patients. Despite its advantages in predicting stroke risk, transcranial Doppler screening has limitations that restrict its applicability, highlighting the need for emerging prognostic tools. Thrombospondin-1 plays a crucial role in endothelial injury, platelet adhesion, and nitric oxide metabolism and may be implicated in stroke pathophysiology. Here, we aimed to evaluate the association of THBS1 genetic variations with the occurrence of stroke in SCA patients MATERIALS AND METHODS: By real-time PCR, 512 SCA patients were fully genotyped for THBS1 A-296G (rs1478605) polymorphism RESULTS: THBS1 GG genotype was associated with a lower risk for stroke occurrence [odds ratio (OR): 0.30; 95% confidence interval (CI): 0.11-0.78; P = 0.011], although these findings were not consistent with multivariate logistic regression analysis (OR: 0.73, 95% CI: 0.12 - 4.37; P = 0.736). In agreement, the cumulative incidence of stroke for patients with AG/AA genotypes was higher when compared to the GG genotype (P = 0.018). However, the association was not maintained in the multivariate proportional hazards model (hazard ratio: 0.67, 95% CI: 0.12-3.61; P = 0.643) CONCLUSIONS: In summary, the present study shows that the THBS1 A-296G (rs1478605) polymorphism may be a potential modifier for stroke in SCA.
Collapse
Affiliation(s)
- Jessica M F Oliveira
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Gabriela S Arcanjo
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Igor F Domingos
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Pronto Socorro Cardiológico de Pernambuco, University of Pernambuco, Recife, Pernambuco, Brazil
| | - Jéssica V G F Batista
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Diego A Pereira-Martins
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Department of Internal Medicine, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Thais H C Batista
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Betânia L D Hatzlhofer
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Department of Pharmaceutical Sciences, Health Sciences Centre, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Diego A Falcão
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Madi V Diniz
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Alexsandro P Silva
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Bárbara C V Pires
- Department of Internal Medicine, Hematology and Hemotherapy Foundation of Pernambuco, Recife, Pernambuco, Brazil
| | - Ana C Dos Anjos
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Department of Internal Medicine, Hematology and Hemotherapy Foundation of Pernambuco, Recife, Pernambuco, Brazil
| | - Fernando F Costa
- Hematology and Hemotherapy Center, University of Campinas, Campinas, São Paulo, Brazil
| | - Aderson S Araujo
- Department of Internal Medicine, Hematology and Hemotherapy Foundation of Pernambuco, Recife, Pernambuco, Brazil
| | | | - Marcos A Bezerra
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Pernambuco, Brazil.
| |
Collapse
|
3
|
Karkoska KA, Gollamudi J, Hyacinth HI. Molecular and environmental contributors to neurological complications in sickle cell disease. Exp Biol Med (Maywood) 2023; 248:1319-1332. [PMID: 37688519 PMCID: PMC10625341 DOI: 10.1177/15353702231187646] [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] [Indexed: 09/11/2023] Open
Abstract
Sickle cell disease (SCD) is an inherited hemoglobinopathy in which affected hemoglobin polymerizes under hypoxic conditions resulting in red cell distortion and chronic hemolytic anemia. SCD affects millions of people worldwide, primarily in Sub-Saharan Africa and the Indian subcontinent. Due to vaso-occlusion of sickled red cells within the microvasculature, SCD affects virtually every organ system and causes significant morbidity and early mortality. The neurological complications of SCD are particularly devastating and diverse, ranging from overt stroke to covert cerebral injury, including silent cerebral infarctions and blood vessel tortuosity. However, even individuals without evidence of neuroanatomical changes in brain imaging have evidence of cognitive deficits compared to matched healthy controls likely due to chronic cerebral hypoxemia and neuroinflammation. In this review, we first examined the biological contributors to SCD-related neurological complications and then discussed the equally important socioenvironmental contributors. We then discuss the evidence for neuroprotection from the two primary disease-modifying therapies, chronic monthly blood transfusions and hydroxyurea, and end with several experimental therapies designed to specifically target these complications.
Collapse
Affiliation(s)
- Kristine A Karkoska
- Division of Hematology & Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45219-0525, USA
| | - Jahnavi Gollamudi
- Division of Hematology & Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45219-0525, USA
| | - Hyacinth I Hyacinth
- Department of Neurology & Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0525, USA
| |
Collapse
|
4
|
Abi Rached NM, Gbotosho OT, Archer DR, Jones JA, Sterling MS, Hyacinth HI. Adhesion molecules and cerebral microvascular hemodynamic abnormalities in sickle cell disease. Front Neurol 2022; 13:976063. [PMID: 36570439 PMCID: PMC9767957 DOI: 10.3389/fneur.2022.976063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Cerebrovascular abnormalities are a common feature of sickle cell disease that may be associated with risk of vaso-occlusive pain crises, microinfarcts, and cognitive impairment. An activated endothelium and adhesion factors, VCAM-1 and P-selectin, are implicated in sickle cell vasculopathy, including abnormal hemodynamics and leukocyte adherence. This study examined the association between cerebral expression of these adhesion factors and cortical microvascular blood flow dynamics by using in-vivo two-photon microscopy. We also examined the impact of blood transfusion treatment on these markers of vasculopathy. Results showed that sickle cell mice had significantly higher maximum red blood cell (RBC) velocity (6.80 ± 0.25 mm/sec, p ≤ 0.01 vs. 5.35 ± 0.35 mm/sec) and more frequent blood flow reversals (18.04 ± 0.95 /min, p ≤ 0.01 vs. 13.59 ± 1.40 /min) in the cortical microvasculature compared to controls. In addition, sickle cell mice had a 2.6-fold (RFU/mm2) increase in expression of VCAM-1 and 17-fold (RFU/mm2) increase in expression of P-selectin compared to controls. This was accompanied by an increased frequency in leukocyte adherence (4.83 ± 0.57 /100 μm/min vs. 2.26 ± 0.37 /100 μm/min, p ≤ 0.001). We also found that microinfarcts identified in sickle cell mice were 50% larger than in controls. After blood transfusion, many of these parameters improved, as results demonstrated that sickle cell mice had a lower post-transfusion maximum RBC velocity (8.30 ± 0.98 mm/sec vs. 11.29 ± 0.95 mm/sec), lower frequency of blood flow reversals (12.80 ± 2.76 /min vs. 27.75 ± 2.09 /min), and fewer instances of leukocyte adherence compared to their pre-transfusion imaging time point (1.35 ± 0.32 /100 μm/min vs. 3.46 ± 0.58 /100 μm/min). Additionally, we found that blood transfusion was associated with lower expression of adhesion factors. Our results suggest that blood transfusion and adhesion factors, VCAM-1 and P-selectin, are potential therapeutic targets for addressing cerebrovascular pathology, such as vaso-occlusion, in sickle cell disease.
Collapse
Affiliation(s)
- Noor Mary Abi Rached
- Neuroscience and Behavioral Biology Undergraduate Program, Emory University, Atlanta, GA, United States
| | - Oluwabukola T. Gbotosho
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - David R. Archer
- Aflac Cancer and Blood Disorders Center, Emory University Department of Pediatrics and Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Jayre A. Jones
- Aflac Cancer and Blood Disorders Center, Emory University Department of Pediatrics and Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Morgan S. Sterling
- Aflac Cancer and Blood Disorders Center, Emory University Department of Pediatrics and Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Hyacinth I. Hyacinth
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
5
|
Karki NR, Saunders K, Kutlar A. A critical evaluation of crizanlizumab for the treatment of sickle cell disease. Expert Rev Hematol 2021; 15:5-13. [PMID: 34942078 DOI: 10.1080/17474086.2022.2023007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION P-selectin is a key adhesion molecule in the pathogenesis of sickle cell disease, including acute painful event(s). Many of the mediators activated in prototypical pain crisis are also involved in other complications seen in this population. Crizanlizumab is a monoclonal antibody approved in the US in 2019 for patients of all genotypes of sickle cell disease. By blocking P-selectin, it effectively prevents acute painful event(s) and has a manageable safety profile. AREAS COVERED In this review, we provide an overview of the (i) biology of P-selectin in sickle cell disease, (ii) various agents inhibiting P-selectin, (iii) pharmacology of crizanlizumab, (iv) preclinical and clinical data on crizanlizumab, and (v) its potential for other indications, ongoing studies, regulatory status, and cost issues. Further, we describe its position among other approved agents in sickle cell disease and project future directions as well. EXPERT OPINION Crizanlizumab holds great promise in modulating the natural history of sickle cell disease and may have pleotropic effects. Studies are ongoing to define its role in preventing other sickle cell-related complications, non-sickle cell inflammatory states, and thrombotic disorders.
Collapse
Affiliation(s)
- Nabin Raj Karki
- Division of Hematology/Oncology, Augusta University, Augusta, GA, USA
| | | | - Abdullah Kutlar
- Division of Hematology/Oncology, Augusta University, Augusta, GA, USA
| |
Collapse
|
6
|
Pitanga TN, Santana SS, Zanette DL, Guarda CC, Santiago RP, Maffili VV, Lima JB, Carvalho GQ, Filho JR, Ferreira JRD, Aleluia MM, Nascimento VML, Carvalho MOS, Lyra IM, Borges VM, Oliveira RR, Goncalves MS. Effect of lysed and non-lysed sickle red cells on the activation of NLRP3 inflammasome and LTB4 production by mononuclear cells. Inflamm Res 2021; 70:823-834. [PMID: 34196737 DOI: 10.1007/s00011-021-01461-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 03/24/2021] [Accepted: 03/30/2021] [Indexed: 10/21/2022] Open
Abstract
OBJECTIVE AND DESIGN This study tested the hypothesis that sickle red blood cell (SS-RBC) can induce inflammasome NLRP3 components gene expression in peripheral blood mononuclear cells (PBMCs) as well as interleukin-1β (IL-1β) and leukotriene B4 (LTB4) production. Additionally, we investigated the effect of hydroxyurea (HU) treatment in these inflammatory markers. METHODS PBMCs from healthy donors (AA-PBMC) were challenged with intact and lysed RBCs from SCA patients (SS-RBC) and from healthy volunteers (AA-RBC). NLRP3, IL-1β, IL-18 and Caspase-1 gene expression levels were assessed by quantitative PCR (qPCR). IL-1β protein levels and LTB4 were measured by ELISA. RESULTS We observed that lysed SS-RBC induced the expression of inflammasome NLRP3 components, but this increase was more prominent for CASP1 and IL18 expression levels. Moreover, we observed that intact SS-RBC induced higher production of IL-1β and LTB4 than lysed SS-RBC. Although SCA patients treated with HU have a reduction in NLRP3 gene expression and LTB4 production, this treatment did not modulate the expression of other inflammasome components or IL-1β production. CONCLUSIONS Thus, our data suggest that caspase-1, IL-1β and IL-18 may contribute to the inflammatory status observed in SCA and that HU treatment may not interfere in this inflammatory pathway.
Collapse
Affiliation(s)
- Thassila N Pitanga
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil.,Universidade Católica do Salvador (UCSAL), Salvador, Bahia, Brazil
| | - Sânzio S Santana
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil.,Universidade Católica do Salvador (UCSAL), Salvador, Bahia, Brazil
| | - Dalila L Zanette
- Fundação Oswaldo Cruz, Instituto Carlos Chagas (ICC-FIOCRUZ/PR), Curitiba, Paraná, Brazil
| | - Caroline C Guarda
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | - Rayra P Santiago
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | - Vitor V Maffili
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | - Jonilson B Lima
- Universidade Federal do Oeste da Bahia (UFOB), Barreiras, Bahia, Brazil
| | - Graziele Q Carvalho
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | - Jaime R Filho
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | | | - Milena M Aleluia
- Universidade Estadual de Santa Cruz (UESC), Ilhéus, Bahia, Brazil
| | - Valma M L Nascimento
- Fundação de Hematologia e Hemoterapia da Bahia (HEMOBA), Salvador, Bahia, Brazil
| | - Magda O S Carvalho
- Hospital Universitário Professor Edgard Santos (HUPES), UFBA, Salvador, Bahia, Brazil
| | - Isa M Lyra
- Hospital Universitário Professor Edgard Santos (HUPES), UFBA, Salvador, Bahia, Brazil
| | - Valéria M Borges
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | - Ricardo R Oliveira
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil
| | - Marilda S Goncalves
- Instituto Gonçalo Moniz, FIOCRUZ Bahia, Fundação Oswaldo Cruz / FIOCRUZ, Rua Waldemar Falcão, n. 121, Candeal, Salvador, Bahia, 40296710, Brazil. .,Universidade Federal da Bahia (UFBA), Salvador, Bahia, Brazil.
| |
Collapse
|
7
|
Complement in sickle cell disease and targeted therapy: I know one thing, that I know nothing. Blood Rev 2021; 48:100805. [PMID: 33504459 DOI: 10.1016/j.blre.2021.100805] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 11/10/2020] [Accepted: 01/13/2021] [Indexed: 12/19/2022]
Abstract
Sickle cell disease (SCD) is a common inherited clinical syndrome, characterized by the presence of hemoglobin S. Anemia, susceptibility to infections and episodes of vaso-occlusive crisis (VOC) are among its features. Since SCD complications (VOC or delayed hemolytic transfusion reaction/DHTR) lead to significant morbidity and mortality, a number of studies have addressed their pathophysiology Although SCD pathophysiology has been mainly attributed to the interaction between sickle cells and neutrophils, platelets or endothelial cells in small vessels leading to hemolysis, the role of complement activation has been increasingly investigated. Importantly, complement inhibition with eculizumab has shown beneficial effects in DHTR. Given the unmet clinical need of novel therapeutics in SCD, our review summarizes current understanding of (a) complement system for the clinician, (b) complement activation in SCD both in asymptomatic state and severe clinical manifestations, (c) probable underlying mechanisms of complement activation in SCD, and (d) new therapeutic perspective of complement inhibition.
Collapse
|
8
|
Hebbel RP, Belcher JD, Vercellotti GM. The multifaceted role of ischemia/reperfusion in sickle cell anemia. J Clin Invest 2020; 130:1062-1072. [PMID: 32118586 DOI: 10.1172/jci133639] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sickle cell anemia is a unique disease dominated by hemolytic anemia and vaso-occlusive events. The latter trigger a version of ischemia/reperfusion (I/R) pathobiology that is singular in its origin, cyclicity, complexity, instability, perpetuity, and breadth of clinical consequences. Specific clinical features are probably attributable to local I/R injury (e.g., stroke syndromes) or remote organ injury (e.g., acute chest syndrome) or the systematization of inflammation (e.g., multifocal arteriopathy). Indeed, by fashioning an underlying template of endothelial dysfunction and vulnerability, the robust inflammatory systematization no doubt contributes to all sickle pathology. In this Review, we highlight I/R-targeting therapeutics shown to improve microvascular blood flow in sickle transgenic mice undergoing I/R, and we suggest how such insights might be translated into human therapeutic strategies.
Collapse
|
9
|
Lizarralde-Iragorri MA, Shet AS. Sickle Cell Disease: A Paradigm for Venous Thrombosis Pathophysiology. Int J Mol Sci 2020; 21:ijms21155279. [PMID: 32722421 PMCID: PMC7432404 DOI: 10.3390/ijms21155279] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023] Open
Abstract
Venous thromboembolism (VTE) is an important cause of vascular morbidity and mortality. Many risk factors have been identified for venous thrombosis that lead to alterations in blood flow, activate the vascular endothelium, and increase the propensity for blood coagulation. However, the precise molecular and cellular mechanisms that cause blood clots in the venous vasculature have not been fully elucidated. Patients with sickle cell disease (SCD) demonstrate all the risk factors for venous stasis, activated endothelium, and blood hypercoagulability, making them particularly vulnerable to VTE. In this review, we will discuss how mouse models have elucidated the complex vascular pathobiology of SCD. We review the dysregulated pathways of inflammation and coagulation in SCD and how the resultant hypercoagulable state can potentiate thrombosis through down-regulation of vascular anticoagulants. Studies of VTE pathogenesis using SCD mouse models may provide insight into the intersection between the cellular and molecular processes involving inflammation and coagulation and help to identify novel mechanistic pathways.
Collapse
|
10
|
Vercellotti GM, Dalmasso AP, Schaid TR, Nguyen J, Chen C, Ericson ME, Abdulla F, Killeen T, Lindorfer MA, Taylor RP, Belcher JD. Critical role of C5a in sickle cell disease. Am J Hematol 2019; 94:327-337. [PMID: 30569594 DOI: 10.1002/ajh.25384] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/16/2018] [Accepted: 12/10/2018] [Indexed: 12/31/2022]
Abstract
Innate immune complement activation may contribute to sickle cell disease (SCD) pathogenesis. Ischemia-reperfusion physiology is a key component of the inflammatory and vaso-occlusive milieu in SCD and is associated with complement activation. C5a is an anaphylatoxin, a potent pro-inflammatory mediator that can activate leukocytes, platelets, and endothelial cells, all of which play a role in vaso-occlusion. We hypothesize that hypoxia-reoxygenation (H/R) in SCD mice activates complement, promoting inflammation and vaso-occlusion. At baseline and after H/R, sickle Townes-SS mice had increased C3 activation fragments and C5b-9 deposition in kidneys, livers and lungs and alternative pathway Bb fragments in plasma compared to control AA-mice. Activated complement promoted vaso-occlusion (microvascular stasis) in SS-mice; infusion of zymosan-activated, but not heat-inactivated serum, induced substantial vaso-occlusion in the skin venules of SS-mice. Infusion of recombinant C5a induced stasis in SS, but not AA-mice that was blocked by anti-C5a receptor (C5aR) IgG. C5a-mediated stasis was accompanied by inflammatory responses in SS-mice including NF-κB activation and increased expression of TLR4 and adhesion molecules VCAM-1, ICAM-1, and E-selectin in the liver. Anti-C5aR IgG blocked these inflammatory responses. Also, C5a rapidly up-regulated Weibel-Palade body P-selectin and von Willebrand factor on the surface of human umbilical vein endothelial cells in vitro and on vascular endothelium in vivo. In SS-mice, a blocking antibody to P-selectin inhibited C5a-induced stasis. Similarly, an antibody to C5 that blocks murine C5 cleavage or an antibody that blocks C5aR inhibited H/R-induced stasis in SS-mice. These results suggest that inhibition of C5a may be beneficial in SCD.
Collapse
Affiliation(s)
- Gregory M. Vercellotti
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | | | - Terry R. Schaid
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Julia Nguyen
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Chunsheng Chen
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Marna E. Ericson
- Department of Dermatology; University of Minnesota; Minneapolis Minnesota
| | - Fuad Abdulla
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Trevor Killeen
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| | - Margaret A. Lindorfer
- Department of Biochemistry and Molecular Genetics; University of Virginia School of Medicine; Charlottesville Virginia
| | - Ronald P. Taylor
- Department of Biochemistry and Molecular Genetics; University of Virginia School of Medicine; Charlottesville Virginia
| | - John D. Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation; University of Minnesota; Minneapolis Minnesota
| |
Collapse
|
11
|
Abstract
The primary β-globin gene mutation that causes sickle cell disease (SCD) has significant pathophysiological consequences that result in hemolytic events and the induction of the inflammatory processes that ultimately lead to vaso-occlusion. In addition to their role in the initiation of the acute painful vaso-occlusive episodes that are characteristic of SCD, inflammatory processes are also key components of many of the complications of the disease including autosplenectomy, acute chest syndrome, pulmonary hypertension, leg ulcers, nephropathy and stroke. We, herein, discuss the events that trigger inflammation in the disease, as well as the mechanisms, inflammatory molecules and cells that propagate these inflammatory processes. Given the central role that inflammation plays in SCD pathophysiology, many of the therapeutic approaches currently under pre-clinical and clinical development for the treatment of SCD endeavor to counter aspects or specific molecules of these inflammatory processes and it is possible that, in the future, we will see anti-inflammatory drugs being used either together with, or in place of, hydroxyurea in those SCD patients for whom hematopoietic stem cell transplants and evolving gene therapies are not a viable option.
Collapse
Affiliation(s)
- Nicola Conran
- Hematology Center, University of Campinas - UNICAMP, Cidade Universitária, Campinas-SP, Brazil
| | - John D Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Vascular Biology Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
12
|
Horton RE. Microfluidics for investigating vaso-occlusions in sickle cell disease. Microcirculation 2018; 24. [PMID: 28376286 DOI: 10.1111/micc.12373] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/30/2017] [Indexed: 12/17/2022]
Abstract
SCD stems from amutation in the beta globin gene. Upon deoxygenation, hemoglobin polymerizes and triggers RBC remodeling. This phenomenon is central to SCD pathogenesis as individuals suffering from the disease are plagued by painful vaso-occlusive crises episodes. These episodes are the result of a combination of processes including inflammation, thrombosis, and blood cell adhesion to the vascular wall which leads to blockages within the vasculature termed vaso-occlusions. Vaso-occlusive episodes deprive tissues of oxygen and are a major contributor to SCD-related complications; unfortunately, the complex mechanisms that contribute to vaso-occlusions are not well understood. Vaso-occlusions can occur in post-capillary venules; hence, the microvasculature is a prime target for SCD therapies. Traditional in vitro systems poorly recapitulate architectural and dynamic flow properties of in vivo systems. However, microfluidic devices can capture features of the native vasculature such as cellular composition, flow, geometry, and ECM presentation. This review, although not comprehensive, highlights microfluidic approaches that aim to improve our current understanding of the pathophysiological mechanisms surrounding SCD. Microfluidic platforms can aid in identifying factors that may contribute to disease severity and can serve as suitable test beds for novel treatment strategies which may improve patient outcomes.
Collapse
Affiliation(s)
- Renita E Horton
- Agricultural and Biological Engineering Department, College of Agriculture and Life Sciences, James Worth Bagley College of Engineering, Mississippi State University, Starkville, MS, USA
| |
Collapse
|
13
|
Pitanga TN, Oliveira RR, Zanette DL, Guarda CC, Santiago RP, Santana SS, Nascimento VML, Lima JB, Carvalho GQ, Maffili VV, Carvalho MOS, Alcântara LCJ, Borges VM, Goncalves MS. Sickle red cells as danger signals on proinflammatory gene expression, leukotriene B4 and interleukin-1 beta production in peripheral blood mononuclear cell. Cytokine 2016; 83:75-84. [PMID: 27045344 DOI: 10.1016/j.cyto.2016.03.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/08/2016] [Accepted: 03/21/2016] [Indexed: 02/06/2023]
Abstract
This study tested the hypothesis that sickle red blood cell (SS-RBC) induce Toll-like receptors (TLR) and Nod-like receptor family, pyrin domain containing 3 (NLRP3)- inflammasome expression in peripheral blood mononuclear cells (PBMC). TLR and NLRP3 inflammasome could contribute to the maintenance of the inflammatory status in sickle cell anemia (SCA) patients, since SS-RBC act as danger signals activating these pathways. In this study, first, we evaluated TLR (2, 4, 5 and 9), NLRP3, Caspase-1, interleukin (IL)-1β and IL-18 expression in PBMC freshly isolated from SCA patients (SS-PBMC) in comparison with PBMC from healthy individuals (AA-PBMC). In the second moment, we investigated whether SS-RBC could interfere with the expression of these molecules in PBMC from healthy donor, in the absence or presence of hydroxyurea (HU) in vitro. TLRs and NLRP3 inflammasome expression were investigated by qPCR. IL-1β, Leukotriene-B4 (LTB4) and nitrite production were measured in PBMC (from healthy donor) culture supernatants. TLR2, TLR4, TLR5, NLRP3 and IL-1β were highly expressed in SS-PBMC when compared to AA-PBMC. Additionally, SS-RBC induced TLR9, NLRP3, Caspase-1, IL-1β and IL-18 expression and induced IL-1β, LTB4 and nitrite production in PBMC cultures. HU did not prevent TLR and NLRP3 inflammasome expression, but increased TLR2 and IL-18 expression and reduced nitrite production. In conclusion, our data suggest that TLR and inflammasome complexes may be key inducers of inflammation in SCA patients, probably through SS-RBC; also, HU does not prevent NLRP3 inflammasome- and TLR-dependent inflammation, indicating the need to develop new therapeutic strategies to SCA patients that act with different mechanisms of those observed for HU.
Collapse
Affiliation(s)
- Thassila N Pitanga
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil; Instituto de Ciências da Saúde (ICS), Universidade Federal da Bahia (UFBA), 40110902 Salvador, Bahia, Brazil
| | - Ricardo R Oliveira
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Dalila L Zanette
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Caroline C Guarda
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Rayra P Santiago
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Sanzio S Santana
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Valma M L Nascimento
- Fundação de Hematologia e Hemoterapia da Bahia (HEMOBA), 40286240 Salvador, Bahia, Brazil
| | - Jonilson B Lima
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Graziele Q Carvalho
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Vitor V Maffili
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Magda O S Carvalho
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil; Hospital Universitário Professor Edgard Santos (HUPES), UFBA, 40110060 Salvador, Bahia, Brazil
| | - Luiz C J Alcântara
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Valéria M Borges
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil
| | - Marilda S Goncalves
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz - FIOCRUZ/BA, 40296710 Salvador, Bahia, Brazil; Faculdade de Farmácia, UFBA, 40170115 Salvador, Bahia, Brazil.
| |
Collapse
|
14
|
Martin C, Pialoux V, Faes C, Charrin E, Skinner S, Connes P. Does physical activity increase or decrease the risk of sickle cell disease complications? Br J Sports Med 2015; 52:214-218. [PMID: 26701924 DOI: 10.1136/bjsports-2015-095317] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2015] [Indexed: 12/11/2022]
Abstract
Sickle cell disease (SCD) is the most common inherited disease in the world. Red blood cell sickling, blood cell-endothelium adhesion, blood rheology abnormalities, intravascular haemolysis, and increased oxidative stress and inflammation contribute to the pathophysiology of SCD. Because acute intense exercise may alter these pathophysiological mechanisms, physical activity is usually contra-indicated in patients with SCD. However, recent studies in sickle-cell trait carriers and in a SCD mice model show that regular physical activity could decrease oxidative stress and inflammation, limit blood rheology alterations and increase nitric oxide metabolism. Therefore, supervised habitual physical activity may benefit patients with SCD. This article reviews the literature on the effects of acute and chronic exercise on the biological responses and clinical outcomes of patients with SCD.
Collapse
Affiliation(s)
- Cyril Martin
- Center of Research and Innovation on Sports (CRIS EA647), Team 'Vascular Biology and Red Blood Cell', University of Lyon 1, University of Lyon, Lyon, France.,Laboratory of Excellence in Red Blood Cell (LABEX GR-Ex), PRES Sorbonne, Paris, France
| | - Vincent Pialoux
- Center of Research and Innovation on Sports (CRIS EA647), Team 'Vascular Biology and Red Blood Cell', University of Lyon 1, University of Lyon, Lyon, France.,Laboratory of Excellence in Red Blood Cell (LABEX GR-Ex), PRES Sorbonne, Paris, France.,Institut Universitaire de France, Paris, France
| | - Camille Faes
- Center of Research and Innovation on Sports (CRIS EA647), Team 'Vascular Biology and Red Blood Cell', University of Lyon 1, University of Lyon, Lyon, France.,Laboratory of Excellence in Red Blood Cell (LABEX GR-Ex), PRES Sorbonne, Paris, France
| | - Emmanuelle Charrin
- Center of Research and Innovation on Sports (CRIS EA647), Team 'Vascular Biology and Red Blood Cell', University of Lyon 1, University of Lyon, Lyon, France.,Laboratory of Excellence in Red Blood Cell (LABEX GR-Ex), PRES Sorbonne, Paris, France
| | - Sarah Skinner
- Center of Research and Innovation on Sports (CRIS EA647), Team 'Vascular Biology and Red Blood Cell', University of Lyon 1, University of Lyon, Lyon, France.,Laboratory of Excellence in Red Blood Cell (LABEX GR-Ex), PRES Sorbonne, Paris, France
| | - Philippe Connes
- Center of Research and Innovation on Sports (CRIS EA647), Team 'Vascular Biology and Red Blood Cell', University of Lyon 1, University of Lyon, Lyon, France.,Laboratory of Excellence in Red Blood Cell (LABEX GR-Ex), PRES Sorbonne, Paris, France.,Institut Universitaire de France, Paris, France
| |
Collapse
|
15
|
Andemariam B, Adami AJ, Singh A, McNamara JT, Secor ER, Guernsey LA, Thrall RS. The sickle cell mouse lung: proinflammatory and primed for allergic inflammation. Transl Res 2015; 166:254-68. [PMID: 25843670 PMCID: PMC4537824 DOI: 10.1016/j.trsl.2015.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/09/2015] [Accepted: 03/10/2015] [Indexed: 01/22/2023]
Abstract
Comorbid asthma in sickle cell disease (SCD) confers higher rates of vaso-occlusive pain and mortality, yet the physiological link between these two distinct diseases remains puzzling. We used a mouse model of SCD to study pulmonary immunology and physiology before and after the induction of allergic airway disease (AAD). SCD mice were sensitized with ovalbumin (OVA) and aluminum hydroxide by the intraperitoneal route followed by daily, nose-only OVA-aerosol challenge to induce AAD. The lungs of naive SCD mice showed signs of inflammatory and immune processes: (1) histologic and cytochemical evidence of airway inflammation compared with naive wild-type mice; (2) bronchoalveolar lavage (BAL) fluid contained increased total lymphocytes, %CD8+ T cells, granulocyte-colony stimulating factor, interleukin 5 (IL-5), IL-7, and chemokine (C-X-C motif) ligand (CXCL)1; and (3) lung tissue and hilar lymph node (HLN) had increased CD4+, CD8+, and regulatory T (Treg) cells. Furthermore, SCD mice at AAD demonstrated significant changes compared with the naive state: (1) BAL fluid with increased %CD4+ T cells and Treg cells, lower %CD8+ T cells, and decreased interferon gamma, CXCL10, chemokine (C-C motif) ligand 2, and IL-17; (2) serum with increased OVA-specific immunoglobulin E, IL-6, and IL-13, and decreased IL-1α and CXCL10; (3) no increase in Treg cells in the lung tissue or HLN; and (4) hyporesponsiveness to methacholine challenge. In conclusion, SCD mice have an altered immunologic pulmonary milieu and physiological responsiveness. These findings suggest that the clinical phenotype of AAD in SCD mice differs from that of wild-type mice and that individuals with SCD may also have a unique, divergent phenotype perhaps amenable to a different therapeutic approach.
Collapse
Affiliation(s)
- Biree Andemariam
- Division of Hematology-Oncology, Lea Center for Hematologic Disorders, Adult Sickle Cell Center, University of Connecticut Health Center, Farmington, Conn.
| | - Alexander J Adami
- Department of Immunology, University of Connecticut Health Center, Farmington, Conn
| | - Anurag Singh
- Department of Immunology, University of Connecticut Health Center, Farmington, Conn
| | - Jeffrey T McNamara
- Department of Immunology, University of Connecticut Health Center, Farmington, Conn
| | - Eric R Secor
- Department of Immunology, University of Connecticut Health Center, Farmington, Conn
| | - Linda A Guernsey
- Department of Immunology, University of Connecticut Health Center, Farmington, Conn
| | - Roger S Thrall
- Department of Immunology, University of Connecticut Health Center, Farmington, Conn
| |
Collapse
|
16
|
McEver RP. Selectins: initiators of leucocyte adhesion and signalling at the vascular wall. Cardiovasc Res 2015; 107:331-9. [PMID: 25994174 PMCID: PMC4592324 DOI: 10.1093/cvr/cvv154] [Citation(s) in RCA: 374] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/06/2015] [Accepted: 04/29/2015] [Indexed: 12/28/2022] Open
Abstract
The selectins are transmembrane, Ca(2+)-dependent lectins that mediate leucocyte rolling on vascular surfaces, the first adhesive step during inflammation and immune surveillance. Leucocytes express L-selectin, activated platelets express P-selectin, and activated endothelial cells express E- and P-selectin. Rolling involves force-regulated, rapidly reversible interactions of selectins with a limited number of glycosylated cell surface ligands. Rolling permits leucocytes to interact with immobilized chemokines that convert β2 integrins to high-affinity conformations, which mediate arrest, post-arrest adhesion strengthening, and transendothelial migration. However, rolling leucocytes also transduce signals through selectin ligands, the focus of this review. These signals include serial activation of kinases and recruitment of adaptors that convert integrins to intermediate-affinity conformations, which decrease rolling velocities. In vitro, selectin signalling enables myeloid cells to respond to suboptimal levels of chemokines and other agonists. This cooperative signalling triggers effector responses such as degranulation, superoxide production, chemokine synthesis, and release of procoagulant/proinflammatory microparticles. In vivo, selectin-mediated adhesion and signalling likely contributes to atherosclerosis, arterial and deep vein thrombosis, ischaemia-reperfusion injury, and other cardiovascular diseases.
Collapse
Affiliation(s)
- Rodger P McEver
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, and Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 825 N.E. 13th Street, Oklahoma City, OK 73104, USA
| |
Collapse
|
17
|
Novelli EM, Elizabeth Sarles C, Jay Aizenstein H, Ibrahim TS, Butters MA, Connelly Ritter A, Erickson KI, Rosano C. Brain venular pattern by 7T MRI correlates with memory and haemoglobin in sickle cell anaemia. Psychiatry Res 2015; 233:18-22. [PMID: 26002434 PMCID: PMC7147953 DOI: 10.1016/j.pscychresns.2015.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 01/22/2015] [Accepted: 04/20/2015] [Indexed: 11/23/2022]
Abstract
Sickle cell anaemia (SCA) is a hereditary hemoglobinopathy characterised by extensive vascular dysfunction that stems from inflammation, thrombosis and occlusion of post-capillary venules. Cognitive impairment is a neurological complication of SCA whose pathogenesis is unknown. We hypothesised that cerebral venular abnormalities are linked to cognitive impairment in SCA. Thus, we employed 7T magnetic resonance imaging (MRI) to examine the association between venular density and cognitive function in homozygous SCA. We quantified the density of total, long, and short venules in pre-defined regions of interest between the frontal and occipital cornu on each hemisphere. Cognitive function was assessed using the Hopkins Verbal Learning Test - Revised (HVLT-R) test of learning and memory. Patients (n=11) were compared with race, age and gender-equated controls (n=7). Compared to controls, patients had an overall venular rarefaction, with significantly lower density of long venules and greater density of short venules which was inversely related to HVLT-R performance and haemoglobin. To our knowledge, this is the first 7T MRI study in SCA and first report of associations between cerebral venular patterns and cognitive performance and haemoglobin. Future studies should examine whether these novel neuroimaging markers predict cognitive impairment longitudinally and are mechanistically linked to severity of anaemia.
Collapse
Affiliation(s)
- Enrico M Novelli
- University of Pittsburgh Medical Center (UPMC) and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - C Elizabeth Sarles
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Tamer S Ibrahim
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Meryl A Butters
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Kirk I Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Caterina Rosano
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
18
|
Belcher JD, Chen C, Nguyen J, Abdulla F, Nguyen P, Nguyen M, Okeley NM, Benjamin DR, Senter PD, Vercellotti GM. The fucosylation inhibitor, 2-fluorofucose, inhibits vaso-occlusion, leukocyte-endothelium interactions and NF-ĸB activation in transgenic sickle mice. PLoS One 2015; 10:e0117772. [PMID: 25706118 PMCID: PMC4338063 DOI: 10.1371/journal.pone.0117772] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 12/30/2014] [Indexed: 02/02/2023] Open
Abstract
2-Fluorofucose (2FF) blocks the fucosylation and the tethering of sialyl-Lewisx tetrasaccharide and structural variants on leukocytes and red blood cells to P- and E-selectins on activated endothelial cell surfaces. Because P- and E-selectin are required for vaso-occlusion in murine sickle cell disease (SCD), we investigated whether 2FF would inhibit vaso-occlusion in SCD mice. Microvascular stasis was measured in subcutaneous venules in NY1DD and HbSS-Townes SCD mice with dorsal skin-fold chambers after infusion of hemoglobin or exposure to hypoxia/reoxygenation. 2FF in drinking water or administered by gavage inhibited stasis in sickle mice in a dose-responsive manner. Significant inhibitory effects on stasis were seen 1 day post-treatment. 2FF treatment of SCD mice also significantly reduced leukocyte rolling and adhesion along the vessel walls of SCD mice and the static adhesion of neutrophils and sickle red blood cells isolated from 2FF-treated SCD mice to resting and activated endothelial cells. Total white blood cell counts increased in response to 2FF. NF-ĸB activation and VCAM-1 and E-selectin expression were inhibited in the livers of SCD mice consistent with an overall decrease in vascular inflammation and ischemia-reperfusion physiology. Pretreatment with 2FF completely eliminated heme-induced lethality in HbSS-Townes mice, consistent with the observed anti-inflammatory and anti-adhesive properties of 2FF in SCD mice. These data suggest that 2FF may be beneficial for preventing or treating vaso-occlusive crises in SCD patients.
Collapse
Affiliation(s)
- John D. Belcher
- Department of Medicine, Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Minneapolis, Minnesota, United States of America
- * E-mail:
| | - Chunsheng Chen
- Department of Medicine, Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Minneapolis, Minnesota, United States of America
| | - Julia Nguyen
- Department of Medicine, Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Minneapolis, Minnesota, United States of America
| | - Fuad Abdulla
- Department of Medicine, Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Minneapolis, Minnesota, United States of America
| | - Phong Nguyen
- Department of Medicine, Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Minneapolis, Minnesota, United States of America
| | - Minh Nguyen
- Department of Medicine, Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Minneapolis, Minnesota, United States of America
| | - Nicole M. Okeley
- Seattle Genetics, Inc., Bothell, Washington, United States of America
| | | | - Peter D. Senter
- Seattle Genetics, Inc., Bothell, Washington, United States of America
| | - Gregory M. Vercellotti
- Department of Medicine, Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Minneapolis, Minnesota, United States of America
| |
Collapse
|
19
|
Sumagin R, Sarelius IH. Emerging understanding of roles for arterioles in inflammation. Microcirculation 2014; 20:679-92. [PMID: 23701383 DOI: 10.1111/micc.12068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 05/20/2013] [Indexed: 12/20/2022]
Abstract
Arterioles, capillaries, and venules all actively change their cellular functions and phenotypes during inflammation in ways that are essential for maintenance of homeostasis and self-defense, and are also associated with many inflammatory disorders. ECs, together with pericytes and ECM proteins, can regulate blood flow, the coagulation cascade, fluid and solute exchange, and leukocyte trafficking. While capillary and venular functions in inflammation are well characterized, the arteriolar contribution to inflammation has only recently come into focus. Arterioles differ from venules in structure, EC morphology, shear environment, expression, and distribution of surface ligands; hence, regulation and function of arteriolar wall cells during inflammation may also be distinct from venules. Recent work indicates that in response to proinflammatory stimuli, arterioles alter barrier function, and support leukocyte and platelet interactions through upregulation of adhesion molecules. This suggests that in addition to their role in blood flow regulation, arterioles may also participate in inflammatory responses. In this review, we will discuss mechanisms that characterize arteriolar responses to proinflammatory stimuli. We will detail how distinct arteriolar features contribute to regulation of barrier function and leukocyte-EC interactions in inflammation, and further highlight the potential priming effects of arteriolar responses on venular function and progression of inflammatory responses.
Collapse
Affiliation(s)
- Ronen Sumagin
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | | |
Collapse
|
20
|
Ghoshal P, Rajendran M, Odo N, Ikuta T. Glycosylation inhibitors efficiently inhibit P-selectin-mediated cell adhesion to endothelial cells. PLoS One 2014; 9:e99363. [PMID: 24945938 PMCID: PMC4063735 DOI: 10.1371/journal.pone.0099363] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/13/2014] [Indexed: 02/02/2023] Open
Abstract
Adhesion molecules play a critical role in the adhesive interactions of multiple cell types in sickle cell disease (SCD). We previously showed that anti-P-selectin aptamer efficiently inhibits cell adhesion to endothelial cells (ECs) and permits SCD mice to survive hypoxic stress. In an effort to discover new mechanisms with which to inhibit P-selectin, we examined the role of glycosylation. P-selectin is a 90 kDa protein but was found to migrate as 90 and 140 kDa bands on gel electrophoresis. When P-selectin isolated from ECs was digested with peptide N-glycosidase F, but not O-glycosidase, the 140 kDa band was lost and the 90 kDa band was enhanced. Treatment of ECs with tunicamycin, an N-glycosylation inhibitor, suppressed CD62P (P-selectin) expression on the cell surface as well as the 140 kDa form in the cytoplasm. These results indicate that the 140 kDa band is N-glycosylated and glycosylation is critical for cell surface expression of P-selectin in ECs. Thrombin, which stimulates P-selectin expression on ECs, induced AKT phosphorylation, whereas tunicamycin inhibited AKT phosphorylation, suggesting that AKT signaling is involved in the tunicamycin-mediated inhibition of P-selectin expression. Importantly, the adhesion of sickle red blood cells (sRBCs) and leukocytes to ECs induced by thrombin or hypoxia was markedly inhibited by two structurally distinct glycosylation inhibitors; the levels of which were comparable to that of a P-selectin monoclonal antibody which most strongly inhibited cell adhesion in vivo. Knockdown studies of P-selectin using short-hairpin RNAs in ECs suppressed sRBC adhesion, indicating a legitimate role for P-selectin in sRBC adhesion. Together, these results demonstrate that P-selectin expression on ECs is regulated in part by glycosylation mechanisms and that glycosylation inhibitors efficiently reduce the adhesion of sRBCs and leukocytes to ECs. Glycosylation inhibitors may lead to a novel therapy which inhibits cell adhesion in SCD.
Collapse
Affiliation(s)
- Pushpankur Ghoshal
- Department of Anesthesiology and Perioperative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
| | - Mythilypriya Rajendran
- Department of Anesthesiology and Perioperative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
| | - Nadine Odo
- Department of Anesthesiology and Perioperative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
| | - Tohru Ikuta
- Department of Anesthesiology and Perioperative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
| |
Collapse
|
21
|
Heme triggers TLR4 signaling leading to endothelial cell activation and vaso-occlusion in murine sickle cell disease. Blood 2013; 123:377-90. [PMID: 24277079 DOI: 10.1182/blood-2013-04-495887] [Citation(s) in RCA: 525] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Treatment of sickle cell disease (SCD) is hampered by incomplete understanding of pathways linking hemolysis to vaso-occlusion. We investigated these pathways in transgenic sickle mice. Infusion of hemoglobin or heme triggered vaso-occlusion in sickle, but not normal, mice. Methemoglobin, but not heme-stabilized cyanomethemoglobin, induced vaso-occlusion, indicating heme liberation is necessary. In corroboration, hemoglobin-induced vaso-occlusion was blocked by the methemoglobin reducing agent methylene blue, haptoglobin, or the heme-binding protein hemopexin. Untreated HbSS mice, but not HbAA mice, exhibited ∼10% vaso-occlusion in steady state that was inhibited by haptoglobin or hemopexin infusion. Antibody blockade of adhesion molecules P-selectin, von Willebrand factor (VWF), E-selectin, vascular cell adhesion molecule 1, intercellular adhesion molecule 1, platelet endothelial cell (EC) adhesion molecule 1, α4β1, or αVβ3 integrin prevented vaso-occlusion. Heme rapidly (5 minutes) mobilized Weibel-Palade body (WPB) P-selectin and VWF onto EC and vessel wall surfaces and activated EC nuclear factor κB (NF-κB). This was mediated by TLR4 as TAK-242 blocked WPB degranulation, NF-κB activation, vaso-occlusion, leukocyte rolling/adhesion, and heme lethality. TLR4(-/-) mice transplanted with TLR4(+/+) sickle bone marrow exhibited no heme-induced vaso-occlusion. The TLR4 agonist lipopolysaccharide (LPS) activated ECs and triggered vaso-occlusion that was inhibited by TAK-242, linking hemolysis- and infection-induced vaso-occlusive crises to TLR4 signaling. Heme and LPS failed to activate VWF and NF-κB in TLR4(-/-) ECs. Anti-LPS immunoglobulin G blocked LPS-induced, but not heme-induced, vaso-occlusion, illustrating LPS-independent TLR4 signaling by heme. Inhibition of protein kinase C, NADPH oxidase, or antioxidant treatment blocked heme-mediated stasis, WPB degranulation, and oxidant production. We conclude that intravascular hemolysis in SCD releases heme that activates endothelial TLR4 signaling leading to WPB degranulation, NF-κB activation, and vaso-occlusion.
Collapse
|
22
|
Stokes KY, Granger DN. Platelets: a critical link between inflammation and microvascular dysfunction. J Physiol 2011; 590:1023-34. [PMID: 22183721 DOI: 10.1113/jphysiol.2011.225417] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Inflammation is an underlying feature of a variety of human diseases. An important manifestation of this pathophysiological response is microvascular dysfunction, which includes the activation of vascular endothelial cells, and circulating leucocytes and platelets. While endothelial cells and leucocytes are widely accepted as critical players in the microvascular alterations induced by inflammation, recent attention has focused on the modulatory role of platelets, which act both as effector and target cells in inflamed microvessels. Evidence is presented to demonstrate the capacity for 'cross-talk' between platelets and other cells (endothelial cells, leucocytes) that contribute to an inflammatory response, and to illustrate the pathophysiological consequences of these interactions of platelets with other cells within the microvasculature.
Collapse
Affiliation(s)
- Karen Y Stokes
- Department of Molecular & Cellular Physiology, LSU Health Sciences Centre-Shreveport, 1501 Kings Highway Shreveport, LA 71130-3932, USA.
| | | |
Collapse
|
23
|
Morgan SP. Can new optical techniques for in vivo imaging and flow cytometry of the microcirculation benefit sickle cell disease research? Cytometry A 2011; 79:766-74. [PMID: 21744494 DOI: 10.1002/cyto.a.21101] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 04/25/2011] [Accepted: 06/09/2011] [Indexed: 01/24/2023]
Abstract
Intravital microscopy is a valuable tool for research into sickle cell disease with studies being carried out on transgenic mice and human volunteers. The method has helped to develop an explanation for sickle crises based on cell adhesion to the vascular endothelium followed by logjamming of rigid sickle cells and has stimulated much research into new treatments. In recent years there have been numerous new optical techniques developed for imaging the microcirculation and understanding the circulation of cells within the body, many of which have been further developed into in vivo flow cytometry techniques. This brief review highlights some of the progress made to date in the understanding of sickle cell disease using intravital microscopy. New techniques for imaging the microcirculation and their potential uses in understanding sickle cell disease are discussed.
Collapse
Affiliation(s)
- Stephen P Morgan
- Electrical Systems and Optics Research Division, Faculty of Engineering, University of Nottingham, University Park, Nottingham. NG7 2RD, United Kingdom.
| |
Collapse
|
24
|
Mechanisms of enhanced thrombus formation in cerebral microvessels of mice expressing hemoglobin-S. Blood 2011; 117:4125-33. [PMID: 21304105 DOI: 10.1182/blood-2010-08-301366] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The microvasculature assumes an inflammatory and procoagulant state in a variety of different diseases, including sickle cell disease (SCD), which may contribute to the high incidence of ischemic stroke in these patients. This study provides evidence for accelerated thrombus formation in arterioles and venules in the cerebral vasculature of mice that express hemoglobin-S (β(s) mice). Enhanced microvascular thrombosis in β(s) mice was blunted by immunologic or genetic interventions that target tissue factor, endothelial protein C receptor, activated protein C, or thrombin. Platelets from β(s) mice also exhibited enhanced aggregation velocity after stimulation with thrombin but not ADP. Neutropenia also protected against the enhanced thrombosis response in β(s) mice. These results indicate that the cerebral microvasculature is rendered vulnerable to thrombus formation in β(s) mice via a neutrophil-dependent mechanism that is associated with an increased formation of and enhanced platelet sensitivity to thrombin.
Collapse
|
25
|
Proença-Ferreira R, Franco-Penteado CF, Traina F, Saad STO, Costa FF, Conran N. Increased adhesive properties of platelets in sickle cell disease: roles for alphaIIb beta3-mediated ligand binding, diminished cAMP signalling and increased phosphodiesterase 3A activity. Br J Haematol 2010; 149:280-8. [PMID: 20136824 DOI: 10.1111/j.1365-2141.2010.08087.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Whilst high pro-coagulant activity is reported in sickle cell disease (SCD), the precise role of platelets (PLTs) in SCD inflammatory and vaso-occlusive processes is unclear. Adhesion of PLTs from healthy controls (CON), SCD individuals (SCD) and SCD patients on hydroxycarbamide (SCDHC) to fibrinogen (FB) was compared using static adhesion assays. PLT adhesion molecules and intraplatelet cyclic adenosine monophosphate (icAMP) were observed by flow cytometry and enzyme-linked immunosorbent assay. SCD-PLTs demonstrated significantly greater adhesion than CON-PLTs to FB. Participation of the alpha(IIb)beta(3)-integrin in SCD-PLT adhesion was implicated by increased alpha(IIb)beta(3) activation and data showing that an alpha(IIb)beta(3)-function-inhibiting antibody significantly diminished SCD-PLT adhesion to FB. Platelet activation was potentiated by reductions in icAMP; cAMP levels were decreased in SCD-PLTs, being comparable to those of thrombin-stimulated CON-PLTs. Furthermore, SCD-PLT adhesion to FB was significantly reduced by cilostazol, an inhibitor of cAMP-hydrolyzing phosphodiesterase 3A (PDE3A). Both alpha(IIb)beta(3)-integrin activation and icAMP correlated significantly with fetal haemoglobin in SCD. Accordingly, hydroxycarbamide therapy was associated with lower PLT adhesion and higher icAMP. SCD-PLTs may be capable of adhering to proteins encountered on the inflamed vascular wall and, potentially, participate in vaso-occlusive processes. Hydroxycarbamide and, speculatively, nitric oxide donor or cyclic-nucleotide-targeted therapies may aid in the reversal of PLT adhesive properties in SCD.
Collapse
|
26
|
Abstract
Heme is an essential molecule in aerobic organisms. Heme consists of protoporphyrin IX and a ferrous (Fe(2+)) iron atom, which has high affinity for oxygen (O(2)). Hemoglobin, the major oxygen-carrying protein in blood, is the most abundant heme-protein in animals and humans. Hemoglobin consists of four globin subunits (alpha(2)beta(2)), with each subunit carrying a heme group. Ferrous (Fe(2+)) hemoglobin is easily oxidized in circulation to ferric (Fe(3+)) hemoglobin, which readily releases free hemin. Hemin is hydrophobic and intercalates into cell membranes. Hydrogen peroxide can split the heme ring and release "free" redox-active iron, which catalytically amplifies the production of reactive oxygen species. These oxidants can oxidize lipids, proteins, and DNA; activate cell-signaling pathways and oxidant-sensitive, proinflammatory transcription factors; alter protein expression; perturb membrane channels; and induce apoptosis and cell death. Heme-derived oxidants induce recruitment of leukocytes, platelets, and red blood cells to the vessel wall; oxidize low-density lipoproteins; and consume nitric oxide. Heme metabolism, extracellular and intracellular defenses against heme, and cellular cytoprotective adaptations are emphasized. Sickle cell disease, an archetypal example of hemolysis, heme-induced oxidative stress, and cytoprotective adaptation, is reviewed.
Collapse
Affiliation(s)
- John D Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Minneapolis, Minnesota 55455, USA
| | | | | | | | | |
Collapse
|
27
|
|
28
|
|
29
|
Abstract
Periodic recurrence of painful vaso-occlusive crisis is the defining feature of sickle cell disease. Among multiple pathologies associated with this disease, sickle red cell-endothelium interaction has been implicated as a potential initiating mechanism in vaso-occlusive events. This review focuses on various interrelated mechanisms involved in human sickle red cell adhesion. We discuss in vitro and microcirculatory findings on sickle red cell adhesion, its potential role in vaso-occlusion, and the current understanding of receptor-ligand interactions involved in this pathological phenomenon. In addition, we discuss the contribution of other cellular interactions (leukocytes recruitment and leukocyte-red cell interaction) to vaso-occlusion, as observed in transgenic sickle mouse models. Emphasis is given to recently discovered adhesion molecules that play a predominant role in mediating human sickle red cell adhesion. Finally, we analyze various therapeutic approaches for inhibiting sickle red cell adhesion by targeting adhesion molecules and also consider therapeutic strategies that target stimuli involved in endothelial activation and initiation of adhesion.
Collapse
Affiliation(s)
- Dhananjay K Kaul
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461, USA.
| | | | | |
Collapse
|
30
|
|
31
|
Abstract
Spectrin is the backbone of the erythroid cytoskeleton; sph/sph mice have severe hereditary spherocytosis (HS) because of a mutation in the murine erythroid alpha-spectrin gene. sph/sph mice have a high incidence of thrombosis and infarction in multiple tissues, suggesting significant vascular dysfunction. In the current study, we provide evidence for both pulmonary and systemic vascular dysfunction in sph/sph mice. We found increased levels of soluble cell adhesion molecules in sph/sph mice, suggesting activation of the vascular endothelium. We hypothesized that plasma hemoglobin released by intravascular hemolysis initiates endothelial injury through nitric oxide (NO) scavenging and oxidative damage. Likewise, electron paramagnetic resonance spectroscopy showed that plasma hemoglobin is much greater in sph/sph mice. Moreover, plasma from sph/sph mice had significantly higher oxidative potential. Finally, xanthine oxidase, a potent superoxide generator, is decreased in subpopulations of liver hepatocytes and increased on liver endothelium in sph/sph mice. These results indicate that vasoregulation is abnormal, and NO-based vasoregulatory mechanisms particularly impaired, in sph/sph mice. Together, these data indicate that sph/sph mice with severe HS have increased plasma hemoglobin and NO scavenging capacity, likely contributing to aberrant vasoregulation and initiating oxidative damage.
Collapse
|
32
|
van Beers EJ, Goedhart PT, Unger M, Biemond BJ, Ince C. Normal sublingual microcirculation during painful crisis in sickle cell disease. Microvasc Res 2008; 76:57-60. [DOI: 10.1016/j.mvr.2008.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Revised: 01/19/2008] [Accepted: 02/13/2008] [Indexed: 11/25/2022]
|
33
|
|
34
|
Schmitt-Sody M, Metz P, Gottschalk O, Birkenmaier C, Zysk S, Veihelmann A, Jansson V. Platelet P-selectin is significantly involved in leukocyte-endothelial cell interaction in murine antigen-induced arthritis. Platelets 2007; 18:365-72. [PMID: 17654306 DOI: 10.1080/09537100701191315] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
There is growing evidence that platelets play an important role in the development and maintenance of rheumatoid arthritis. Activation and adherence of platelets in the synovial microcirculation might be in part responsible for endothelial damage and activation of leukocytes. Recent findings show a direct influence of P-selectin on platelet- and leukocyte-endothelial cell interaction in mice with Antigen-induced Arthritis (AiA). P-selectin is only expressed by platelets and endothelial cells, not by leukocytes. Therefore, the aim of the present study was to investigate the differential influence of platelet and endothelial P-selectin on the extent of inflammation in AiA. AiA was induced in wild-type mice and in P-selectin-deficient mice from the same genetic background (four groups: each n = 7). Intravital fluorescence microscopy (IVM) was used to visualize platelets and leukocytes in the synovial microcirculation at day 8 after AiA. Platelets from either strain were fluorescence-labelled ex vivo and transferred into either strain. We were able to demonstrate a significant decrease of platelet- and leukocyte-endothelial cell interaction in P-selectin-deficient mice with AiA in comparison to wild-type mice with AiA. When wild-type platelets were donated into P-selectin-deficient AiA recipients, the leukocyte-endothelial cell interaction was significantly increased compared to the group consisting of P-selectin-deficient recipient and donor mice. These are the first in vivo results showing that the P-selectin stored in platelets is at least partly responsible for the leukocyte-endothelial cell interaction and the resulting tissue damage in AiA. In the future, a suppression of platelet P-selectin could potentially become a treatment option for reducing the effects of rheumatoid arthritis.
Collapse
MESH Headings
- Animals
- Antigens/toxicity
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/genetics
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Arthritis, Experimental/therapy
- Arthritis, Rheumatoid/chemically induced
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/therapy
- Blood Platelets/immunology
- Blood Platelets/pathology
- Cell Adhesion/immunology
- Cell Communication/genetics
- Cell Communication/immunology
- Endothelial Cells/immunology
- Endothelial Cells/pathology
- Female
- Leukocytes/immunology
- Leukocytes/pathology
- Mice
- Mice, Knockout
- P-Selectin/genetics
- P-Selectin/immunology
- Platelet Adhesiveness/genetics
- Platelet Adhesiveness/immunology
- Platelet Transfusion
- Synovial Membrane/immunology
- Synovial Membrane/pathology
Collapse
Affiliation(s)
- M Schmitt-Sody
- Department of Orthopaedics, Ludwig Maximilians University, Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
35
|
Wood KC, Granger DN. Sickle cell disease: role of reactive oxygen and nitrogen metabolites. Clin Exp Pharmacol Physiol 2007; 34:926-32. [PMID: 17645642 DOI: 10.1111/j.1440-1681.2007.04639.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
1. Sickle cell disease (SCD) is an inherited disorder of haemoglobin synthesis that is associated with significant morbidity and mortality due to sequelae of episodic vaso-occlusive events: pain crises and multiorgan damage. The microvascular responses to the initiation, progression and resolution of vaso-occlusive events are consistent with an inflammatory phenotype as suggested by activation of multiple cell types, an oxidatively stressed environment and endothelial cell dysfunction. 2. Decreased anti-oxidant defences in SCD patients and mice are accompanied by activation of enzymatic (NADPH oxidase, xanthine oxidase) and non-enzymatic (sickle haemoglobin auto-oxidation) sources of reactive oxygen species. The resultant oxidative stress leads to dysfunction/activation of arteriolar and venular endothelial cells, resulting in impaired vasomotor function and blood cell-endothelial cell adhesion. 3. Changes in substrate and cofactor availability for endothelial cell nitric oxide synthase may underlie reactive oxygen- and nitrogen-induced events that contribute to SCD-induced vasculopathy. 4. The emerging role of reactive oxygen and nitrogen species in the pathogenesis of SCD provides a platform for the development of novel agents to treat this painful and lethal disease.
Collapse
Affiliation(s)
- Katherine C Wood
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932, USA
| | | |
Collapse
|
36
|
Finnegan EM, Turhan A, Golan DE, Barabino GA. Adherent leukocytes capture sickle erythrocytes in an in vitro flow model of vaso-occlusion. Am J Hematol 2007; 82:266-75. [PMID: 17094094 DOI: 10.1002/ajh.20819] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Recent in vivo studies suggest that adherent leukocytes bind RBCs and contribute to the microvascular pathology that characterizes sickle cell disease (SCD). A parallel-plate flow assay was used: to investigate the capture of RBCs by adherent neutrophils, monocytes, and T-lymphocytes; to examine whether RBC capture is elevated in patients with SCD; and to determine whether hydroxyurea (HU) therapy affects these interactions. Four measures of cell-cell adhesion were used: adhesion of leukocytes to TNF-alpha-treated human umbilical vein endothelial cells (HUVECs), percent of adherent leukocytes that captured RBCs, number of RBCs captured per interacting leukocyte, and duration of RBC capture. Leukocyte subpopulations from sickle patients were more adherent to activated ECs and captured more RBCs per interacting leukocyte than the corresponding subpopulations from healthy controls. While HU did not affect leukocyte adhesion to activated ECs, it reduced the proportion of adherent leukocytes that captured RBCs, as well as the number of RBCs captured per neutrophil. T-lymphocytes demonstrated elevated adhesion in all measures, and may be the leukocyte subpopulation whose behavior is most altered in SCD. Our findings suggest that neutrophils, monocytes, and T-lymphocytes could all be involved in adhesive interactions with autologous RBCs in patients with SCD.
Collapse
Affiliation(s)
- Eileen M Finnegan
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
37
|
Abstract
Endothelial cells, which form the inner cellular lining of blood vessels and lymphatics, display remarkable heterogeneity in structure and function. This is the second of a 2-part review on the phenotypic heterogeneity of blood vessel endothelial cells. The first part discusses the scope, the underlying mechanisms, and the diagnostic and therapeutic implications of phenotypic heterogeneity. Here, these principles are applied to an understanding of organ-specific phenotypes in representative vascular beds including arteries and veins, heart, lung, liver, and kidney. The goal is to underscore the importance of site-specific properties of the endothelium in mediating homeostasis and focal vascular pathology, while at the same time emphasizing the value of approaching the endothelium as an integrated system.
Collapse
Affiliation(s)
- William C Aird
- Division of Molecular and Vascular Medicine, Department of Medicine, and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass 02215, USA.
| |
Collapse
|
38
|
Dasgupta T, Hebbel RP, Kaul DK. Protective effect of arginine on oxidative stress in transgenic sickle mouse models. Free Radic Biol Med 2006; 41:1771-80. [PMID: 17157180 PMCID: PMC1948977 DOI: 10.1016/j.freeradbiomed.2006.08.025] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Revised: 07/31/2006] [Accepted: 08/11/2006] [Indexed: 01/24/2023]
Abstract
Sickle cell disease (SCD) is characterized by reperfusion injury and chronic oxidative stress. Oxidative stress and hemolysis in SCD result in inactivation of nitric oxide (NO) and depleted arginine levels. We hypothesized that augmenting NO production by arginine supplementation will reduce oxidative stress in SCD. To this end, we measured the effect of arginine (5% in mouse chow) on NO metabolites (NOx), lipid peroxidation (LPO), and selected antioxidants in transgenic sickle mouse models. Untreated transgenic sickle (NY1DD) mice (expressing approximately 75% beta(S)-globin of all beta-globins; mild pathology) and knockout sickle (BERK) mice (expressing exclusively hemoglobin S; severe pathology) showed reduced NOx levels and significant increases in the liver LPO compared with C57BL mice, with BERK mice showing maximal LPO increase in accordance with the disease severity. This was accompanied by reduced activity of antioxidants (glutathione, total superoxide dismutase, catalase, and glutathione peroxidase). However, GSH levels in BERK were higher than in NY1DD mice, indicating a protective response to greater oxidative stress. Importantly, dietary arginine significantly increased NOx levels, reduced LPO, and increased antioxidants in both sickle mouse models. In contrast, nitro-L-arginine methylester, a potent nonselective NOS inhibitor, worsened the oxidative stress in NY1DD mice. Thus, the attenuating effect of arginine on oxidative stress in SCD mice suggests its potential application in the management of this disease.
Collapse
Affiliation(s)
- Trisha Dasgupta
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461
| | - Robert P Hebbel
- Department of Medicine, University of Minnesota Medical School, 420 Delware Street, SE, Minneapolis, MN 55455
| | - Dhananjay K. Kaul
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461
| |
Collapse
|
39
|
Petnehazy T, Cooper D, Stokes KY, Russell J, Wood KC, Granger DN. Angiotensin II type 1 receptors and the intestinal microvascular dysfunction induced by ischemia and reperfusion. Am J Physiol Gastrointest Liver Physiol 2006; 290:G1203-10. [PMID: 16469824 DOI: 10.1152/ajpgi.00578.2005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The acute phase of intestinal ischemia-reperfusion (I/R) injury is mediated by leukocytes and is characterized by oxidative stress and blood cell recruitment. Upregulation of angiotensin II type 1 receptors (AT1-R) has been implicated in the pathogenesis of conditions associated with oxidative stress. The AT1-R-antagonist Losartan (Los) attenuates leukocyte recruitment following I/R. However, the role of AT1-R in intestinal I/R injury and the associated platelet-leukocyte interactions remains unclear. The objective of this study was to define the contribution of AT1-R to I/R-induced blood cell recruitment in intestinal venules. Leukocyte and platelet adhesion were quantified by intravital microscopy in the small bowel of C57Bl/6 [wild-type (WT)] mice exposed to sham operation or 45 min of ischemia and 4 h of reperfusion. A separate WT group received Los for 7 days before gut I/R (WT-I/R + Los). AT1-R bone marrow chimeras that express AT1-R on the vessel wall but not blood cells also underwent I/R. Platelet and leukocyte adhesion as well as AT1-R expression in the gut microvasculature were significantly elevated after I/R. All of these responses were attenuated in the WT-I/R + Los group, compared with untreated I/R mice. A comparable abrogation of I/R-induced blood cell adhesion was noted in AT1-R bone marrow chimeras. I/R-induced platelet adhesion was unaltered in mice overexpressing Cu,Zn-SOD or mice deficient in NAD(P)H oxidase. These data suggest that although gut I/R upregulates endothelial expression of AT1-R, engagement of these angiotensin II receptors on blood cells is more important in eliciting the prothrombogenic and proinflammatory state observed in postischemic gut venules, through a superoxide-independent pathway.
Collapse
Affiliation(s)
- Thomas Petnehazy
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, 71130, USA
| | | | | | | | | | | |
Collapse
|
40
|
Wood KC, Hebbel RP, Lefer DJ, Granger DN. Critical role of endothelial cell-derived nitric oxide synthase in sickle cell disease-induced microvascular dysfunction. Free Radic Biol Med 2006; 40:1443-53. [PMID: 16631534 DOI: 10.1016/j.freeradbiomed.2005.12.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Revised: 12/07/2005] [Accepted: 12/14/2005] [Indexed: 01/18/2023]
Abstract
Superoxide, which can limit nitric oxide bioavailability, has been implicated in blood cell-vessel wall interactions observed in sickle cell transgenic (beta(S)) mice. Here we report that nonselective chemical inhibition of nitric oxide synthase isoforms dramatically reduces the enhanced leukocyte and platelet adhesion normally observed in cerebral venules of beta(S) mice. Although genetic deficiency of vascular wall inducible nitric oxide synthase does not alter adhesion responses in beta(S) mice, a significant attenuation is noted in beta(S) mice with vascular wall endothelial nitric oxide synthase (eNOS) deficiency, while the adhesion responses are exacerbated when eNOS is overexpressed in microvessels. The eNOS-mediated enhancement of blood cell adhesion is reversible by pretreatment with sepiapterin (which generates the eNOS cofactor tetrahydrobiopterin) or polyethyleneglycol-superoxide dismutase, implicating a role for eNOS-dependent superoxide production. These findings suggest that an imbalance between eNOS-derived nitric oxide and superoxide, both generated by the vessel wall, is critical to the proinflammatory and prothrombogenic phenotype that is assumed by the microvasculature in sickle cell disease.
Collapse
Affiliation(s)
- Katherine C Wood
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, 71130, USA
| | | | | | | |
Collapse
|
41
|
Canalli AA, Costa FF, Saad STO, Conran N. Granulocytic adhesive interactions and their role in sickle cell vaso-occlusion. ACTA ACUST UNITED AC 2006; 10:419-25. [PMID: 16273736 DOI: 10.1080/10245330500141259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Recent research has high-lighted the importance of leukocytes in sickle cell disease (SCD). Here we summarize evidence to show that the granulocytes may play a role in SCD due to their increased numbers and adhesive properties, facilitating their participation in the vaso-occlusive process.
Collapse
|
42
|
Petnehazy T, Stokes KY, Wood KC, Russell J, Granger DN. Role of Blood Cell–Associated AT1 Receptors in the Microvascular Responses to Hypercholesterolemia. Arterioscler Thromb Vasc Biol 2006; 26:313-8. [PMID: 16254200 DOI: 10.1161/01.atv.0000193625.32499.71] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Hypercholesterolemia elicits a proinflammatory and prothrombogenic phenotype in the microvasculature that is characterized by activation and adhesion of blood cells. The angiotensin II receptor-1 antagonist Losartan prevents the induction of these responses. The objective of this study was to determine the relative contributions of blood cell-associated versus endothelium-associated AT1a-R to these hypercholesterolemia-induced microvascular alterations. METHODS AND RESULTS Leukocyte adhesion and emigration and platelet adhesion were quantified by intravital microscopy in postcapillary venules. C57Bl/6 mice were placed on a normal (ND) or high-cholesterol (HCD) diet for 2 weeks. AT1a-R bone marrow chimeras that express AT1a-R on the vessel wall but not blood cells and AT1a-R knockouts were placed on HCD. Venular shear rate was comparable in all groups. Platelet and leukocyte adhesion and leukocyte emigration were significantly increased in HCD mice versus ND. Leukocyte recruitment was significantly reduced in the HCD-AT1a-R bone marrow chimera group, whereas platelet adhesion remained at HCD levels. However, in HCD-AT1a-R knockout mice, platelet and leukocyte adhesion were reduced to ND levels. CONCLUSIONS These data indicate that the platelet-vessel wall adhesion elicited by hypercholesterolemia is mediated by AT1a-R engagement on the endothelial cell rather than the platelet, whereas leukocyte recruitment is mediated by blood cell-associated AT1a-R.
Collapse
Affiliation(s)
- Thomas Petnehazy
- Department of Molecular and Cellular Physiology, Louisiana State University, Health Sciences Center, Shreveport, LA 71130-3932, USA
| | | | | | | | | |
Collapse
|
43
|
Wood K, Jawahar A, Smelley C, Mullapudi S, DeLaune A, Nanda A, Granger DN. Exposure of Brain to High-Dose, Focused Gamma Rays Irradiation Produces Increase in Leukocytes-Adhesion and Pavementing in Small Intracerebral Blood Vessels. Neurosurgery 2005; 57:1282-8; discussion 1282-8. [PMID: 16331177 DOI: 10.1227/01.neu.0000187318.17832.55] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Radiosurgery is used to destroy a predetermined target within the brain, with minimal radiation injury to the surrounding tissue. We hereby present our in vivo model to study the effects of single-session, high-dose radiation on the cerebral vessels that are targeted with radiosurgery using the Leksell Gamma Knife. METHODS The study was conducted in 29 adult male WT C57BL/6J mice weighing 21 to 28 g (6-8 wk old). The animals were exposed to 100 Gy single-session focused gamma ray irradiation using the Leksell Gamma Knife, and subsequently underwent intravital microscopy at different time intervals to study leukocytes and platelets adhesion patterns to the endothelium of the irradiated cerebral micro-vessels. RESULTS The leukocyte adhesion response showed a bell-shaped curve upon quantitative analysis with a steady increase in the number of adherent cells during the first four hours and a subsequent plateau response that was maintained during the next 24 hours. The platelet adhesion response did not demonstrate any particular pattern similar to the leukocyte response. CONCLUSION The experiment was able to establish in vivo increased leukocyte adhesion to the cerebral vascular endothelial cells in response to radiation injury and elaborate the time frame within which the leukocyte adhesion response increases, reaches a peak and then starts decreasing.
Collapse
Affiliation(s)
- Katherine Wood
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Kato GJ, Martyr S, Blackwelder WC, Nichols JS, Coles WA, Hunter LA, Brennan ML, Hazen SL, Gladwin MT. Levels of soluble endothelium-derived adhesion molecules in patients with sickle cell disease are associated with pulmonary hypertension, organ dysfunction, and mortality. Br J Haematol 2005; 130:943-53. [PMID: 16156864 PMCID: PMC2065864 DOI: 10.1111/j.1365-2141.2005.05701.x] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Endothelial cell adhesion molecules orchestrate the recruitment and binding of inflammatory cells to vascular endothelium. With endothelial dysfunction and vascular injury, the levels of endothelial bound and soluble adhesion molecules increase. Such expression is modulated by nitric oxide (NO), and in patients with sickle cell disease (SCD), these levels are inversely associated with measures of NO bioavailability. To further evaluate the role of endothelial dysfunction in a population study of SCD, we have measured the levels of soluble endothelium-derived adhesion molecules in the plasma specimens of 160 adult patients with SCD during steady state. Consistent with a link between endothelial dysfunction and end-organ disease, we found that higher levels of soluble vascular cell adhesion molecule-1 (sVCAM-1) were associated with markers indicating renal dysfunction and hepatic impairment. Analysis of soluble intercellular cell adhesion molecule-1 (sICAM-1), sE-selectin and sP-selectin levels indicated partially overlapping associations with sVCAM-1, with an additional association with inflammatory stress and triglyceride levels. Importantly, increased soluble adhesion molecule expression correlated with severity of pulmonary hypertension, a clinical manifestation of endothelial dysfunction. Soluble VCAM-1, ICAM-1, and E-selectin were independently associated with the risk of mortality in this cohort. Our data are consistent with steady state levels of soluble adhesion molecules as markers of pulmonary hypertension and risk of death.
Collapse
Affiliation(s)
- Gregory J Kato
- Vascular Therapeutics Section, Cardiovascular Branch, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The vaso-occlusion model has evolved impressively over the past several decades from polymerization-based concepts to a complex, wide-ranging schema that involves multistep, heterogeneous, and interdependent interactions among sickle erythrocytes (SSRBCs), adherent leukocytes, endothelial cells, plasma proteins, and other factors. Endothelial activation, induced directly or indirectly by the proinflammatory behavior of SSRBCs, is the most likely initiating step toward vaso-occlusion. Given the complexity and dynamic relationships of the potential mechanisms leading to vaso-occlusion, further in vivo studies in relevant sickle cell animal models will most likely yield the greatest advances and promote the development of novel, more effective therapeutic strategies.
Collapse
Affiliation(s)
- Elaine Y Chiang
- Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | |
Collapse
|
46
|
Wood KC, Hebbel RP, Granger DN. Endothelial cell NADPH oxidase mediates the cerebral microvascular dysfunction in sickle cell transgenic mice. FASEB J 2005; 19:989-91. [PMID: 15923406 DOI: 10.1096/fj.04-3218fje] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Although blood cell-endothelial cell adhesion and oxidative stress have been implicated in the pathogenesis of sickle cell disease (SCD), the nature of the linkage between these vascular responses in SCD remains unclear. The objective of this study was to determine whether superoxide derived from endothelial cell-associated NADPH oxidase mediates the leukocyte-endothelial (L/E) and platelet-endothelial cell (P/E) adhesion that is observed in the cerebral microvasculature of sickle cell transgenic (betaS) mice. Intravital fluorescence microscopy was used to monitor L/E and P/E adhesion in brain postcapillary venules of wild-type (WT), SOD1 transgenic (SOD1-TgN), and gp91phox (NADPH oxidase)-deficient mice that were transplanted with bone marrow from betaS mice. Hypoxia/reoxygenation (H/R) yielded intense P/E and L/E adhesion responses in cerebral venules of betaS/WT chimeras that were significantly attenuated in both betaS/SOD1-TgN, and betaS/gp91phox-/- chimeras. Pretreatment of betaS/WT chimeras with the iron-chelator desferroxamine blunted the blood cell-endothelial cell adhesion responses to H/R, whereas pretreatment with the xanthine oxidase inhibitor allopurinol had no effect. These findings suggest that superoxide derived from endothelial cell NADPH-oxidase and catalytically active iron contribute to the proinflammatory and prothrombogenic responses associated with sickle cell disease.
Collapse
Affiliation(s)
- Katherine C Wood
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, Louisiana 71130-3932, USA
| | | | | |
Collapse
|
47
|
Canalli AA, Conran N, Fattori A, Saad STO, Costa FF. Increased adhesive properties of eosinophils in sickle cell disease. Exp Hematol 2004; 32:728-34. [PMID: 15308324 DOI: 10.1016/j.exphem.2004.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2004] [Revised: 04/23/2004] [Accepted: 04/29/2004] [Indexed: 11/16/2022]
Abstract
OBJECTIVE A role for leukocytes in vasoocclusion is becoming increasingly recognized. Here we investigate a possible role for the eosinophil in sickle cell anemia (SCA). PATIENTS AND METHODS Eosinophils were isolated from whole blood samples of 59 steady-state SCA homozygous SS and control individuals using Percoll gradient separation, followed by immunomagnetic sorting. Adhesion of cells to FN was evaluated using static adhesion assays (60 min, 37 degrees C, 5% CO2) and eosinophil adhesion molecular expression was observed by flow cytometry. RESULTS SCA patients presented significantly elevated absolute eosinophil numbers. Furthermore, eosinophils isolated from these individuals demonstrated a significantly greater adhesion ( approximately 70% increased) to fibronectin (FN) than normal eosinophils in static adhesion assays. Coincubation of eosinophils with integrin-blocking monoclonal antibodies in adhesion assays showed that an association of the VLA-4, LFA-1, and Mac-1 integrins mediate the adhesion of SCA eosinophils to FN. Flow cytometry demonstrated that the expression of these integrins, however, was unaltered on the surface of SCA eosinophils, suggesting that the increased SCA eosinophil adhesion is a consequence of increased integrin affinity or avidity. SCA eosinophil adhesion to FN was further increased by the cytokine, GM-CSF, indicating that inflammation processes may further stimulate eosinophil adhesion in these patients. CONCLUSION We report that eosinophil numbers may be significantly increased in SCA individuals and that these cells appear to exist in an activated state. Such alterations may indicate a role for the eosinophil in the vasooclusive process.
Collapse
|