Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Aug 1.
Published in final edited form as: Transl Res. 2022 Mar 27;246:1–14. doi: 10.1016/j.trsl.2022.03.010

Microfluidic Methods to Advance Mechanistic Understanding and Translational Research in Sickle Cell Disease

Melissa Azul *, Eudorah F Vital **, Wilbur A Lam **, David K Wood ***, Joan D Beckman ****
PMCID: PMC9218997  NIHMSID: NIHMS1793078  PMID: 35354090

Abstract

Sickle cell disease (SCD) is caused by a single point mutation in the β-globin gene of hemoglobin, which produces an altered sickle hemoglobin (HbS). The ability of HbS to polymerize under deoxygenated conditions gives rise to chronic hemolysis, oxidative stress, inflammation, and vaso-occlusion. Herein, we review recent findings using microfluidic technologies that have elucidated mechanisms of oxygen-dependent and -independent induction of HbS polymerization and how these mechanisms elicit the biophysical and inflammatory consequences in SCD pathophysiology. We also discuss how validation and use of microfluidics in SCD provides the opportunity to advance development of numerous therapeutic strategies, including curative gene therapies.

Keywords: sickle cell disease, red blood cell, microfluidics, rheology, oxygen tension

Introduction

Sickle cell disease (SCD) is an autosomal recessive disorder caused by a single point mutation in the β-globin gene of hemoglobin. This mutation results in altered sickle hemoglobin (HbS). Under deoxygenation, polymerization of HbS fibers occur within the red blood cell (RBC) and deforms the shape of the sickle RBC (sRBC). Additionally, a spectrum of molecular and biophysical consequences within the sRBC, on the sRBC’s surface, and in the local vascular environment simultaneously contribute to the altered hemorheology seen in SCD.1

Since the discovery of the single point mutation in Hb more than 60 years ago, our understanding of SCD pathophysiology has increased dramatically. However, the clinical phenotype varies widely, with marked heterogeneity in symptoms and severity. The heterogeneity in clinical phenotypes is poorly understood, hence there is no standard protocol to risk stratify according to disease severity. This lack of risk stratification contributes to difficulties in disease management by healthcare providers.2 For example, clinical symptoms can manifest as early as infancy and can continue to adulthood. Chronic disease burden often leads to end-organ damage and early mortality in those with SCD, who have a median life expectancy of 54 years.3 Among the many potential acute clinical manifestations of SCD, extreme episodes of pain known as vaso-occlusive episodes (VOEs) remain the clinical hallmark of the disease. VOEs are the most common reason for hospitalizations in SCD and can significantly reduce an individual’s quality of life.46

Development of new therapies for SCD has languished for many non-biologic reasons,7 however lack of sufficient and validated biomarkers to stratify patient phenotypes and serve as clinical trial endpoints has also hindered progress. Additionally, lack of biomarkers also has limited development of standardized treatments for SCD individuals. Despite these barriers, three new SCD-modifying therapies have been brought to market since 2017, with more under investigation — including potential curative gene therapies.8,9 With more therapies clinically available and many more under various stages of clinical development,8 now more than ever there is a need to fundamentally define how HbS and sRBCs drive SCD pathophysiology in order to accelerate development of new SCD therapies and provide evidence-based recommendations for personalized SCD management.

In the past decade, developments in microfluidic technologies have advanced the scientific understanding of sRBC biophysical interactions in SCD pathophysiology. Furthermore, new therapeutics targeting the sRBC and its biophysical composition are currently under development. Herein, we highlight how these technologies have advanced understanding and treatment of SCD and discuss how the development and use of these technologies can be incorporated into the validation pipelines of new SCD therapeutic approaches, including gene therapies.

Contributions of Oxygen-Dependent Microfluidics to Assess RBC Function

Intravital microscopy in SCD mouse models has been and remains a mainstay experimental tool for evaluating sRBC behavior.10 However, in vivo methods often lack quantitative control of vital biophysical parameters such as shear stress, blood vessel geometry, and mass transport, all of which may contribute to vaso-occlusive physiology. Microfluidics advancements allow for researchers to capture the dynamic effects of the deoxygenated-induced polymerization on hemorheology in a controlled, physiologically relevant system. In microfluidic devices that lack an endothelial cell layer, but otherwise recapitulate the in vitro vasculature anatomy and shear rates, blood can be perfused through the device at a constant pressure and exposed to decreased oxygen tensions while sRBC rheological flow response can be measured (Fig 2A). These microfluidic devices can be used to demonstrate that deoxygenation initiates alterations in sRBC rheology that are sufficient to increase viscosity, slow sRBC velocity, and impede sickle cell blood flow; these data provide additional insight into the dynamics of vaso-occlusive events (Fig 2B).1120 Furthermore, microfluidic studies show that sRBC velocity is reduced when oxygen tension is reduced and that there is an oxygen tension threshold where blood flow transitions from oxygen independent flow to oxygen-dependent flow (Fig 2C). Using these devices, oxygen tension can be decreased in a step-wise fashion to determine the lowest oxygen tension that can be tolerated without a reduction in sRBC velocity. For example, in a set of experiments using SCD patient blood, increased viscosity was observed at oxygen tensions as high as 80 mmHg, similar to that of arterial circulation.21 These findings support the arterial clinical manifestations seen in SCD such as stroke, moyamoya, and pulmonary hypertension.22,23 These studies suggest that arterial oxygen tensions may not be protective against stasis-mediated physiology.

Figure 2: Microfluidics devices to interrogate oxygen-dependent sickle RBC rheology.

Figure 2:

A. Photo of a microfluidic device for scale, top-down view. B. Still image of red blood cells moving through microfluidic device. C-D: Raw representative RBC velocity data from a SCD subject. C. RBC velocity is tracked through the device while environmental oxygen tension within the device is varied between 0 % and 21%. In the untreated sample, RBC velocity is dependent upon oxygen tension as RBC flow velocity is reduced with hypoxia. D. RBCs from the same SCD subject after 1-hour incubation with 500 μM of voxelotor at 37C ex vivo. Identical oxygen tension protocol as in B; however, voxelotor-treated RBCs experience smaller velocity reductions with hypoxia and reach oxygen-independent flow at lower oxygen tensions than untreated samples.

As microfluidics technologies have been useful in evaluating the biophysical consequences of HbS polymerization under deoxygenated conditions, they also have revealed several other mechanisms known to be fundamental to SCD pathologic rheology. By first observing the pathology in these devices, microfluidic studies can serve as an indicator of efficacy in current and potential treatments involved in the management of SCD. As many therapies target hypoxia-induced polymerization, oxygen-dependent devices particularly can serve as a marker of drug efficacy. For example, there has been a long-standing observation that the presence of fetal hemoglobin (HbF) reduces the concentration of HbS and thereby also reduces polymerization rates.2428 Therefore, the co-inheritance of additional genetic mutations that increase HbF concentrations such as hereditary persistence of HbF, or reduce HbS concentration, such as α-thalassemia or βC allele with βS may reduce disease severity.3 Based on this premise, induction of HbF has been a long-standing treatment for SCD, and hydroxyurea, which induces HbF in patients, has been approved for use in adults SCD since 1998.29,30 Indeed, in microfluidics models, sRBC treated with hydroxyurea demonstrate decreased oxygen-dependent viscosity changes compared to untreated blood.13,15 Collectively, these data offer a benchmark by which to compare efficacy and benefits of other HbF induction strategies, including gene therapies.

In addition to evaluating the effects of reducing HbS concentration by increasing HbF, oxygen-dependent microfluidics can also be used to screen for treatment effects of oxygen-dissociation agents on rheology. Approved since 2018 to prevent hemolysis in SCD, voxelotor (GBT 440) is thought to stabilize HbS in the oxygenated state, thereby reducing HbS polymerization. In a phase 3 randomized, double-blind, placebo-controlled trial, voxelotor demonstrated that patients’ hemoglobin improved by 1g/dL after 6 months of use along with a reduction in hemolysis markers.31 This clinical finding was confirmed in oxygen-dependent fluidic devices (Fig 2A), where sRBCs incubated ex vivo with voxelotor were resistant to a decrease in hypoxia-induced velocity slowing (Fig 2 B, D); this finding indicates less oxygen-dependent rheology. In a representative patient sample (Fig 2C-D), it appears that oxygen-independent flow is achieved around 6% O2 (45mmHg) in the voxelotor-treated sample. However, in the untreated sample (Fig 2C), there continues to be oxygen-dependent flow at the same oxygen tension. Collectively, these data suggest that the development, validation, and use of oxygen-sensitive microfluidic technologies may identify strategies to prevent oxygen-dependent sRBC rheologic changes and inform strategies to better treat and prevent SCD complications.

sRBC Dehydration through Activation of Cation Transport Channels during Deoxygenation

The ability to evaluate sRBC oxygen-sensitive rheology also permits for concurrent assessment of the interactions between oxygen-dependent HbS polymerization and sRBC cellular dehydration. HbS polymerization and decreased oxygen tensions lead to changes in RBC membrane permeability and permits the passage of cations and other small molecules to efflux through several channels (Fig 3A). Altered permeability ultimately leads to dehydration and increases in mean corpuscular hemoglobin concentration (MCHC). Increased MCHC in a sRBC increases HbS concentration, making cells at increased risk of polymerization. For example, under deoxygenation and polymerization, the Psickle channel, potentially identified as the PIEZO1 channel, becomes activated and allows an intracellular influx of sodium (Na+) and calcium (Ca2+) and an efflux of potassium (K+). Subsequent activation of the RBC Gardos (KCCN4) channel by the influx of Ca2+ causes a net loss of K+ and dehydration.32 Further dehydration also occurs due to activation of the K-Cl cotransporter. Additionally, Ca2+ efflux leads to increased exposure of pro-coagulant phospholipid phosphatidylserine (PS) and increased vascular adhesion.33,34 Using several methods to assess sRBC biophysical changes, several groups have noted that cyclic deoxygenation augments sRBC channel permeability leading to decreased deformability and significant shifts in RBC rheology (Table 1).16,17,20,3538 Overall, deoxygenation triggers HbS polymerization, which increases sRBC dehydration, which compounds polymerization and ultimately drives development of a rigid, dehydrated sRBC containing polymerized HbS.16,17

Figure 3: Implications of sickle red blood cell dehydration.

Figure 3:

A. Channels involved in sickle cell red cell dehydration. The Na-K pump (Na, K-ATPase) is more active in sickle RBCs. Monensin and other drugs can target this pump. The P-sickle channel, which may be PIEZO1, leads to Ca2+ influx. Furthermore, activation of GPCR and other cytokine-mediated channels leads to Ca2+ influx, which leads to Gardos channel activation. This leads to K+ and water efflux by aquaporins (AQ1). In SCD, the K-Cl cotransporter is also activated at low oxygen tensions, causing efflux of K+ and subsequently loss of water by aquaporins (AQ1).. Image created in Biorender. Fluid tonicity effects sRBC adherence to endothelium. B. Fluorescent-stained sRBC (red) adhered to human umblical vein endothelial cells. C. Bar graph illustrating how sRBC from 6 indivudals were less adherent to endothelium after exposure to hypotonic fluids. Figure modified and used with permission from Carden et. al. 2017.

Table 1:

Examples of microfluidics models applied for investigation of sickle RBC biophysical properties

Model Type Measurements Findings References
Microvasculature-on-a-chip Transit time
Time to occlusion
Adherent sickle cells
Platelet and fibrin deposition
Endothelial-sRBC interactions and geometry contribute to vaso-occlusion Carden et al. 2017. Microcirculation
Carden et al. 2017 Blood
Mathur et al. 2021. Bioeng Transl Med
Oxygen-Tension modulation
Microvasculature model
Shear-thinning
Oxygen-dependent velocity
Oxygen-dependent rheology
Oxygen-gradient and vascular size
Oxygen-dependent viscosity changes
Sickle Hb concentration increases blood viscosity with decreasing oxygen tension Wood et al. 2012. Sci Transl Med
Lu et al. 2018. Am J Hematol
Valdez et al. 2019. APL Bioeng
Single-RBC Hemoglobin polymer content Measurement of single-RBC
Hb polymer
Semi-quantitative measurement of hemoglobin polymer in single RBCs as function of oxygen DiCaprio et al. 2019. PNAS
Red cell biomechanics and rheology Sickling kinetics
Image-based quantification of SRBC deformity
RBC cortical tension
Splenic-like slit device
pH and oxygen associated biomechanics
sRBC behavior in cycles of hypoxia
Cortical tension in sRBC and pH increase stiffness of RBCs
Shape-dependent velocities are important.
Abbyad et al. 2010. Lab Chip
Guraprasad et al. 2019. Am J Hematol.
Li et al. 2017. PLoS Comput. Biol.
Gambhire, P et al. 2017. Small
Guo et al. 2014. J Biomech.
Du E. and Dao M. 2019. Exp Mech.
RBC adhesion Flow dependent RBC adhesion to laminin and fibronectin
RBC deformability
Sickle RBC adhesion to endothelial ligands or activated endothelium Alapan et al. 2014. Scientific Rep
Alapan et al. 2016. Transl Res
Kucukal et al. 2018. Am J Hem
OcclusionChip
Microfluidic electrical impedance
RBC-mediated microvascular occlusion Percent occlusions and percent impedance change across device correlates with HbS phenotype Man et al. 2021. Microcirculation
Man et al 2021. Lab Chip
Liu et al. 2018. Phys Fluids.
Pulsatile parallel plate flow adhesion Sickle RBC adhesion to VCAM-1, Laminin Sickle RBC adhesion to endothelial ligands or activated endothelium can be
blocked by antibodies
White J et al. 2015. Clin Hemorheol Microcirc

Clinically, the interplay between sRBC dehydration and permeability is important. For example, previous research has evaluated if systemic fluid resuscitation in individuals with SCD, particularly during times of VOE, can mitigate some of the effects of sRBC dehydration and reduce VOE. However, the most recent American Society of Hematology (ASH) Clinical Guidelines for SCD on managing acute or chronic pain does not offer recommendations regarding management of supplemental fluids to aid patients with SCD.39 This is in part due to the known pathologic permeability of sRBCs and the shift in cellular osmolality that it presents. By administering hyperosmolar fluids, there is a theoretical potential to increase cellular dehydration by increasing plasma osmolality and changing the efflux of potassium and calcium through transport channels (Fig 3B-C).4042 In fact, some suggest using hypo-osmolar solutions for hydration for the potential reverse effect.42 For example, in Carden et al. microvasculature-on-a-chip model, when comparing sRBCs exposed to hypo-osmolar solutions, sRBCs exposed to normal saline in vitro were stiffer, had longer transit times, and were more likely to occlude a microfluidic channel under normoxic and hypoxic conditions (Fig 3C).43 Interestingly, in a retrospective cohort study of SCD patients receiving intravenous fluid and pain medication, administration of normal saline was associated with poorer pain control.40 Collectively, these studies justify further work to identify how sRBC hydration, patient fluid resuscitation, and volume homeostasis affect HbS polymerization and VOE outcomes. Furthermore, this work highlights that incorporation of microfluidics into clinical studies investigating the effects of various osmolar fluids on sRBC rheology may provide better insight into our understanding of sRBC hydration on a cellular level and can help answer important clinical questions regarding how to best use fluids to manage patients.

Microfluidics to Model Sickle Red Cell Interactions with Endothelium

Decades of evidence demonstrates that sRBC adhesion to the endothelium contributes to disease pathophysiology.44,45 In addition to heme-mediated activation of endothelial cells46, the increased expression of surface molecules on the sRBC are known to contribute to endothelial adhesion. Increased expression of glycoprotein IV, integrin VLA-4, Landsteiner-Wiener intercellular adhesion molecule 4 (ICAM-4), and endothelial Lutheran/basal-cell adhesion molecule (Lu/BCAM) have all been studied and reported in the literature to play a role in increased sRBC adhesion.4651 Furthermore, as HbS polymerization increases sRBC density, early adhesion studies conducted under static conditions demonstrated that dense sRBCs exhibited higher amounts of endothelial adhesion when compared to light-density sRBCs.18,45 Therefore, sRBC interactions with the endothelium or endothelial activation ligands is an important consideration in modeling.

Recently, a series of microfluidic systems have served as a tool to recapitulate RBC-endothelial adhesion occurring in SCD (Table 1). Collectively, results from these studies demonstrate that increased sRBC adhesion molecule expression contributes to direct vaso-occlusion or increased capillary transit time, leading to longer exposure to deoxygenation and increased polymerization rates.14,5257 For example, in Papageorgiou et al.’s microfluidic device, which recapitulates the molecular and dimensional environment of post-capillary venules and is coated with the endothelial matrix protein fibronectin, hypoxia enhances sRBC adhesion four-fold. Furthermore, during HbS polymerization, the growing HbS fiber bundles also contribute to increased adhesion.57 Other groups have used microfluidic devices coated with endothelial matrix proteins laminin and VCAM, and demonstrated increased endothelial adhesion strength among the most non-deformable sRBCs as compared to more deformable sRBCs due to increased glycoprotein IV, integrin VLA-4, ICAM-4, and Lu/BCAM expression.5863 Likewise, White et al. has also modeled the microvasculature with a VCAM-1-coated device and has shown that pulsatile flow similar to that in vivo may lead to increased sRBC adhesion.64,65 Furthermore, Mathur et al. have also cultured SCD patient-derived blood outgrowth endothelial cells inside microfluidics to assess flow-mediated endothelial changes and sRBC adhesion.66,67 These model devices provide in-depth insight into the complex interactions occurring at the endothelium and how they might be contributing to vaso-occlusive events.

Red cell heterogeneity and implications for use of ektacytometry and microfluidics

sRBC Heterogeneity

While the various mechanical characteristics of sRBCs as a whole are important to SCD pathophysiology, RBC deformability and adhesion can vary across the sRBC lifespan.16 At any given time, RBCs of different ages co-exist and add to the complexity and heterogeneity of SCD pathophysiology. Given that RBCs must pass through the narrowest of vessels to deliver oxygen in the microcirculation, deformability plays a critical role in the survival of an RBC and SCD pathology.16,18,36,38 For example, as previously mentioned, HbS polymerization amplifies other circulation-dependent changes in RBC deformability and rigidity. Within the circulation, all RBCs undergo dynamic, transient deformations in order to migrate through various capillary diameters or the spleen.37,68,69 Over time, these deformations lead to alterations in membrane structure, generation of microvesicles (MV), and increased exposure of negatively-charged PS.70 Indeed, in computational studies of splenic function, RBC surface-to-volume ratio is a critical parameter of splenic filtration. 37,69 Additionally, using models of splenic slits, sRBCs exhibit cytoskeletal changes that result in a threefold increase in RBC cytoplasmic viscosity.37 Compared to healthy controls, return to equipoise after splenic transition is delayed. Collectively, these data suggest that even at steady state and under fully oxygenated conditions compared to non-sickle RBCs, sRBC undergo alterations in cell membrane plus cycles of polymerization throughout circulation that contribute to an overall decrease in deformability in RBCs from patients with SCD.71 The increase in sRBC rigidity can cause RBCs to be retained in the spleen, leading to extravascular hemolysis and contributing to chronic anemia in SCD.37,69 In early studies investigating cellular densities in sRBCs, 4 different subpopulations of cells based on density were identified: reticulocytes, discocytes, dense discocytes, and irreversibly sickled cells (ISCs). These subpopulations demonstrated corresponding increases in density with MCHC and increases in bulk viscosity measured in a viscosimeter.72 Furthermore, the extent to which these subpopulations exist within a patient varies and may contribute to an individuals’ clinical disease.36,72 The critical level at which each subpopulation causes clinical complications or severe disease is unknown. However, with the ability to measure adhesion and deformability, investigators may begin to understand the impact of each sRBC subpopulation and how its adhesive and deformable properties can drive SCD pathology, providing for targeted therapeutic development. Overall, the best technique to accurately measure deformability or density remains under debate; however, several techniques are being used, such as ektacytometry and single-cell measurements (Table 1)

Deformability by Ektacytometry

Ektacytometry is useful in determining RBC deformability, particularly while measuring deformability in red cell populations in flow. Because of this, ektacytometry is often used to study SCD and the effects on blood flow of sRBCs’ decreased deformability and increased rigidity.73 In one study using ektacytometry in SCD, a correlation of decreased deformability and increased rates of hemolysis was observed.74 Likewise, compared to HbSS individuals with high RBC deformability by ektacytometry, HbSS individuals with lower RBC deformability had significantly more leg ulcers and increased VOE events.75,76

Unfortunately, ektacytometry does not measure single-cell deformability and often provides unclear data on specific rigid, or dense RBC subpopulations that are often implicated in SCD pathology. This is, in part, because ektacytometry methodology is dependent on diffraction patterns that lack universal definitions.73 When looking at the two most common assays to measure deformability, ektacytometry and micro-pore filtration, neither technique can accurately predict the ability of RBCs to perfuse a microvascular network.77 One reason for this inaccuracy is that the distribution of polymer is bimodal, making bulk flow deformability measurements incomplete and inconclusive, as has been shown that under finite oxygen tensions.78 Therefore, as microfluidics models reveal the importance of sRBC subpopulations and cellular heterogeneity, assessing general deformability across all cells may not provide a specific nor accurate reflection of sRBC deformability and lead to inaccurate data interpretation. Hence, development of strategies to evaluate single-cell deformability and behavior may improve upon ektacytometry.

Deformability and Single-Cell Measurements by Microfluidics

More recently, with microfluidic devices, the ability to probe single-cell behavior provides better insight into the variable behaviors of individual sRBC subpopulations and their effect on blood flow. Man et al. have developed microfluidic models of progressively narrowing channels from 20 to 4 μm to recapitulate a capillary bed. To assess deformability, the percent of occlusions after a fixed time of blood flow may be measured using the occlusion index (OI). Lower OI has correlated with patients with more severe phenotypes, providing potential for a tool to assist in clinical risk stratification or predictive outcomes.79,80 Further studies by Li et al. also found correlation between deformability and sRBC flow.81 ISCs are not thought to contain polymerized hemoglobin, but are thought to be a product of membrane damage (i.e., MV and PS exposure) during repeated cycles of polymerization. ISCs are poorly deformable, have a short lifespan, and are highly correlated with hemolytic anemia.82 In a microfluidic chip designed with narrow channels as small as 5 μm to mimic the mechanical stress exerted on sRBCs in microcirculation, a significant reduction in ISCs were observed at the outlet of the device, suggesting the ISCs hemolyzed in transit through the device as a result of mechanical stress.83 This finding is not surprising given that ISCs are prone to hemolysis compared to other RBC populations.54 Within patients with identical genotypes of HbSS disease, it has been reported that ISCs comprise a wide, variable range between 5–50% of the total red cell population. The effects of a higher proportion of ISCs on whole blood rheology and its manifestations are highlighted in a study that correlated ISC proportion to spleen function, an organ that filters RBCs through endothelial slits 1–3 μm wide. In this study, pediatric patients that had higher percentages of ISCs experienced more episodes of acute splenic sequestration and splenic dysfunction.84

At present, defining and assessing the contribution of sRBC heterogeneity to SCD pathogenesis has been a major barrier to predicting success of many therapeutics, including gene therapy. As discussed earlier, induction of HbF has been a long-standing treatment for SCD. HbF is heterocellularly expressed only in a subset of sRBCs, F-cells, which vary in concentration in individuals ranging from 2 to 80% of sRBCs.85 However, the HbF concentration within each F-cell has a variable distribution. These individual differences in HbF expression and varied distribution among F-cells are likely controlled genetically and continue to be poorly understood.86 Although high levels of HbF are strongly associated with fewer vaso-occlusive complications, such as VOEs and acute chest syndrome,87 as polymerization inhibition requires a threshold intracellular HbF concentration, it is the distribution of HbF concentrations among F-cells that has a greater impact on disease.85 In recent work, Di Caprio et al. developed a single-RBC hemoglobin polymer microfluidic model designed to assess oxygen-dependent Hb polymers.78 This systems demonstrates that sRBC morphology and HbS polymer distribution is oxygen sensitive. Additionally, at a single-cell level this system identified patient-specific variable oxygen saturation distribution that is correlated to Hb polymer fraction.78 Therefore, moving beyond ektacytometry, microfluidics and single-RBC analysis may offer the ability to assess single RBC HbS polymers and HbF concentration as it pertains to the effects of hemoglobin modification.78

Application of Microfluidic Technologies to Assess New SCD-Directed Therapies

A persistent challenge in understanding the pathophysiology of SCD is the broad clinical phenotypic heterogeneity and lack of correlative biomarkers to stratify phenotypes. These factors significantly complicate the diagnosis and treatment process for patients. A recent hallmark study, the ELIPSIS study, monitored patients with SCD at home with an electronic patient-reported outcome tool and actigraphy over the course of 6 months. Additionally, a mobile-based blood collection team obtained blood samples when VOE was reported-permitting investigators to analyze trends over the course of a patient-reported VOE.65 During times of patient-reported VOE at home, whole blood adhesion to P-selectin was evaluated in a fluidic-based assay and found to be significantly elevated from individual patient’s baseline.65 These studies are notable as they provide insight into pathophysiology during VOE and substantiate previous clinical trial findings demonstrating that patients receiving crizanlizumab, a human monoclonal antibody against P-selectin, experienced a reduction in VOE.88

The ELIPIS study elegantly highlights that microfluidics assessment has the potential to identify and validate treatment strategies. As recently extensively reviewed by Telen et al., multiple strategies directed toward sRBC rheology and interactions continue to be explored.8 With expanding categories of interventions (anti-adhesion, anti-dehydration, oxygen-dissociation), when and how to proceed with treatment in a phenotypically heterogenous patient population will need to be identified. Going forward, combining microfluidic measurements, along with other tools such as computational modeling, may allow for non-invasive, upfront strategies to predict ability of candidate molecules to meaningfully alter patient-specific SCD parameters. An example of how this could be integrated is illustrated in Figure 4. First, patient-specific pre-intervention studies can be correlated to patient-reported outcomes (ie pain), and standard biometrics (blood work, vitals). If, with integration of these parameters, a subject is found to have increased adhesive properties on oxygen-dependent sRBC adhesion or occlusion models, one may select for anti-adhesive based therapies (ie crizanulizumab). However, if a subject has increased single-RBC Hb polymer content or oxygen-dependent sRBC velocity changes, one may instead chose oxygen-dissociation agents (ie voxeletor). After intervention, use of these technologies also will permit for monitoring of response, which along with patient-reported outcomes related to pain and clinical data may permit for increased standardization between trials. Finally, as multiple agents are in the developmental therapeutic pipeline-leveraging computational methods to quantify and predict patient-specific rheological patterns may inform studies combining therapies to maximize benefit and minimize toxicity. For example, if cohort of subject has rheologic patterns with increased single RBC polymer content and increased adhesion, combining anti-adhesive agent (crizanulizumab) with hemoglobin F induction (hydroxyurea) may be beneficial.89,90 As microfluidic models are non-invasive and can be standardized, incorporation of microfluidic measurements into the clinical development pipeline for SCD therapies is justified.91

Figure 4: Potential applications of microfluidic technologies in the development of SCD therapies.

Figure 4:

1. Samples can be collected before or after treatment. 2. Ex vivo studies from individuals with SCD can be used. These would include oxygen-dependent sRBC velocity, oxygen-dependent sRBC adhesion studies, and oxygen-dependent occlusion studies to evaluate RBC behavior under physiologic relevant conditions. Furthermore, single-RBC polymer content can be established. 3. These findings can be integrated with clinical parameters, including patient-based assessments, to assess response. Furthermore, combined with computational modeling, cycles of further interventions to optimize combination therapies may be completed. Image created in Biorender.com.

Strategies to Leverage Microfluidics Technologies in Sickle Cell Gene Therapy

Gene therapy to prevent HbS polymerization is the ultimate panacea in SCD treatment.92 At present, there are several ongoing clinical trials evaluating gene addition and gene editing strategies to cure SCD (Table 2). In general, gene addition approaches use lentiviral vectors to carry therapeutic genes into hematopoietic stem cells (HSCs). Gene editing using CRISPR-Cas is also underway to edit hemoglobin S translation.9395 Early clinical trials using gene additive strategies to introduce alternative γ-globin genes have shown clinical efficacy in patients with SCD and β-thalassemia.9,9499 Since then, several clinical trials have emerged to introduce anti-sickling globins (Table 2). Recently, two of the largest trials did report serious adverse outcomes prompting temporary cessation of enrollment.100,101 However, for those patients with SCD who have received lentiviral therapy, overall survival (OS) and event-free survival (EFS) are both 96% compared to historical allogeneic matched sibling HSCT OS and EFS of 95% and 92%, respectively. Importantly, after treatment, SCD subjects had no episodes of acute chest syndrome or VOE. Moreover, participants reported clinically meaningful improvements in pain reduction at 12 months post-treatment.100

Table 2:

Clinical Trials for Gene therapy and editing in sickle cell disease

Strategy NCT Number Phase/Enrollment/Status Results Citations
Lentiviral transfer of γ-globinG16D – ARU-1801 NCT02186418 Phase 1/2
Enrollment- 10
Status: open
Increased F-cells, no VOE Grimley M, et al. 2020. Blood
Lentiviral transfer of modified HBB encoding antisickling variant βA87Thr:Gln[βA-T87Q] LentiGlobin BB305 NCT02151526 Phase 1/2
Enrollment- 7
Status: closed
Stable CBC,
Decreased reticulocyte, decreased RBC sickling under hypoxia and improved RBC deformability
Ribel JA et al. 2017. NEJM
NCT02140554 Phase 1/2
Enrollment- 50
Status: not recruiting*
NCT04293185 Phase 3
Enrollment- 35
Status: recruiting*
AS3-FB vector transduced peripheral blood CD34+ cells NCT02247843 Phase 1/2
Enrollment- 6
Status: not recruiting
None reported Pre-clinical: decreased sickling, engraftment of mice Romero Z. et al. 2013. JCI
Lentiviral targeting of γ-globin repressor BCL11A with BCH-BB694 BCL11A shmiR vector NCT03282656 Phase 1
Enrollment- 15
Status: SUSPENDED*
Increased HbF, increased % F-cells, increased HbF per F cells Esrick, EB. Et al. 2021. NEJM
Reactivation of fetal hemoglobin by using Crisper-Cas (CTX001) editing to suppress BCL11A β-globin switch NCT03745287 Phase 1/2
Enrollment- 45
Status: recruiting
Increased HbF, reduced transfusion needs and no VOE Frangoul, H. et al. 2021. NEJM
*

Studies were paused in March 2021 due to the development of myelodysplastic syndrome and acute myeloid leukemia in patients in NCT02140554 and NCT04293185 prompting suspension of NCT03282656 out of caution. NCT02140554 and NCT04293185 currently active.

Overall, for gene therapy and other potentially disease-modifying therapies, use of microfluidic strategies, such as oxygen-dependent velocity, endothelial interaction, and single-RBC Hb polymerization assays will be valuable in identifying how HbF concentration distribution among F-cells correlates to SCD clinical phenotype. Thus far, the largest trial, by Rivel et al. used ektacytometry and morphologic assessment to demonstrate sRBC benefits of lentiglobin modification.97 More recent work by Frangoul et al. and Esrick et al. demonstrated increased F-cell number, with further studies evaluating sRBC biophysical characteristics anticipated as more subjects enroll in the trials.95,98 Collectively, these studies highlight the importance of incorporating measurements of sRBC biophysical behavior to obtain a robust characterization of the myriad of effects driven by the sRBC.

Conclusions

In conclusion, microfluidics models have advanced our fundamental understanding of how sRBCs drive SCD pathogenesis. Thus far, the importance of HbS polymerization has driven much of the evaluation, with ektacytometry studies increasingly being incorporated into clinical trials. Moving forward, adoption of single-RBC polymerization models may permit for further insights into oxygen-sensitive RBC characteristics. Additionally, incorporation of patient samples in microvascular occlusion models offers the ability to rapidly screen candidate therapeutics for further pre-clinical (i.e., mouse) studies. During clinical studies, use of microfluidic assays allows for non-invasive measurement of candidate interventions on hemorheology. For far too long, the SCD therapeutic pipeline has been stagnant. However, with multiple therapeutics and gene therapy under rapid develop, the addition of microfluidic technologies offers the exciting opportunity to accelerate the investigational pipeline for sickle cell therapies.

Figure 1: Sickle hemoglobin fiber polymerization is related to hemoglobin S concentration and is highly inefficient.

Figure 1:

A. Hemoglobin S polymerization (C) as a function of HbS molecules per cell as calculated by Lu, L et al. 2017. Figure moodified and used with permission. B. Differential interference contrast images showing an example of heterogeneous nucleation (left) and the intersection of two separate HbS fibers (right). Red and yellow arrowheads indicate the region where the intensity value was calculated for the multifiber region (nucleation or intersection) or the adjacent region, respectively. Scale bars, 2 μm. (Figure modified and used with permission from Castle et al. 2019).

Funding

Portions of this work were conducted in the Minnesota Nano Center, which is supported by the National Science Foundation through the National Nanotechnology Coordinated Infrastructure (NNCI) under Award Number ECCS-2025124. E.F.V. is supported by F31HL158223. W.A.L. is supported by R35HL145000. W.A.L. and D.K.W. are supported by R01HL140589. D.K.W is supported by HL132906. J.D.B. is supported in part by Institutional Research Grant #129819-IRG-16–189-58-IRG-114 from the American Cancer Society, OT2 HL15275801, an American Society of Hematology Restart Award and American Heart Association Career Development Award.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosures

Dr. Beckman receives funds from Bayer unrelated to content of review. The remaining authors have no conflicts of interest.

References

  • 1.Connes P, Alexy T, Detterich J, Romana M, Hardy-Dessources MD, Ballas SK. The role of blood rheology in sickle cell disease. Blood Rev 2016;30(2):111–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Rees DC, Gibson JS. Biomarkers in sickle cell disease. Br J Haematol 2012;156(4):433–445. [DOI] [PubMed] [Google Scholar]
  • 3.Lubeck D, Agodoa I, Bhakta N, et al. Estimated Life Expectancy and Income of Patients With Sickle Cell Disease Compared With Those Without Sickle Cell Disease. JAMA Netw Open 2019;2(11):e1915374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kauf TL, Coates TD, Huazhi L, Mody-Patel N, Hartzema AG. The cost of health care for children and adults with sickle cell disease. Am J Hematol 2009;84(6):323–327. [DOI] [PubMed] [Google Scholar]
  • 5.National Center on Birth Defects and Developmental Disabilities CfDCaP Center For Disease Control. Vol. 2020. https://www.cdc.gov/ncbddd/sicklecell/data.html:CDC.gov; 2019. [Google Scholar]
  • 6.Esham KS, Rodday AM, Smith HP, et al. Assessment of health-related quality of life among adults hospitalized with sickle cell disease vaso-occlusive crisis. Blood Adv 2020;4(1):19–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kanter J, Smith WR, Desai PC, et al. Building access to care in adult sickle cell disease: defining models of care, essential components, and economic aspects. Blood Adv 2020;4(16):3804–3813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Telen MJ, Malik P, Vercellotti GM. Therapeutic strategies for sickle cell disease: towards a multi-agent approach. Nat Rev Drug Discov 2018. [DOI] [PMC free article] [PubMed]
  • 9.Kanter J, Walters MC, Krishnamurti L, et al. Biologic and Clinical Efficacy of LentiGlobin for Sickle Cell Disease. New England Journal of Medicine 2021. [DOI] [PubMed]
  • 10.Kalambur VS, Mahaseth H, Bischof JC, et al. Microvascular blood flow and stasis in transgenic sickle mice: Utility of a dorsal skin fold chamber for igntravital microscopy. Am J Hematol 2004;77(2):117–125. [DOI] [PubMed] [Google Scholar]
  • 11.Higgins JM, Eddington DT, Bhatia SN, Mahadevan L. Sickle cell vasoocclusion and rescue in a microfluidic device. Proc Natl Acad Sci U S A 2007;104(51):20496–20500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lu X, Wood DK, Higgins JM. Deoxygenation Reduces Sickle Cell Blood Flow at Arterial Oxygen Tension. Biophys J 2016;110(12):2751–2758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wood DK, Soriano A, Mahadevan L, Higgins JM, Bhatia SN. A biophysical indicator of vaso-occlusive risk in sickle cell disease. Sci Transl Med 2012;4(123):123ra126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Valdez JM, Datta YH, Higgins JM, Wood DK. A microfluidic platform for simultaneous quantification of oxygen-dependent viscosity and shear thinning in sickle cell blood. APL Bioeng 2019;3(4):046102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lu X, Chaudhury A, Higgins JM, Wood DK. Oxygen-dependent flow of sickle trait blood as an in vitro therapeutic benchmark for sickle cell disease treatments. American Journal of Hematology 2018;93(10):1227–1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Qiang Y, Liu J, Dao M, Du E. In vitro assay for single-cell characterization of impaired deformability in red blood cells under recurrent episodes of hypoxia. Lab Chip 2021;21(18):3458–3470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Du E, Dao M. Faster Sickling Kinetics and Sickle Cell Shape Evolution during Repeated Deoxygenation and Oxygenation Cycles. Exp Mech 2019;59(3):319–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Atwell S, Badens C, Charrier A, Helfer E, Viallat A. Dynamics of Individual Red Blood Cells Under Shear Flow: A Way to Discriminate Deformability Alterations. Front Physiol 2021;12:775584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Du E, Diez-Silva M, Kato GJ, Dao M, Suresh S. Kinetics of sickle cell biorheology and implications for painful vasoocclusive crisis. Proceedings of the National Academy of Sciences 2015;112(5):1422–1427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Abbyad P, Tharaux P-L, Martin J-L, Baroud CN, Alexandrou A. Sickling of red blood cells through rapid oxygen exchange in microfluidic drops. Lab on a Chip 2010;10(19):2505–2512. [DOI] [PubMed] [Google Scholar]
  • 21.Lu X, Galarneau MM, Higgins JM, Wood DK. A microfluidic platform to study the effects of vascular architecture and oxygen gradients on sickle blood flow. Microcirculation 2017;24(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Usmani A, Machado RF. Vascular complications of sickle cell disease. Clin Hemorheol Microcirc 2018;68(2–3):205–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Song H, Keegan PM, Anbazhakan S, et al. Sickle Cell Anemia Mediates Carotid Artery Expansive Remodeling That Can Be Prevented by Inhibition of JNK (c-Jun N-Terminal Kinase). Arterioscler Thromb Vasc Biol 2020;40(5):1220–1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Noguchi CT, Rodgers GP, Schechter AN. Intracellular polymerization. Disease severity and therapeutic predictions. Ann N Y Acad Sci 1989;565:75–82. [DOI] [PubMed] [Google Scholar]
  • 25.Rodgers GP, Dover GJ, Noguchi CT, Schechter AN, Nienhuis AW. Induction of fetal hemoglobin in sickle cell patients by hydroxyurea: the N.I.H. experience. Prog Clin Biol Res 1989;316b:281–293. [PubMed] [Google Scholar]
  • 26.Rodgers GP, Dover GJ, Noguchi CT, Schechter AN, Nienhuis AW. Hematologic responses of patients with sickle cell disease to treatment with hydroxyurea. N Engl J Med 1990;322(15):1037–1045. [DOI] [PubMed] [Google Scholar]
  • 27.Poillon WN, Kim BC, Rodgers GP, Noguchi CT, Schechter AN. Sparing effect of hemoglobin F and hemoglobin A2 on the polymerization of hemoglobin S at physiologic ligand saturations. Proceedings of the National Academy of Sciences of the United States of America 1993;90(11):5039–5043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Henry ER, Cellmer T, Dunkelberger EB, et al. Allosteric control of hemoglobin S fiber formation by oxygen and its relation to the pathophysiology of sickle cell disease. Proceedings of the National Academy of Sciences 2020;117(26):15018–15027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Fibach E, Burke LP, Schechter AN, Noguchi CT, Rodgers GP. Hydroxyurea increases fetal hemoglobin in cultured erythroid cells derived from normal individuals and patients with sickle cell anemia or beta-thalassemia. Blood 1993;81(6):1630–1635. [PubMed] [Google Scholar]
  • 30.Charache S, Barton FB, Moore RD, et al. Hydroxyurea and sickle cell anemia. Clinical utility of a myelosuppressive “switching” agent. The Multicenter Study of Hydroxyurea in Sickle Cell Anemia. Medicine (Baltimore) 1996;75(6):300–326. [DOI] [PubMed] [Google Scholar]
  • 31.Vichinsky E, Hoppe CC, Ataga KI, et al. A Phase 3 Randomized Trial of Voxelotor in Sickle Cell Disease. N Engl J Med 2019;381(6):509–519. [DOI] [PubMed] [Google Scholar]
  • 32.Huisjes R, Bogdanova A, van Solinge WW, Schiffelers RM, Kaestner L, van Wijk R. Squeezing for Life – Properties of Red Blood Cell Deformability. Frontiers in Physiology 2018;9(656). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lew VL, Bookchin RM. Ion transport pathology in the mechanism of sickle cell dehydration. Physiol Rev 2005;85(1):179–200. [DOI] [PubMed] [Google Scholar]
  • 34.Lew VL, Freeman CJ, Ortiz OE, Bookchin RM. A mathematical model of the volume, pH, and ion content regulation in reticulocytes. Application to the pathophysiology of sickle cell dehydration. J Clin Invest 1991;87(1):100–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Liu J, Qiang Y, Alvarez O, Du E. Electrical Impedance Characterization of Erythrocyte Response to Cyclic Hypoxia in Sickle Cell Disease. ACS Sensors 2019;4(7):1783–1790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Passos A, Sherwood JM, Kaliviotis E, Agrawal R, Pavesio C, Balabani S. The effect of deformability on the microscale flow behavior of red blood cell suspensions. Physics of Fluids 2019;31(9):091903. [Google Scholar]
  • 37.Gambhire P, Atwell S, Iss C, et al. High Aspect Ratio Sub-Micrometer Channels Using Wet Etching: Application to the Dynamics of Red Blood Cell Transiting through Biomimetic Splenic Slits. Small 2017;13(32). [DOI] [PubMed] [Google Scholar]
  • 38.Guo Q, Duffy SP, Matthews K, Santoso AT, Scott MD, Ma H. Microfluidic analysis of red blood cell deformability. Journal of Biomechanics 2014;47(8):1767–1776. [DOI] [PubMed] [Google Scholar]
  • 39.Brandow AM, Carroll CP, Creary S, et al. American Society of Hematology 2020 guidelines for sickle cell disease: management of acute and chronic pain. Blood Advances 2020;4(12):2656–2701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Carden MA, Brousseau DC, Ahmad FA, et al. Normal saline bolus use in pediatric emergency departments is associated with poorer pain control in children with sickle cell anemia and vaso-occlusive pain. Am J Hematol 2019;94(6):689–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Carden MA, Fay ME, Lu X, et al. Extracellular fluid tonicity impacts sickle red blood cell deformability and adhesion. Blood 2017;130(24):2654–2663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Benjamin LJ, Swinson GI, Nagel RL. Sickle cell anemia day hospital: an approach for the management of uncomplicated painful crises. Blood 2000;95(4):1130–1136. [PubMed] [Google Scholar]
  • 43.Carden MA, Fay M, Sakurai Y, et al. Normal saline is associated with increased sickle red cell stiffness and prolonged transit times in a microfluidic model of the capillary system. Microcirculation 2017;24(5). [DOI] [PubMed] [Google Scholar]
  • 44.Hebbel RP, Boogaerts MA, Eaton JW, Steinberg MH. Erythrocyte adherence to endothelium in sickle-cell anemia. A possible determinant of disease severity. N Engl J Med 1980;302(18):992–995. [DOI] [PubMed] [Google Scholar]
  • 45.Hebbel RP, Yamada O, Moldow CF, Jacob HS, White JG, Eaton JW. Abnormal adherence of sickle erythrocytes to cultured vascular endothelium: possible mechanism for microvascular occlusion in sickle cell disease. J Clin Invest 1980;65(1):154–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Murphy MM, Zayed MA, Evans A, et al. Role of Rap1 in promoting sickle red blood cell adhesion to laminin via BCAM/LU. Blood 2005;105(8):3322–3329. [DOI] [PubMed] [Google Scholar]
  • 47.Hines PC, Zen Q, Burney SN, et al. Novel epinephrine and cyclic AMP-mediated activation of BCAM/Lu-dependent sickle (SS) RBC adhesion. Blood 2003;101(8):3281–3287. [DOI] [PubMed] [Google Scholar]
  • 48.Udani M, Zen Q, Cottman M, et al. Basal cell adhesion molecule/lutheran protein. The receptor critical for sickle cell adhesion to laminin. J Clin Invest 1998;101(11):2550–2558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lee SP, Cunningham ML, Hines PC, Joneckis CC, Orringer EP, Parise LV. Sickle cell adhesion to laminin: potential role for the alpha5 chain. Blood 1998;92(8):2951–2958. [PubMed] [Google Scholar]
  • 50.Hebbel RP, Moldow CF, Steinberg MH. Modulation of erythrocyte-endothelial interactions and the vasocclusive severity of sickling disorders. Blood 1981;58(5):947–952. [PubMed] [Google Scholar]
  • 51.White J, Krishnamoorthy S, Gupta D, et al. VLA-4 blockade by natalizumab inhibits sickle reticulocyte and leucocyte adhesion during simulated blood flow. Br J Haematol 2016;174(6):970–982. [DOI] [PubMed] [Google Scholar]
  • 52.Lu X, Chaudhury A, Higgins JM, Wood DK. Oxygen-dependent flow of sickle trait blood as an in vitro therapeutic benchmark for sickle cell disease treatments. Am J Hematol 2018. [DOI] [PMC free article] [PubMed]
  • 53.Lu M, Kanne CK, Reddington RC, Lezzar DL, Sheehan VA, Shevkoplyas SS. Concurrent Assessment of Deformability and Adhesiveness of Sickle Red Blood Cells by Measuring Perfusion of an Adhesive Artificial Microvascular Network. Front Physiol 2021;12:633080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Lizarralde Iragorri MA, El Hoss S, Brousse V, et al. A microfluidic approach to study the effect of mechanical stress on erythrocytes in sickle cell disease. Lab Chip 2018;18(19):2975–2984. [DOI] [PubMed] [Google Scholar]
  • 55.Kucukal E, Man Y, Quinn E, et al. Red blood cell adhesion to ICAM-1 is mediated by fibrinogen and is associated with right-to-left shunts in sickle cell disease. Blood Adv 2020;4(15):3688–3698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kucukal E, Ilich A, Key NS, Little JA, Gurkan UA. Red Blood Cell Adhesion to Heme-Activated Endothelial Cells Reflects Clinical Phenotype in Sickle Cell Disease. Am J Hematol 2018. [DOI] [PMC free article] [PubMed]
  • 57.Papageorgiou DP, Abidi SZ, Chang HY, et al. Simultaneous polymerization and adhesion under hypoxia in sickle cell disease. Proc Natl Acad Sci U S A 2018;115(38):9473–9478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lizarralde-Iragorri MA, Lefevre SD, Cochet S, et al. Oxidative stress activates red cell adhesion to laminin in sickle cell disease. Haematologica 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.White JC, Pawar A, Fu G, et al. TR2/TR4 overexpression in a humanized sickle cell disease mouse model decreases RBC adhesion to VCAM-1. Blood Cells Mol Dis 2015;55(4):316–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lancelot M, White J, Sarnaik S, Hines P. Low molecular weight heparin inhibits sickle erythrocyte adhesion to VCAM-1 through VLA-4 blockade in a standardized microfluidic flow adhesion assay. Br J Haematol 2017;178(3):479–481. [DOI] [PubMed] [Google Scholar]
  • 61.Zhang J, Abiraman K, Jones SM, Lykotrafitis G, Andemariam B. Regulation of Active ICAM-4 on Normal and Sickle Cell Disease RBCs via AKAPs Is Revealed by AFM. Biophys J 2017;112(1):143–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Alapan Y, Little JA, Gurkan UA. Heterogeneous red blood cell adhesion and deformability in sickle cell disease. Sci Rep 2014;4:7173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Alapan Y, Kim C, Adhikari A, et al. Sickle cell disease biochip: a functional red blood cell adhesion assay for monitoring sickle cell disease. Transl Res 2016;173:74–91.e78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.White J, Lancelot M, Sarnaik S, Hines P. Increased erythrocyte adhesion to VCAM-1 during pulsatile flow: Application of a microfluidic flow adhesion bioassay. Clin Hemorheol Microcirc 2015;60(2):201–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Pittman DD, Hines PC, Beidler D, et al. Evaluation of Longitudinal Pain Study in Sickle Cell Disease (ELIPSIS) by patient-reported outcomes, actigraphy, and biomarkers. Blood 2021;137(15):2010–2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mathur T, Flanagan JM, Jain A. Tripartite collaboration of blood-derived endothelial cells, next generation RNA sequencing and bioengineered vessel-chip may distinguish vasculopathy and thrombosis among sickle cell disease patients. Bioeng Transl Med 2021;6(3):e10211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Mathur T, Singh KA, NK RP, et al. Organ-on-chips made of blood: endothelial progenitor cells from blood reconstitute vascular thromboinflammation in vessel-chips. Lab Chip 2019;19(15):2500–2511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Watts T, Barigou M, Nash GB. Comparative rheology of the adhesion of platelets and leukocytes from flowing blood: why are platelets so small? American Journal of Physiology - Heart and Circulatory Physiology 2013;304(11):H1483–H1494. [DOI] [PubMed] [Google Scholar]
  • 69.Pivkin IV, Peng Z, Karniadakis GE, Buffet PA, Dao M, Suresh S. Biomechanics of red blood cells in human spleen and consequences for physiology and disease. Proc Natl Acad Sci U S A 2016;113(28):7804–7809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Burger P, Kostova E, Bloem E, et al. Potassium leakage primes stored erythrocytes for phosphatidylserine exposure and shedding of pro-coagulant vesicles. British Journal of Haematology 2013;160(3):377–386. [DOI] [PubMed] [Google Scholar]
  • 71.Rab MAE, van Oirschot BA, Bos J, et al. Rapid and reproducible characterization of sickling during automated deoxygenation in sickle cell disease patients. Am J Hematol 2019;94(5):575–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kaul DK, Fabry ME, Windisch P, Baez S, Nagel RL. Erythrocytes in sickle cell anemia are heterogeneous in their rheological and hemodynamic characteristics. J Clin Invest 1983;72(1):22–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Parrow NL, Violet PC, Tu H, et al. Measuring Deformability and Red Cell Heterogeneity in Blood by Ektacytometry. J Vis Exp 2018(131). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Connes P, Lamarre Y, Waltz X, et al. Haemolysis and abnormal haemorheology in sickle cell anaemia. Br J Haematol 2014;165(4):564–572. [DOI] [PubMed] [Google Scholar]
  • 75.Ballas SK. Sickle cell anemia with few painful crises is characterized by decreased red cell deformability and increased number of dense cells. Am J Hematol 1991;36(2):122–130. [DOI] [PubMed] [Google Scholar]
  • 76.Rab MAE, van Oirschot BA, Bos J, et al. Characterization of Sickling During Controlled Automated Deoxygenation with Oxygen Gradient Ektacytometry. J Vis Exp 2019(153). [DOI] [PubMed] [Google Scholar]
  • 77.Sosa JM, Nielsen ND, Vignes SM, Chen TG, Shevkoplyas SS. The relationship between red blood cell deformability metrics and perfusion of an artificial microvascular network. Clin Hemorheol Microcirc 2014;57(3):275–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Di Caprio G, Schonbrun E, Gonçalves BP, Valdez JM, Wood DK, Higgins JM. High-throughput assessment of hemoglobin polymer in single red blood cells from sickle cell patients under controlled oxygen tension. Proc Natl Acad Sci U S A 2019;116(50):25236–25242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Man Y, Kucukal E, An R, Bode A, Little JA, Gurkan UA. Standardized microfluidic assessment of red blood cell-mediated microcapillary occlusion: Association with clinical phenotype and hydroxyurea responsiveness in sickle cell disease. Microcirculation 2021;28(2):e12662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Man Y, Maji D, An R, et al. Microfluidic electrical impedance assessment of red blood cell-mediated microvascular occlusion. Lab Chip 2021;21(6):1036–1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Li X, Du E, Dao M, Suresh S, Karniadakis GE. Patient-specific modeling of individual sickle cell behavior under transient hypoxia. PLoS Comput Biol 2017;13(3):e1005426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Rodgers GP, Noguchi CT, Schechter AN. Irreversibly sickled erythrocytes in sickle cell anemia: a quantitative reappraisal. Am J Hematol 1985;20(1):17–23. [DOI] [PubMed] [Google Scholar]
  • 83.Guruprasad P, Mannino RG, Caruso C, et al. Integrated automated particle tracking microfluidic enables high-throughput cell deformability cytometry for red cell disorders. American Journal of Hematology 2019;94(2):189–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.El Hoss S, Cochet S, Marin M, et al. Insights into determinants of spleen injury in sickle cell anemia. Blood Adv 2019;3(15):2328–2336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Steinberg MH, Kumar S, Murphy GJ, Vanuytsel K. Sickle cell disease in the era of precision medicine: looking to the future. Expert Rev Precis Med Drug Dev 2019;4(6):357–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Khandros E, Huang P, Peslak SA, et al. Understanding heterogeneity of fetal hemoglobin induction through comparative analysis of F and A erythroblasts. Blood 2020;135(22):1957–1968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Charache S, Terrin ML, Moore RD, et al. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia. N Engl J Med 1995;332(20):1317–1322. [DOI] [PubMed] [Google Scholar]
  • 88.Ataga KI, Kutlar A, Kanter J, et al. Crizanlizumab for the Prevention of Pain Crises in Sickle Cell Disease. N Engl J Med 2017;376(5):429–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Bazzi MS, Valdez JM, Barocas VH, Wood DK. An Experimental-Computational Approach to Quantify Blood Rheology in Sickle Cell Disease. Biophys J 2020;119(11):2307–2315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Karimi A, Hassani K, Golpaygani AT, Izadi F. A Fluid Structure Interaction Model for the Red Blood Cell Mechanics. Materials Focus 2016;5(6):524–528. [Google Scholar]
  • 91.Lu L, Li Z, Li H, Li X, Vekilov PG, Karniadakis GE. Quantitative prediction of erythrocyte sickling for the development of advanced sickle cell therapies. Sci Adv 2019;5(8):eaax3905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Eaton WA, Bunn HF. Treating sickle cell disease by targeting HbS polymerization. Blood 2017;129(20):2719–2726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Uchida N, Drysdale CM, Nassehi T, et al. Cas9 protein delivery non-integrating lentiviral vectors for gene correction in sickle cell disease. Mol Ther Methods Clin Dev 2021;21:121–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Lattanzi A, Camarena J, Lahiri P, et al. Development of β-globin gene correction in human hematopoietic stem cells as a potential durable treatment for sickle cell disease. Sci Transl Med 2021;13(598). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Frangoul H, Altshuler D, Cappellini MD, et al. CRISPR-Cas9 Gene Editing for Sickle Cell Disease and β-Thalassemia. N Engl J Med 2021;384(3):252–260. [DOI] [PubMed] [Google Scholar]
  • 96.Cavazzana-Calvo M, Payen E, Negre O, et al. Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia. Nature 2010;467(7313):318–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ribeil JA, Hacein-Bey-Abina S, Payen E, et al. Gene Therapy in a Patient with Sickle Cell Disease. N Engl J Med 2017;376(9):848–855. [DOI] [PubMed] [Google Scholar]
  • 98.Esrick EB, Lehmann LE, Biffi A, et al. Post-Transcriptional Genetic Silencing of BCL11A to Treat Sickle Cell Disease. N Engl J Med 2021;384(3):205–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Romero Z, Urbinati F, Geiger S, et al. β-globin gene transfer to human bone marrow for sickle cell disease. J Clin Invest 2013;123(8):3317–3330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Leonard A, Tisdale JF. A pause in gene therapy: Reflecting on the unique challenges of sickle cell disease. Mol Ther 2021;29(4):1355–1356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Goyal S, Tisdale J, Schmidt M, et al. Acute Myeloid Leukemia Case after Gene Therapy for Sickle Cell Disease. New England Journal of Medicine 2021;386(2):138–147. [DOI] [PubMed] [Google Scholar]

RESOURCES