scholarly journals Metabolomic and molecular insights into sickle cell disease and innovative therapies

2019 ◽  
Vol 3 (8) ◽  
pp. 1347-1355 ◽  
Author(s):  
Morayo G. Adebiyi ◽  
Jeanne M. Manalo ◽  
Yang Xia

Abstract Sickle cell disease (SCD) is an autosomal-recessive hemolytic disorder with high morbidity and mortality. The pathophysiology of SCD is characterized by the polymerization of deoxygenated intracellular sickle hemoglobin, which causes the sickling of erythrocytes. The recent development of metabolomics, the newest member of the “omics” family, has provided a powerful new research strategy to accurately measure functional phenotypes that are the net result of genomic, transcriptomic, and proteomic changes. Metabolomics changes respond faster to external stimuli than any other “ome” and are especially appropriate for surveilling the metabolic profile of erythrocytes. In this review, we summarize recent pioneering research that exploited cutting-edge metabolomics and state-of-the-art isotopically labeled nutrient flux analysis to monitor and trace intracellular metabolism in SCD mice and humans. Genetic, structural, biochemical, and molecular studies in mice and humans demonstrate unrecognized intracellular signaling pathways, including purinergic and sphingolipid signaling networks that promote hypoxic metabolic reprogramming by channeling glucose metabolism to glycolysis via the pentose phosphate pathway. In turn, this hypoxic metabolic reprogramming induces 2,3-bisphosphoglycerate production, deoxygenation of sickle hemoglobin, polymerization, and sickling. Additionally, we review the detrimental role of an impaired Lands’ cycle, which contributes to sickling, inflammation, and disease progression. Thus, metabolomic profiling allows us to identify the pathological role of adenosine signaling and S1P-mediated erythrocyte hypoxic metabolic reprogramming and hypoxia-induced impaired Lands' cycle in SCD. These findings further reveal that the inhibition of adenosine and S1P signaling cascade and the restoration of an imbalanced Lands' cycle have potent preclinical efficacy in counteracting sickling, inflammation, and disease progression.

Genes ◽  
2022 ◽  
Vol 13 (1) ◽  
pp. 144
Author(s):  
Olivia Edwards ◽  
Alicia Burris ◽  
Josh Lua ◽  
Diana J. Wilkie ◽  
Miriam O. Ezenwa ◽  
...  

This review outlines the current clinical research investigating how the haptoglobin (Hp) genetic polymorphism and stroke occurrence are implicated in sickle cell disease (SCD) pathophysiology. Hp is a blood serum glycoprotein responsible for binding and removing toxic free hemoglobin from the vasculature. The role of Hp in patients with SCD is critical in combating blood toxicity, inflammation, oxidative stress, and even stroke. Ischemic stroke occurs when a blocked vessel decreases oxygen delivery in the blood to cerebral tissue and is commonly associated with SCD. Due to the malformed red blood cells of sickle hemoglobin S, blockage of blood flow is much more prevalent in patients with SCD. This review is the first to evaluate the role of the Hp polymorphism in the incidence of stroke in patients with SCD. Overall, the data compiled in this review suggest that further studies should be conducted to reveal and evaluate potential clinical advancements for gene therapy and Hp infusions.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. LBA-3-LBA-3
Author(s):  
Yujin Zhang ◽  
Vladimir Berka ◽  
Wei Wang ◽  
Weiru Zhang ◽  
Chen Ning ◽  
...  

Abstract LBA-3 Sickle cell disease (SCD) is a debilitating hemolytic disorder with high morbidity and mortality affecting millions of individuals worldwide. Although SCD was first identified a century ago, we still lack effective mechanism-based safe therapies to treat this disease. Thus, identification of specific molecules triggering sickling, the central pathogenic process of the disease, is extremely important to advance our understanding of the molecular basis for the pathogenesis of SCD and to develop novel therapeutics. Using non-biased metabolomic screening, we found that sphingosine-1-phosphate (S1P) is significantly elevated in the blood of SCD mice. Further analysis revealed that the activity of sphingosine kinase 1 (Sphk1, the enzyme that produces S1P) is significantly elevated in erythrocytes of SCD mice. Chronic treatment of SCD mice with a SphK1 inhibitor significantly attenuated sickling, hemolysis, inflammation and multiple tissue damage by reducing erythrocyte and plasma S1P levels. Erythrocyte S1P levels were further elevated following hypoxia/reoxygenation-induced acute sickle crisis (ASC) in SCD mice and blocking its elevation by a Sphk1 specific inhibitor significantly reduced hallmark features associated with ASC. As with SCD mice, we found that erythrocyte Sphk1 activity and erythrocyte and plasma S1P levels were significantly elevated in humans with SCD compared to normal individuals. Inhibition of SphK1 in cultured primary human erythrocytes isolated from SCD patients inhibited hypoxia-induced elevation of erythrocyte S1P levels and reduced sickling. Thus, we have revealed for the first time that SphK1-mediated S1P elevation in SCD erythrocytes is a key contributor to sickling in SCD and that Sphk1 inhibition can attenuate both acute and chronic sickling events and disease progression. S1P is an important signaling molecule regulating diverse biological processes. Although S1P is predominantly produced and stored in RBCs, nothing was known about the physiological role of S1P in normal RBCs or the pathophysiological role of S1P in SCD until we conducted a metabolomic screen. In an effort to determine the molecular mechanism underlying S1P-induced sickling, we unexpectedly found that S1P directly binds with Hb and results in a reduced Hb-O2 affinity. This finding led us to further discover that 2,3-diphosphoglycerate, another erythrocyte specific allosteric modulator, is required for S1P-mediated allosteric modulation and that these two endogenous heterotropic modulators work cooperatively to induce a substantial reduction in Hb-O2 affinity. Supporting the biochemical and functional findings, molecular modeling predicts that S1P binds near the water filled central cavity of HbA at a site that is different from the Hb-2,3-DPG binding site. Thus, our discovery adds a significant new chapter to erythrocyte physiology by revealing S1P is a novel allosteric modulator of Hb-O2 affinity and also providing a mechanism underlying S1P-mediated sickling by promoting the formation of deoxyHbS. Thus, the work reported here could be the foundation leading to future human trials and a possible therapy for SCD, a life-threatening hemolytic disorder for which the current treatment is extremely limited. The significance of our findings extends well beyond SCD. Our findings reveal a previously unrecognized important role for S1P in erythrocyte physiology and indicate a new concept for the regulation of O2 release from Hb under normal and sickle cell disease conditions. For SCD, elevated S1P is detrimental because reduced Hb-O2 affinity leads to more deoxygenation of HbS, increased sickling and subsequent multiple life-threatening complications. However, for normal erythrocytes, elevated S1P is likely beneficial by decreasing Hb-O2 affinity allowing for more O2 release to hypoxic tissues. Thus, for humans with normal Hb, if elevated S1P can induce O2 release to hypoxic tissues it may be a novel therapeutic target for a range of disorders, from chronic heart failure to diabetic retinopathy, traumatic blood loss, pulmonary disease and even cancer. In this way our findings reveal important novel opportunities to treat and prevent not only SCD but also multiple cardiovascular and pulmonary diseases associated with hypoxia. Thus, the impact of our novel finding is significant and enormous. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. LBA-3-LBA-3
Author(s):  
Yujin Zhang ◽  
Vladimir Berka ◽  
Wei Wang ◽  
Weiru Zhang ◽  
Chen Ning ◽  
...  

Abstract Abstract LBA-3 Sickle cell disease (SCD) is a debilitating hemolytic disorder with high morbidity and mortality affecting millions of individuals worldwide. Although SCD was first identified a century ago, we still lack effective mechanism-based safe therapies to treat this disease. Thus, identification of specific molecules triggering sickling, the central pathogenic process of the disease, is extremely important to advance our understanding of the molecular basis for the pathogenesis of SCD and to develop novel therapeutics. Using non-biased metabolomic screening, we found that sphingosine-1-phosphate (S1P) is significantly elevated in the blood of SCD mice. Further analysis revealed that the activity of sphingosine kinase 1 (Sphk1, the enzyme that produces S1P) is significantly elevated in erythrocytes of SCD mice. Chronic treatment of SCD mice with a SphK1 inhibitor significantly attenuated sickling, hemolysis, inflammation and multiple tissue damage by reducing erythrocyte and plasma S1P levels. Erythrocyte S1P levels were further elevated following hypoxia/reoxygenation-induced acute sickle crisis (ASC) in SCD mice and blocking its elevation by a Sphk1 specific inhibitor significantly reduced hallmark features associated with ASC. As with SCD mice, we found that erythrocyte Sphk1 activity and erythrocyte and plasma S1P levels were significantly elevated in humans with SCD compared to normal individuals. Inhibition of SphK1 in cultured primary human erythrocytes isolated from SCD patients inhibited hypoxia-induced elevation of erythrocyte S1P levels and reduced sickling. Thus, we have revealed for the first time that SphK1-mediated S1P elevation in SCD erythrocytes is a key contributor to sickling in SCD and that Sphk1 inhibition can attenuate both acute and chronic sickling events and disease progression. S1P is an important signaling molecule regulating diverse biological processes. Although S1P is predominantly produced and stored in RBCs, nothing was known about the physiological role of S1P in normal RBCs or the pathophysiological role of S1P in SCD until we conducted a metabolomic screen. In an effort to determine the molecular mechanism underlying S1P-induced sickling, we unexpectedly found that S1P directly binds with Hb and results in a reduced Hb-O2 affinity. This finding led us to further discover that 2,3-diphosphoglycerate, another erythrocyte specific allosteric modulator, is required for S1P-mediated allosteric modulation and that these two endogenous heterotropic modulators work cooperatively to induce a substantial reduction in Hb-O2 affinity. Supporting the biochemical and functional findings, molecular modeling predicts that S1P binds near the water filled central cavity of HbA at a site that is different from the Hb-2,3-DPG binding site. Thus, our discovery adds a significant new chapter to erythrocyte physiology by revealing S1P is a novel allosteric modulator of Hb-O2 affinity and also providing a mechanism underlying S1P-mediated sickling by promoting the formation of deoxyHbS. Thus, the work reported here could be the foundation leading to future human trials and a possible therapy for SCD, a life-threatening hemolytic disorder for which the current treatment is extremely limited. The significance of our findings extends well beyond SCD. Our findings reveal a previously unrecognized important role for S1P in erythrocyte physiology and indicate a new concept for the regulation of O2 release from Hb under normal and sickle cell disease conditions. For SCD, elevated S1P is detrimental because reduced Hb-O2 affinity leads to more deoxygenation of HbS, increased sickling and subsequent multiple life-threatening complications. However, for normal erythrocytes, elevated S1P is likely beneficial by decreasing Hb-O2 affinity allowing for more O2 release to hypoxic tissues. Thus, for humans with normal Hb, if elevated S1P can induce O2 release to hypoxic tissues it may be a novel therapeutic target for a range of disorders, from chronic heart failure to diabetic retinopathy, traumatic blood loss, pulmonary disease and even cancer. In this way our findings reveal important novel opportunities to treat and prevent not only SCD but also multiple cardiovascular and pulmonary diseases associated with hypoxia. Thus, the impact of our novel finding is significant and enormous. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3232-3232
Author(s):  
Anren Song ◽  
Yujin Zhang ◽  
Chen Ning ◽  
Almut Grenz ◽  
Holger Etlzschig ◽  
...  

Abstract Abstract 3232 Sickle cell disease (SCD) is a severe genetic disorder with a high morbidity and mortality. Understanding the molecular basis responsible for sickling, a central pathogenesis of SCD, is critical for developing new therapeutic strategies. Using a nonbiased high throughput metabolomic screen, coupled with genetic and pharmacological approaches, recent studies demonstrated that excessive adenosine signaling through the A2B adenosine receptor triggers sickling by induction of 2,3-bisphosphoglycerate (2,3-BPG), an erythroid specific metabolite that induces O2 release from hemoglobin. Adenosine is a signaling nucleoside that elicits many physiological effects by engaging membrane receptors. Notably, equlibrative nucleoside transporters (ENTs) on erythrocytes have been long speculated to regulate extracellular adenosine concentrations under hypoxic conditions. Thus, we hypothesize that ENT is likely a key molecule responsible for elevated circulating adenosine levels and contributing to pathophysiology of SCD. To test this hypothesis, we first conducted western blot analysis to compare expression profiles of ENTs on normal and sickle erythrocytes. We found that ENT1 is the major ENT expressed on both mouse and human erythrocytes. Unexpectedly, ENT1 levels were significantly reduced in sickle erythrocytes compared to normal erythrocytes in both humans and mice, suggesting that ENT1 may contribute to increased adenosine levels seen in SCD. Next, we performed pharmacological studies to determine the exact role of ENT in normal and sickle erythrocytes. We found that treatment with dipyridamole or an ENT1 specific inhibitor (NBMPR) enhanced adenosine-induced elevation of 2,3-BPG in cultured mouse RBCs. Using Hemox Analyzer, we found that co-treatment of adenosine with either dipyridamole or NBMPR resulted in a further right shift of oxygen equilibrium curve (OEC) and further increase in P50 compared to the cells treated with adenosine alone. Similar to our pharmacological studies, we found that genetic deletion of ENT1 further enhanced adenosine-induced 2,3-BPG production in cultured erythrocytes, additional right shift of OEC and increased P50. Extending mouse studies to human, we demonstrate that co-treatment of adenosine with either dipyridamole or NBMPR further enhanced the adenosine alone-mediated 2,3-BPG induction in cultured erythrocytes isolated from normal individuals and SCD patients. Finally, we found that dipyridamole treatment significantly enhanced hypoxia-mediated 2,3-BPG production, right shift of OEC and substantial sickling in cultured erythrocytes isolated from SCD patients. Overall, our studies demonstrate that 1) ENT1 is a major transporter expressed by RBCs and that inhibition or deletion of ENT1 results in enhanced adenosine-mediated 2,3-BPG induction and deoxygenation in normal RBCs; 2) Decreased ENT1 expression in sickle erythrocytes is responsible for elevated circulating adenosine and thereby contributes to sickling by promoting 2,3-BPG production and triggering deoxygenation. Therefore, our findings reveal a previously unrecognized role of ENT1 in erythrocyte physiology, add a new insight to the pathophysiology of SCD and open up new therapies for the disease. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 27 ◽  
pp. 107602962110029
Author(s):  
Mira Merashli ◽  
Alessia Arcaro ◽  
Maria Graf ◽  
Matilde Caruso ◽  
Paul R. J. Ames ◽  
...  

The relationship between antiphospholipid antibodies (aPL) and sickle cell disease (SCD) has never been systematically addressed. Our aim was to evaluate potential links between SCD and aPL in all age groups. EMBASE/PubMed was screened from inception to May 2020 and Peto odds ratios for rare events were calculated. The pooled prevalence (PP) of IgG anticardiolipin antibodies (aCL) was higher in individuals with SCD than in controls (27.9% vs 8.7%, P < 0.0001), that of IgM aCL was similar in the two groups (2.9% vs 2.7%); only individuals with SCD were positive for lupus anticoagulant (LA) (7.7% vs 0%, P < 0.0001). The PP of leg ulcers was similar between aPL positive and negative individuals (44% vs 53%) and between patients in acute crisis and stable patients (5.6% vs 7.3%). Reporting of aPL as a binary outcome and not as a titer precluded further interpretation. The results indicate that a prospective case-control study with serial measurements of a panel of aPL in SCD patients might be warranted, in order to understand further the possible pathogenic role of aPL in SCD.


Antioxidants ◽  
2021 ◽  
Vol 10 (2) ◽  
pp. 296
Author(s):  
Rosa Vona ◽  
Nadia Maria Sposi ◽  
Lorenza Mattia ◽  
Lucrezia Gambardella ◽  
Elisabetta Straface ◽  
...  

Sickle cell disease (SCD) is the most common hereditary disorder of hemoglobin (Hb), which affects approximately a million people worldwide. It is characterized by a single nucleotide substitution in the β-globin gene, leading to the production of abnormal sickle hemoglobin (HbS) with multi-system consequences. HbS polymerization is the primary event in SCD. Repeated polymerization and depolymerization of Hb causes oxidative stress that plays a key role in the pathophysiology of hemolysis, vessel occlusion and the following organ damage in sickle cell patients. For this reason, reactive oxidizing species and the (end)-products of their oxidative reactions have been proposed as markers of both tissue pro-oxidant status and disease severity. Although more studies are needed to clarify their role, antioxidant agents have been shown to be effective in reducing pathological consequences of the disease by preventing oxidative damage in SCD, i.e., by decreasing the oxidant formation or repairing the induced damage. An improved understanding of oxidative stress will lead to targeted antioxidant therapies that should prevent or delay the development of organ complications in this patient population.


Hematology ◽  
2007 ◽  
Vol 2007 (1) ◽  
pp. 84-90 ◽  
Author(s):  
Marilyn J. Telen

AbstractA number of lines of evidence now support the hypothesis that vaso-occlusion and several of the sequelae of sickle cell disease (SCD) arise, at least in part, from adhesive interactions of sickle red blood cells, leukocytes, and the endothelium. Both experimental and genetic evidence provide support for the importance of these interactions. It is likely that future therapies for SCD might target one or more of these interactions.


Hematology ◽  
2013 ◽  
Vol 2013 (1) ◽  
pp. 362-369 ◽  
Author(s):  
Deepa Manwani ◽  
Paul S. Frenette

Abstract Recurrent and unpredictable episodes of vaso-occlusion are the hallmark of sickle cell disease. Symptomatic management and prevention of these events using the fetal hemoglobin–reactivating agent hydroxyurea are currently the mainstay of treatment. Discoveries over the past 2 decades have highlighted the important contributions of various cellular and soluble participants in the vaso-occlusive cascade. The role of these elements and the opportunities for therapeutic intervention are summarized in this review.


2019 ◽  
Author(s):  
Yann Iboudo ◽  
Melanie E. Garrett ◽  
Pablo Bartolucci ◽  
Carlo Brugnara ◽  
Clary B. Clish ◽  
...  

ABSTRACTIn a recent clinical trial, the metabolite L-glutamine was shown to reduce painful crises in sickle cell disease (SCD) patients. To confirm this observation and identify other metabolites implicated in SCD clinical heterogeneity, we profiled 129 metabolites in the plasma of 705 SCD patients. We tested correlations between metabolite levels and six SCD-related complications (painful crises, cholecystectomy, retinopathy, leg ulcer, priapism, aseptic necrosis) or estimated glomerular filtration rate (eGFR), and used Mendelian randomization (MR) to assess causality. We found a causal relationship between L-glutamine levels and painful crises (N=1,278, odds ratio (OR) [95% confidence interval] = 0.68 [0.52 – 0.89], P=0.0048). In two smaller SCD cohorts (N=299 and 406), the protective effect of L-glutamine was observed (OR=0.82 [0.50-1.34]), although the MR result was not significant (P=0.44). We identified 66 significant correlations between the levels of other metabolites and SCD-related complications or eGFR. We tested these correlations for causality using MR analyses and found no significant causal relationship. The baseline levels of quinolinic acid was associated with prospectively ascertained survival in SCD patients, and this effect was dependent on eGFR. Metabolomics provide a promising approach to prioritize small molecules that may serve as biomarkers or drug targets in SCD.


Sign in / Sign up

Export Citation Format

Share Document