Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Nov 1.
Published in final edited form as: Br J Haematol. 2024 Sep 15;205(5):1974–1984. doi: 10.1111/bjh.19758

Genetic variants associated with white blood cell count among individuals with sickle cell disease

Mina Cintho Ozahata 1, Yuelong Guo 2, Isabel Gomes 3, Barbara Malta 3, André Belisário 3, Luiz Amorim 4, Dahra Teles 5, Miriam Park 6, Shannon Kelly 7, Ester C Sabino 9, Grier P Page 2, Brian Custer 8, Carla L Dinardo 9, International Component of the NHLBI Recipient Epidemiology and Donor Evaluation Study (REDS-III) and for the TOPMed (NHLBI TransOmics for Precision Medicine) SCD working
PMCID: PMC11568933  NIHMSID: NIHMS2020222  PMID: 39279196

Abstract

Background:

Sickle cell disease (SCD) is a Mendelian disorder characterized by a point mutation in the β-globin gene that leads to sickling of erythrocytes. Several studies have shown that absolute neutrophil count is strongly associated with clinical severity of SCD, suggesting an apparent role of white blood cells (WBC) in SCD pathology. However, the mechanism by which genetic variants lead to white blood cells count differences in SCD patients remains unclear.

Methods:

Genome wide association (GWA) analyses were carried out among a cohort of 2,409 Brazil SCD participants. Association of white blood cells (WBC) count and genetic markers were investigated in homozygous sickle cell anemia (HbSS) participants and compound heterozygous sickle cell hemoglobin C (HbSC) participants.

Results:

GWA analysis showed that variants in genes TERT, ACKR1 and FAM3C are associated with WBC count variation. The well-studied association between WBC count and Duffy null phenotype (variant in ACKR1) in healthy populations was replicated, reinforcing the influence of the SNP rs2814778 (T>C) in WBC count.

Conclusion:

Genetics plays an important role in regulating WBC count in patients with SCD. Our results point to possible mechanisms involved in WBC count variation and as increased WBC count is associated with more severe SCD, these results could suggest potential therapeutic targets for individuals with SCD.

Graphical Abstract

graphic file with name nihms-2020222-f0001.jpg

Several studies have shown that absolute neutrophil count is strongly associated with clinical severity of SCD, suggesting an apparent role of white blood cells (WBC) in SCD pathology. This genome wide association (GWA) analyses, carried out among a cohort of 2,409 Brazil SCD participants, showed that variants in genes TERT, ACKR1 and FAM3C are associated with WBC count variation. The well-studied association between WBC count and Duffy null phenotype (variant in ACKR1) in healthy populations was replicated, reinforcing the influence of the SNP rs2814778 (T>C) in WBC count. Genetics plays an important role in regulating WBC count in patients with SCD and our results point to the possible mechanisms involved in WBC count variation and potential therapeutic targets for patients with SCD.

Introduction

Sickle cell disease (SCD) is caused by a variety of mutations in the β-globin gene, resulting in the production of sickle-shaped red blood cells (RBCs). Disease severity varies widely and is in part associated with individual genetics. For example, patients with sickle cell anemia (SCA) who carry two copies of hemoglobin S (HbS; SCA patients commonly referred to as HbSS patients) usually present more severe clinical phenotypes compared to patients who carry one copy of HbS and one copy of hemoglobin C (HbC; patients commonly referred to as HbSC patients).

Despite SCD being a genetic disorder that primarily leads to hemoglobin S (HbS) polymerization, which alters RBC morphology, white blood cells (WBC) also play a role in disease progression (reviewed in references (1-3)). Early observational studies have shown that high neutrophil count is associated with severity of clinical manifestations (4), elevated white blood cell count is positively correlated with early death (5), clinically overt strokes (6), silent cerebral infarction (7), and acute chest syndromes (8) in patients with SCD. The observation that sickle RBCs bind to neutrophils both in vitro (9) and in vivo in Berkeley mice (10,11) suggests that neutrophils may be directly involved in vascular occlusion. Recent studies have linked inflammatory cytokines and inflammatory responses to clinical complications including pulmonary hypertension (12) and leg ulcers, (13) suggesting another plausible mechanism for how WBC may be involved in SCD clinical manifestations.

Hydroxyurea has been widely used to treat severe clinical complications such as vaso-occlusive pain episodes and acute chest syndrome in patients with SCD. (14,15) One mechanism of action of hydroxyurea is through inducing fetal hemoglobin (HbF) production. (16,17) Interestingly, some patients who have no increase in HbF after hydroxyurea therapy experience clinical benefits (14) and most patients who respond well to treatment have a decrease in neutrophil count. (15) This suggests a plausible causal relationship between WBC count and SCD clinical complications. Studying genetic factors that affect WBC may have implications for optimizing hydroxyurea as achieving maximum tolerated dose may be challenging in patients with lower baseline WBC.

Several GWA studies in European, (18-23) Asian, (24-26),African or African American (21-23,27,28) and Hispanic populations, (29) have demonstrated the contribution of genetics to WBC count. Most noticeably, a GWA study that included more than 170,000 participants (from the UK Biobank study, (30) the UK BiLEVE study, (31) and the INTERVAL study (32)) identified 180 loci associated with total WBC count and 764 loci associated with at least one WBC index.(18) Arguably, one of the most successful stories on functional studies following GWA-identified variants is the one addressing the mechanisms of benign ethnic neutropenia (BEN), that identified the association between reduced WBC count and the Duffy antigen promoter variant rs2814778 (ACKR1: c.−67T>C). This variant is most prevalent among African populations (1000 genome African C allele frequency [AF] 0.964) and is rare among other populations (1000 genome European AF 0.006, American AF 0.08, and Asian AF 0). A functional study focusing on this variant explained the mechanism by which this variant leads to neutropenia. (33) Despite these successes, these studies enrolled mostly healthy participants who are not exposed to chronic baseline inflammation; therefore, our knowledge remains limited on how genetics are associated with WBC regulation under inflammatory exposure. We address specifically the inflammatory exposure of patients with SCD in this study. Several GWA studies have aimed to investigate alloimmunization susceptibility, (34,35) vaso-occlusive pain, (36) and fetal hemoglobin levels (37,38) among patients with SCD, but less is unknown about the genetic variation that contributes to WBC count. To identify genetic factors associated with WBC count in patients with an inflammatory pathophysiology such as SCD, we performed a GWA analysis in Brazilian individuals with HbSS and HbSC.

Methods

Study Population

The Brazil SCD cohort was established as part of the NHLBI-funded Recipient Epidemiology and Donor Evaluation Study-III (REDS-III). (40) In total, 2,793 participants enrolled into the study with SCD type diagnosed in the treating hemocenters and independently confirmed by pyrosequencing. HbSS was the most common genotype (1973 participants) followed by HbSC (644 participants), Sβ0 (83 participants), and Sβ+ (80 participants). (40)

Protection of human subjects was conducted according to federal regulations and protocols applicable to all human subject research and was supervised by NHLBI’s REDS-III data coordinating center (DCC, RTI International, Rockville, MD). All study procedures were approved by Brazilian Center’s IRB, Vitalant Research Institute, and RTI International.

Data collection and instruments

Clinical and laboratory data were collected according to standardized procedures previously described. (40) Clinical outcomes were defined according to standardized definitions of SCD phenotypic manifestations. (41) Steady-state WBC counts were measured for routine clinical care using automated analyzers. Steady state was defined as no hospitalizations, blood transfusions (for those not on chronic blood transfusion) or clinical complications that may affect the laboratory parameters in the one month prior to measurement of white blood cell count.

Data collected in the Recipient Epidemiology and Donor Evaluation Study (REDS-III) is available through the American National Institute of Health database of Genotypes and Phenotypes (dbGaP) upon request.

Statistical Analyses

Qualitative variables were shown as frequencies, and quantitative variables by mean (if it was normally distributed) or median (otherwise). The quantitative variable’s normality was assessed by Shapiro-Wilk test. The WBC count was transformed using log plus one function to reach the normality. WBC values outside mean ± 4 standard deviations were considered outliers and removed. The association between selected characteristics and the transformed WBC count were evaluated using a linear regression model. The R (42) version 4.1.2 was used to perform the analysis, considering significant p<0.05.

Genome-wide association analysis

Blood collection (40), sequence data processing, quality control and genotype calling were described elsewhere, (43) as whole-genome sequencing was accomplished by the Trans-Omics in Precision Medicine (TOPMed) program supported by the National Heart, Lung and Blood Institute (NHLBI). Of the total of 2617 study participants with HbSS and HbSC, 2409 had whole genome sequence and WBC data available after quality control procedures. Association analysis was conducted using ENCORE (University of Michigan), a web-based tool which allows the execution of GWA analysis of uploaded phenotypes against TOPMed genotypes. A linear mixed model was used, with adjustment for kinship and all previously identified covariates that were found to be associated with WBC counts in addition to the 10 first principal components to correct for population stratification.

Results

Overall Distribution of WBC Counts in the Study Population

There were 2793 participants included in the overall REDS-III Brazil SCD cohort (1973 HbSS, 644 HbSC, 83 HbSβ0-thalassemia, 80 HbSβ+-thalassemia and 13 with other rare compound hemoglobin mutations). Due to low numbers of HbSβ-thalassemia and other genotypes in the cohort, this analysis included only the HbSS and HbSC participants with available whole genome sequencing data. Both genotype groups were similar in age with median and mean ages of 15 and 20.7 years in the HbSC participants and 17 and 20.0 years in the HbSS participants, respectively. The distribution of sexes was also comparable in the two groups, with males accounting for 44.4% of HbSC participants and 45% of the HbSS participants. Regarding treatment, 7.3% of HbSC participants were treated with hydroxyurea, whereas 38.9% of HbSS participants were treated. Additionally, 4.97% of HbSC participants had received RBC transfusion in the last 12 months, compared to 37.6% of HbSS participants. The median WBC count of analyzed participants was 10,600 cells/μL (mean 10,978 cells/μL). Participants with HbSC had lower WBC (mean 8,835 cells/μL; median 8,510 cells/μL) than participants with HbSS (mean 11,685 cells/μL; median 11,343 cells/μL; p < 0.0001) (Table 1).

Table 1.

Demographic and clinical variables associated with WBC count. WBC count is shown as cells/μL. P-value was calculated using age as a numerical, not categorical variable.

Characteristic WBC count (median) P-value
SCD genotype <0.0001
 SC 8,510
 SS 11,343.33
Sex 0.017
 female 10,800
 male 10,400
Age <0.0001
 0 - 4 13,692
 5 - 9 11,533
 10 - 17 10,731
 18 -29 10,685
 30 - 39 9,800
 40 - 49 8,220
 50 - 59 8,308
 60 + 7,318
Enrollment center
 Hemominas HBH 9,430 <0.0001
 Hemominas JFO 10,200 0.29
 Hemominas MOC 11,266.67 0.006
 Hemope 11,140 <0.0001
 Hemorio 11,293.33 <0.0001
 ITACI SP 9,983.33 0.24
Recent hydroxyurea use (yes) 9,877.92 <0.0001
Recent transfusion 11,903.33 <0.0001
Ever transfused 10,905 <0.0001
Number of transfusions <0.0001
Number of hospitalized pains <0.0001

WBC Counts Associated with Age, SCD genotype, Hydroxyurea Use, Hemocenter, and Transfusion

Overall distribution of WBC count results among participants with HbSS and HbSC was right skewed as shown in Figure 1a. Log plus one transformation brought the data to approximately normal distribution (Figure 1b). WBC count was associated with age (p < 0.0001), enrollment center (p < 0.0001), SCD genotype (p < 0.0001), recent hydroxyurea (p < 0.0001) use and recent transfusion history (p < 0.0001) (Table 1).

Figure 1.

Figure 1.

Distribution of leukocyte results. a. Distribution of raw WBC counts in HbSS (blue) and HbSC (red) participants. The QQ plot insert shows the distribution of WBC count data (x-axis) as compared to quantile of normal distribution (y-axis), blue line is a 45-degree line that goes through origin. b. Distribution of log plus one transformed WBC counts in HbSS (blue) and HbSC (red) participants. QQ plot shows that log plus one transformation effectively brings leukocyte results in approximately normal distribution.

Multivariate regression was used to model association between WBC counts and demographic and clinical variables. WBC count was negatively associated with age, with only slight decrease as age increases (Table 2). Current use of hydroxyurea was associated with significant decrease of WBC counts (p < 0.0001). The effect size of hydroxyurea use was −0.104, suggesting that the mean log transformed WBC count result was 0.104 lower in participants who recently used hydroxyurea compared to those who did not. The raw WBC count was approximately 14% lower in the recent hydroxyurea use group. The effect of age and hydroxyurea use were similar among participants with HbSS and HbSC as shown in the stratified models (Table 2).

Table 2.

Multivariate modeling the effect of age, sex, hemo centers, hydroxyurea use, transfusion, and hospitalized pain episodes on leukocyte results. Three statistical models were conducted, one jointly analyzing HbSS and HbSC participants, one restricted to HbSS only participants, and one restricted to HbSC only participants. Shown in the table are effect sizes of each variable on the log transformed leukocyte results after adjusting for other covariates in the model (Beta column), the equivalent percent difference after reverse transformation (Pct. Diff. column), and the corresponding p-values (P column).

Combined model HbSS participants HbSC participants
Beta P Beta P Beta P
Intercept 5.136 5.140 4.916
SCD type −0.136 <0.0001
Age −0.003 <0.0001 −0.003 <0.0001 −0.001 <0.0001
Sex (female) −0.004 0.051 −0.006 0.353 0.002 0.177
Hemominas JFO 0.019 0.084 0.022 0.065 0.013 0.512
Hemominas MOC 0.033 0.001 0.045 0.0001 0.010 0.390
Hemope 0.052 <0.0001 0.067 <0.0001 −0.056 0.082
Hemorio 0.043 <0.0001 0.055 <0.0001 0.003 0.095
ITACI SP −0.0079 0.716 −0.008 0.721 0.005 0.148
Recent hydroxyurea use −0.104 <0.0001 −0.105 <0.0001 −0.103 <0.0001
Transfused within 12 months −0.030 <0.0001 −0.032 <0.0001 0.039 0.388
Number of hospitalized pains −0.00004 0.888 −0.00004 0.873 0.0016 0.255

Transfusion history was also associated with increased WBC counts (Table 1). This may be because patients requiring more frequent transfusions are more severe and the elevated WBC reflects that. Participant transfusion history was coded into three variables: (a) whether a participant had ever received blood transfusion (ever transfused), (b) number of historical transfusions received by a participant (number of transfusions), and (c) whether a participant received a blood transfusion within 12 months prior to enrollment (recent transfusion). We conducted additional statistical modeling to evaluate which of these variables better models the effect to WBC count. Although all three ways of coding transfusion history showed association with WBC counts, recent transfusions were most correlated with elevated WBC counts. Ever transfused and number of historical transfusions were not significantly associated with WBC count when adjusted for recent transfusions (Table 1).

Number of pain episodes hospitalizations were associated with elevated WBC counts when modeled in a univariate model (Table 1) but were not statistically significant when adjusted for transfusion history (Table 2). Therefore, recent transfusion history was used as a covariate in the genetic models.

Genome Wide Association of WBC Counts

Genetic markers associated with WBC count were interrogated using genome wide association analyses, adjusted for the identified demographic and clinical covariates (age, sex, SCD genotype, enrollment center, recent hydroxyurea use, and recent transfusion history). Results are shown in Figure 2. There were 4 genome wide significant hits (P < 5 x 10−8).

Figure 2.

Figure 2.

Manhattan plot and QQ plot for GWA analyses restricted to HbSS participants.

Top variant rs772442796 (p-value 1.14 x 10−8, beta −2.18) , is a C deletion with minor allele frequency 0.1038% in the study population (0.0219% in Topmed, 0% in African and 0.02% in European ALFA population (39)) located in chromosome 5. This variant is in the intronic region of gene telomerase reverse transcriptase (TERT) (Figure 3). The presence of one copy of the rs772442796 variant was linked to an approximate reduction of 2.18 in the log plus one wbc count.

Figure 3.

Figure 3.

LocusZoom single variant association regional association plot for GWA top hit on chromosome 5.

Variant rs2814778 (p-value 1.16 x 10−8, beta −0.16) in chromosome 1 also reached genomic wide significance. One copy of the rs2814778 variant was associated with an estimated decrease in log plus one wbc count by 0.16. rs2814778 (c.−67T>C) is in the promoter region of atypical chemokine receptor 1 (ACKR1) gene (Figure 4). The C allele of this variant is most prevalent in the African population (1000 genome African allele frequency 0.964, European allele frequency 0.006) and is associated with approximately 20% decrease of WBC count in healthy individuals. The Brazilian population is an admixture population that exhibits both C and T alleles with modest frequency for rs2814778 (616 C/C, 1,290 C/T, and 838 T/T in the study population).

Figure 4.

Figure 4.

LocusZoom single variant association regional association plot for GWA top hit on chromosome 1

Two other SNPs reached genome wide significance, SNPs rs151288099 (p-value 2.24 x 10−8, beta −0.28) and rs60492718 (p-value 3.14 x 10−8, beta 0.27) in gene FAM3 metabolism regulating signaling molecule C (FAM3C) in chromosome 7 (Figure 5). 8 other SNPs in FAM3C had P-values close to genome wide significance (P < 8 x 10−8), adding to evidence of association between the gene and WBC. The presence of one copy of the rs151288099 variant was associated with an estimated decrease in log plus one wbc count by 0.28 and the presence of one copy of the rs60492718 variant was associated with an estimated increase in log plus one wbc count by 0.27.

Figure 5.

Figure 5.

LocusZoom single variant association regional association plot for GWA top hit on chromosome 7

In order to test if the SNPs significantly associated with WBC were only associated with WBC as a surrogate marker of disease severity, we tested the association of the SNPs with other markers of disease severity such as 3 or main pain crisis hospitalizations per year, stroke, priapism and acute chest syndrome. However, none of the SNPs associated with WBC were associated with these phenotypes

Discussion

The mutation that causes sickle cell disease only directly affects RBCs, however, it is known that WBCs contribute to the pathophysiology . Sickle cell severity varies significantly from patient to patient and WBC counts are associated with important SCD complications (4-8,12,13), vaso-occlusion (7,9) and mortality (4,5), Therefore, in an effort to help elucidate genetic factors influencing WBC count, and consequently disease severity, a GWAS within the REDS-III Brazil SCD cohort was performed.

The observed median WBC count (10,600 cells/μL with mean 10,978 cells/μL) was congruent with the observation of elevated WBC in SCD patients (5) (normal range 3,500 to 10,500 cells/μL). WBC count was associated with age, recent hydroxyurea use and recent transfusion history. In the genetic analysis, 4 SNPs were found to be associated with WBC count in this cohort.

The GWAS top hit was in TERT, a telomerase protein subunit that adds TTAGGG repeats to chromosome ends with the help of a template coded by gene TERC. (40) Variations in TERT were previously associated with telomere length in TOPMED data. (41) Telomeres are regions of repetitive DNA sequence in the end of chromosomes.

Although TERT has been associated with telomere length, it has also been associated with the development of bone marrow failure. (42,43) The association may be related to the occurrence of short telomeres in patients with aplastic anemia. (44,45) In aplastic anemia, the bone marrow consists mainly of fat and very few hematopoietic cells.(43)

Previous GWAS also found associations between polymorphisms in TERT and platelet (18,46,47), granulocyte (48), neutrophil (46,47) and eosinophil (49) counts. GWAS also found associations between SNPs in TERT and neutrophil percentage of leukocytes, (18) myeloid white cell counts, (18) eosinophil percentage of leukocytes, (49) monocyte percentage of white cells, (49) lymphocyte percentage of leukocytes (49) and myeloproliferative disorder. (50)

A known association of WBC and SNP rs2814778 was replicated in this study. rs2814778 is known to be associated with WBC count and has been replicated across multiple previous GWA studies. (18,20,28,29,51,52) This variant (c.−67T>C) is in the FY*B promoter region of ACKR1 gene responsible for a common phenotype Fy(a-b-) present most in West Africans and rare in Caucasians. (53,54) ACKR1 is also known as the Duffy Antigen Receptor for Chemokines, or DARC. Chemokines attract specific subpopulations of WBCs to inflammation sites. (55) In individuals with HbSS, variant in rs2814778 was associated with end-organ damage (56) and Duffy expression was also associated with WBC count. (56)

Two SNPs in FAM3C reached genome wide significance and 8 other SNPs had P-values close to genome significance (P-values < 8 x 10−8). Polymorphisms in FAM3C were previously associated with bone mineral density (BMD) and fractures. (50,53,54,56) FAM3C is hypothesized to have roles in epithelial-to-mesenchymal transition (EMT), tumor growth, metastasis, osteoblast differentiation and bone homeostasis. (57-59) In a study with FAM3C knockout mouses, although no differences were found in female mouses, in male knockout mouses the percentage of neutrophils out of WBCs was significantly increased and the percentage of lymphocytes out of WBCs reduced compared to wild-type male mouses. (59) Additionally, a study found an association between high BMD loss increased neutrophil, decreased monocyte and decreased lymphocyte counts. (60)

Conclusion

In this study, we investigated genetic factors influencing WBC count variation in patients with SCD. We replicated a well-known mutation associated with WBC count in healthy people and found two additional genes. These findings show the importance of genetic factors and help elucidate pathways and genes that contribute to WBC count variation and that might influence SCD severity.

Further studies are required to reveal the specific mechanisms involved in WBC count variation, but if confirmed these pathways and genes may be candidate targets for furture SCD treatments..

Figure 6.

Figure 6.

Frequency of variants that reached genomic significance.

Acknowledgements

Molecular data for the Trans-Omics in Precision Medicine (TOPMed) program was supported by the National Heart, Lung and Blood Institute (NHLBI). WGS for "NHLBI TOPMed: REDSIII" (phs001468) was performed at Washington University (HHSN268201500015C).

Core support including centralized genomic read mapping and genotype calling, along with variant quality metrics and filtering were provided by the TOPMed Informatics Research Center (3R01HL-117626-02S1; contract HHSN268201800002I). Core support including phenotype harmonization, data management, sample-identity QC, and general program coordination were provided by the TOPMed Data Coordinating Center (R01HL-120393; U01HL-120393; contract HHSN268201800001I). We gratefully acknowledge the studies and participants who provided biological samples and data for TOPMed.

REDS-III was supported by the National Institutes of Health, National Heart, Lung, and Blood Institute under Grants HHSN2682011-00001I, −00002I, −00003I, −00004I, −00005I, −00006I, −00007I, −00008I, and −00009I.

Mina Cintho and Yuelong Guo cleaned, analyzed and interpreted the data and drafted the manuscript. Barbara Malta, André Belisário, Luiz Amorim, Dahra Teles and Miriam Park collected the data, reviewed and edited the manuscript. Grier P. Page supervised the analysis and data interpretation. Shannon Kelly, Ester C. Sabino, and Brian Custer supervised the protocol design, and reviewed, edited and approved the manuscript. Carla L. Dinardo conceptualized the work and supervised, reviewed, edited and approved the manuscript

Footnotes

conflict of interest disclosure

Authors have no conflict of interest with regard to publication of this manuscript

ethics approval statement

This research was approved by all relevant ethical committee/IRB in both the US and Brazil, including the Brazilian National Committee of Ethics in Research, local ethical committees at each participating site as well as the IRBs of the REDS-III data coordinating center (RTI International Rockville, MD) and UCSF, the IRB of record for Vitalant Research Institute.

patient consent statement

All participants (or guardians of participants <18 years) signed informed consent to participate in the REDS-III Brazil SCD cohort and allow their DNA and data to be used in GWAS.

data availability statement

Data collected in the Recipient Epidemiology and Donor Evaluation Study (REDS-III) is available through the American National Institute of Health database of Genotypes and Phenotypes (dbGaP) upon request.

References

  • 1.Conran N, Belcher JD. Inflammation in sickle cell disease. Connes P, editor. Clin Hemorheol Microcirc. 2018. Mar 28;68(2–3):263–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Zhang D, Xu C, Manwani D, Frenette PS. Neutrophils, platelets, and inflammatory pathways at the nexus of sickle cell disease pathophysiology. Blood. 2016. Feb 18;127(7):801–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Okpala I. The intriguing contribution of white blood cells to sickle cell disease – a red cell disorder. Blood Rev. 2004. Mar;18(1):65–73. [DOI] [PubMed] [Google Scholar]
  • 4.Anyaegbu CC, Okpala IE, Aken’Ova YA, Salimonu LS. Peripheral blood neutrophil count and candidacidal activity correlate with the clinical severity of sickle cell anaemia (SCA). Eur J Haematol. 1998. Apr;60(4):267–8. [DOI] [PubMed] [Google Scholar]
  • 5.Platt OS, Brambilla DJ, Rosse WF, Milner PF, Castro O, Steinberg MH, et al. Mortality In Sickle Cell Disease -- Life Expectancy and Risk Factors for Early Death. N Engl J Med. 1994. Jun 9;330(23):1639–44. [DOI] [PubMed] [Google Scholar]
  • 6.Ohene-Frempong K, Weiner SJ, Sleeper LA, Miller ST, Embury S, Moohr JW, et al. Cerebrovascular accidents in sickle cell disease: rates and risk factors. Blood. 1998. Jan 1;91(1):288–94. [PubMed] [Google Scholar]
  • 7.Kinney TR, Sleeper LA, Wang WC, Zimmerman RA, Pegelow CH, Ohene-Frempong K, et al. Silent cerebral infarcts in sickle cell anemia: a risk factor analysis. The Cooperative Study of Sickle Cell Disease. Pediatrics. 1999. Mar;103(3):640–5. [DOI] [PubMed] [Google Scholar]
  • 8.Castro O, Brambilla DJ, Thorington B, Reindorf CA, Scott RB, Gillette P, et al. The acute chest syndrome in sickle cell disease: incidence and risk factors. The Cooperative Study of Sickle Cell Disease. Blood. 1994. Jul 15;84(2):643–9. [PubMed] [Google Scholar]
  • 9.Hofstra TC, Kalra VK, Meiselman HJ, Coates TD. Sickle erythrocytes adhere to polymorphonuclear neutrophils and activate the neutrophil respiratory burst. Blood. 1996. May 15;87(10):4440–7. [PubMed] [Google Scholar]
  • 10.Turhan A, Weiss LA, Mohandas N, Coller BS, Frenette PS. Primary role for adherent leukocytes in sickle cell vascular occlusion: a new paradigm. Proc Natl Acad Sci U S A. 2002. Mar 5;99(5):3047–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Pászty C, Brion CM, Manci E, Witkowska HE, Stevens ME, Mohandas N, et al. Transgenic knockout mice with exclusively human sickle hemoglobin and sickle cell disease. Science. 1997. Oct 31;278(5339):876–8. [DOI] [PubMed] [Google Scholar]
  • 12.Ataga KI, Moore CG, Hillery CA, Jones S, Whinna HC, Strayhorn D, et al. Coagulation activation and inflammation in sickle cell disease-associated pulmonary hypertension. Haematologica. 2008. Jan;93(1):20–6. [DOI] [PubMed] [Google Scholar]
  • 13.Minniti CP, Delaney KMH, Gorbach AM, Xu D, Lee CCR, Malik N, et al. Vasculopathy, inflammation, and blood flow in leg ulcers of patients with sickle cell anemia. Am J Hematol. 2014. Jan;89(1):1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Charache S, Terrin ML, Moore RD, Dover GJ, Barton FB, Eckert SV, 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. May 18;332(20):1317–22. [DOI] [PubMed] [Google Scholar]
  • 15.Charache S. Mechanism of action of hydroxyurea in the management of sickle cell anemia in adults. Semin Hematol. 1997. Jul;34(3 Suppl 3):15–21. [PubMed] [Google Scholar]
  • 16.Veith R, Galanello R, Papayannopoulou T, Stamatoyannopoulos G. Stimulation of F-Cell Production in Patients with Sickle-Cell Anemia Treated with Cytarabine or Hydroxyurea. N Engl J Med. 1985. Dec 19;313(25):1571–5. [DOI] [PubMed] [Google Scholar]
  • 17.Platt OS, Orkin SH, Dover G, Beardsley GP, Miller B, Nathan DG. Hydroxyurea enhances fetal hemoglobin production in sickle cell anemia. J Clin Invest. 1984. Aug;74(2):652–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Astle WJ, Elding H, Jiang T, Allen D, Ruklisa D, Mann AL, et al. The Allelic Landscape of Human Blood Cell Trait Variation and Links to Common Complex Disease. Cell. 2016. Nov 17;167(5):1415–1429.e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Soranzo N, Spector TD, Mangino M, Kühnel B, Rendon A, Teumer A, et al. A genome-wide meta-analysis identifies 22 loci associated with eight hematological parameters in the HaemGen consortium. Nat Genet. 2009. Nov;41(11):1182–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Nalls MA, Couper DJ, Tanaka T, van Rooij FJA, Chen MH, Smith AV, et al. Multiple loci are associated with white blood cell phenotypes. PLoS Genet. 2011. Jun;7(6):e1002113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Li J, Glessner JT, Zhang H, Hou C, Wei Z, Bradfield JP, et al. GWAS of blood cell traits identifies novel associated loci and epistatic interactions in Caucasian and African-American children. Hum Mol Genet. 2013. Apr 1;22(7):1457–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Crosslin DR, McDavid A, Weston N, Nelson SC, Zheng X, Hart E, et al. Genetic variants associated with the white blood cell count in 13,923 subjects in the eMERGE Network. Hum Genet. 2012. Apr;131(4):639–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lo KS, Wilson JG, Lange LA, Folsom AR, Galarneau G, Ganesh SK, et al. Genetic association analysis highlights new loci that modulate hematological trait variation in Caucasians and African Americans. Hum Genet. 2011. Mar;129(3):307–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Okada Y, Hirota T, Kamatani Y, Takahashi A, Ohmiya H, Kumasaka N, et al. Identification of nine novel loci associated with white blood cell subtypes in a Japanese population. PLoS Genet. 2011. Jun;7(6):e1002067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kamatani Y, Matsuda K, Okada Y, Kubo M, Hosono N, Daigo Y, et al. Genome-wide association study of hematological and biochemical traits in a Japanese population. Nat Genet. 2010. Mar;42(3):210–5. [DOI] [PubMed] [Google Scholar]
  • 26.Kong M, Lee C. Genetic associations with C-reactive protein level and white blood cell count in the KARE study. Int J Immunogenet. 2013. Apr;40(2):120–5. [DOI] [PubMed] [Google Scholar]
  • 27.Auer PL, Johnsen JM, Johnson AD, Logsdon BA, Lange LA, Nalls MA, et al. Imputation of exome sequence variants into population- based samples and blood-cell-trait-associated loci in African Americans: NHLBI GO Exome Sequencing Project. Am J Hum Genet. 2012. Nov 2;91(5):794–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Reiner AP, Lettre G, Nalls MA, Ganesh SK, Mathias R, Austin MA, et al. Genome-wide association study of white blood cell count in 16,388 African Americans: the continental origins and genetic epidemiology network (COGENT). PLoS Genet. 2011. Jun;7(6):e1002108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Jain D, Hodonsky CJ, Schick UM, Morrison JV, Minnerath S, Brown L, et al. Genome-wide association of white blood cell counts in Hispanic/Latino Americans: the Hispanic Community Health Study/Study of Latinos. Hum Mol Genet. 2017. Mar 15;26(6):1193–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Sudlow C, Gallacher J, Allen N, Beral V, Burton P, Danesh J, et al. UK biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med. 2015. Mar;12(3):e1001779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wain LV, Shrine N, Miller S, Jackson VE, Ntalla I, Artigas MS, et al. Novel insights into the genetics of smoking behaviour, lung function, and chronic obstructive pulmonary disease (UK BiLEVE): a genetic association study in UK Biobank. Lancet Respir Med. 2015. Oct;3(10):769–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Moore C, Sambrook J, Walker M, Tolkien Z, Kaptoge S, Allen D, et al. The INTERVAL trial to determine whether intervals between blood donations can be safely and acceptably decreased to optimise blood supply: study protocol for a randomised controlled trial. Trials. 2014. Dec;15(1):363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Duchene J, Novitzky-Basso I, Thiriot A, Casanova-Acebes M, Bianchini M, Etheridge SL, et al. Atypical chemokine receptor 1 on nucleated erythroid cells regulates hematopoiesis. Nat Immunol. 2017. Jul;18(7):753–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hanchard NA, Moulds JM, Belmont JW, Chen A. A Genome-Wide Screen for Large-Effect Alloimmunization Susceptibility Loci among Red Blood Cell Transfusion Recipients with Sickle Cell Disease. Transfus Med Hemotherapy. 2014;41(6):453–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Dworkis DA, Timofeev N, Milton JN, Hartley SW, Gupta M, Sebastiani P, et al. A Genome-Wide Association Study of the Alloimmunization Responder Phenotype in Sickle Cell Disease. Blood. 2009. Nov 20;114(22):2551–2551. [Google Scholar]
  • 36.Chaturvedi S, Bhatnagar P, Bean CJ, Steinberg MH, Milton JN, Casella JF, et al. Genome-wide association study to identify variants associated with acute severe vaso-occlusive pain in sickle cell anemia. Blood. 2017. Aug 3;130(5):686–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Timofeev N, Sebastiani P, Hartley SH, Baldwin CT, Steinberg MH. Fetal Hemoglobin in Sickle Cell Anemia: A Genome-Wide Association Study of the Response to Hydroxyurea. Blood. 2008. Nov 16;112(11):2471–2471. [Google Scholar]
  • 38.Mtatiro SN, Singh T, Rooks H, Mgaya J, Mariki H, Soka D, et al. Genome wide association study of fetal hemoglobin in sickle cell anemia in Tanzania. PloS One. 2014;9(11):e111464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Phan L, Jin Y, Zhang H, Qiang W, Shekhtman E, Shao D. ALFA: Allele Frequency Aggregator. 2020; [Google Scholar]
  • 40.Weinrich SL, Pruzan R, Ma L, Ouellette M, Tesmer VM, Holt SE, et al. Reconstitution of human telomerase with the template RNA component hTR and the catalytic protein subunit hTRT. Nat Genet. 1997. Dec;17(4):498–502. [DOI] [PubMed] [Google Scholar]
  • 41.Taub MA, Conomos MP, Keener R, Iyer KR, Weinstock JS, Yanek LR, et al. Genetic determinants of telomere length from 109,122 ancestrally diverse whole-genome sequences in TOPMed. Cell Genomics. 2022. Jan 12;2(1):100084, S2666-979X(21)00105-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Vulliamy TJ, Walne A, Baskaradas A, Mason PJ, Marrone A, Dokal I. Mutations in the reverse transcriptase component of telomerase (TERT) in patients with bone marrow failure. Blood Cells Mol Dis. 2005;34(3):257–63. [DOI] [PubMed] [Google Scholar]
  • 43.Yamaguchi H, Calado RT, Ly H, Kajigaya S, Baerlocher GM, Chanock SJ, et al. Mutations in TERT, the gene for telomerase reverse transcriptase, in aplastic anemia. N Engl J Med. 2005. Apr 7;352(14):1413–24. [DOI] [PubMed] [Google Scholar]
  • 44.Ball SE, Gibson FM, Rizzo S, Tooze JA, Marsh JC, Gordon-Smith EC. Progressive telomere shortening in aplastic anemia. Blood. 1998. May 15;91(10):3582–92. [PubMed] [Google Scholar]
  • 45.Brümmendorf TH, Maciejewski JP, Mak J, Young NS, Lansdorp PM. Telomere length in leukocyte subpopulations of patients with aplastic anemia. Blood. 2001. Feb 15;97(4):895–900. [DOI] [PubMed] [Google Scholar]
  • 46.Kachuri L, Jeon S, DeWan AT, Metayer C, Ma X, Witte JS, et al. Genetic determinants of blood-cell traits influence susceptibility to childhood acute lymphoblastic leukemia. Am J Hum Genet. 2021. Oct 7;108(10):1823–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chen MH, Raffield LM, Mousas A, Sakaue S, Huffman JE, Moscati A, et al. Trans-ethnic and Ancestry-Specific Blood-Cell Genetics in 746,667 Individuals from 5 Global Populations. Cell. 2020. Sep 3;182(5):1198–1213.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kanai M, Akiyama M, Takahashi A, Matoba N, Momozawa Y, Ikeda M, et al. Genetic analysis of quantitative traits in the Japanese population links cell types to complex human diseases. Nat Genet. 2018. Mar;50(3):390–400. [DOI] [PubMed] [Google Scholar]
  • 49.Vuckovic D, Bao EL, Akbari P, Lareau CA, Mousas A, Jiang T, et al. The Polygenic and Monogenic Basis of Blood Traits and Diseases. Cell. 2020. Sep 3;182(5):1214–1231.e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Bao EL, Nandakumar SK, Liao X, Bick AG, Karjalainen J, Tabaka M, et al. Inherited myeloproliferative neoplasm risk affects haematopoietic stem cells. Nature. 2020. Oct;586(7831):769–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Tajuddin SM, Schick UM, Eicher JD, Chami N, Giri A, Brody JA, et al. Large-Scale Exome-wide Association Analysis Identifies Loci for White Blood Cell Traits and Pleiotropy with Immune-Mediated Diseases. Am J Hum Genet. 2016. Jul 7;99(1):22–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Reich D, Nalls MA, Kao WHL, Akylbekova EL, Tandon A, Patterson N, et al. Reduced neutrophil count in people of African descent is due to a regulatory variant in the Duffy antigen receptor for chemokines gene. PLoS Genet. 2009. Jan;5(1):e1000360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zhang LS, Hu HG, Liu YJ, Li J, Yu P, Zhang F, et al. A follow-up association study of two genetic variants for bone mineral density variation in Caucasians. Osteoporos Int J Establ Result Coop Eur Found Osteoporos Natl Osteoporos Found USA. 2012. Jul;23(7):1867–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Cho YS, Go MJ, Kim YJ, Heo JY, Oh JH, Ban HJ, et al. A large-scale genome-wide association study of Asian populations uncovers genetic factors influencing eight quantitative traits. Nat Genet. 2009. May;41(5):527–34. [DOI] [PubMed] [Google Scholar]
  • 55.Chesi A, Mitchell JA, Kalkwarf HJ, Bradfield JP, Lappe JM, McCormack SE, et al. A trans-ethnic genome-wide association study identifies gender-specific loci influencing pediatric aBMD and BMC at the distal radius. Hum Mol Genet. 2015. Sep 1;24(17):5053–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Zhang L, Choi HJ, Estrada K, Leo PJ, Li J, Pei YF, et al. Multistage genome-wide association meta-analyses identified two new loci for bone mineral density. Hum Mol Genet. 2014. Apr 1;23(7):1923–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Waerner T, Alacakaptan M, Tamir I, Oberauer R, Gal A, Brabletz T, et al. ILEI: a cytokine essential for EMT, tumor formation, and late events in metastasis in epithelial cells. Cancer Cell. 2006. Sep;10(3):227–39. [DOI] [PubMed] [Google Scholar]
  • 58.Lahsnig C, Mikula M, Petz M, Zulehner G, Schneller D, van Zijl F, et al. ILEI requires oncogenic Ras for the epithelial to mesenchymal transition of hepatocytes and liver carcinoma progression. Oncogene. 2009. Feb 5;28(5):638–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Määttä JA, Bendre A, Laanti M, Büki KG, Rantakari P, Tervola P, et al. Fam3c modulates osteogenic cell differentiation and affects bone volume and cortical bone mineral density. BoneKEy Rep. 2016;5:787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Okpala I. The management of crisis in sickle cell disease. Eur J Haematol. 1998. Jan;60(1):1–6. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data collected in the Recipient Epidemiology and Donor Evaluation Study (REDS-III) is available through the American National Institute of Health database of Genotypes and Phenotypes (dbGaP) upon request.

RESOURCES