-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

3393 Genetic Modifiers Influencing the Development of Albuminuria in Children with Sickle Cell Anemia

Hemoglobinopathies, Excluding Thalassemia – Basic and Translational Science
Program: Oral and Poster Abstracts
Session: 113. Hemoglobinopathies, Excluding Thalassemia – Basic and Translational Science: Poster III
Monday, December 7, 2015, 6:00 PM-8:00 PM
Hall A, Level 2 (Orange County Convention Center)

Beverly A Schaefer, MD1, Jonathan M Flanagan, PhD2, Banu Aygun, MD3, Ofelia A. Alvarez, MD4, Stephen C Nelson, MD5, Kerri A Nottage, MD, MPH6, Carla W Roberts, MD7*, Thad A. Howard, MS1*, Barry R Davis, MD PhD8* and Russell E. Ware, MD, PhD9

1Cincinnati Children's Hospital Medical Center, Cincinnati, OH
2Baylor College of Medicine, Houston, TX
3Cohen Children's Medical Center of New York, New Hyde Park, NY
4Univ. of Miami School of Med., Miami, FL
5Children’s Hospitals and Clinics of Minnesota, Minneapolis, MN
6St. Jude Children's Research Hospital, Memphis, TN
7University of South Carolina, Columbia, SC
8University of Texas School of Public Health, Houston, TX
9Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH

Background:  Nephropathy in sickle cell anemia (SCA) begins in childhood and portends chronic kidney disease, renal failure, and early mortality among affected adults. Individuals of African descent have disproportionately higher rates of developing non-diabetic renal disease. Several candidate genetic variants have been identified, including some specific to African Americans, which are associated with the development of albuminuria and renal disease. The influence of genetic polymorphisms on albuminuria and elevated glomerular filtration rate (GFR) in children with SCA, both early signs of sickle nephropathy, has not been investigated.  

Objectives:  To determine the influence of selected single nucleotide polymorphisms (SNPs) on the development of albuminuria and elevated GFR in children with SCA; to identify novel genetic variants influencing albuminuria and GFR by whole exome sequencing (WES).

Design/Methods:  Genomic DNA was collected on children with SCA enrolled in two prospective studies with pre-hydroxyurea renal assessments (n=185):  (1) Hydroxyurea Study of Long-Term Effects (HUSTLE, NCT00305175, n=79) with no prior disease-modifying therapy; and (2) Transcranial Doppler (TCD) With Transfusions Changing to Hydroxyurea (TWiTCH, NCT 01425307, n=106) on chronic transfusions for abnormal TCD velocities. Albuminuria was defined as ≥30mg albumin/gm creatinine on the pre-hydroxyurea urine specimen. GFR was measured in HUSTLE using plasma DTPA (technetium 99m-labeled diethylenetriaminepentaacetic acid) clearance, and estimated GFR (eGFR) in TWiTCH based on serum creatinine. DNA samples were genotyped for 8 candidate SNPs previously associated with renal disease, using PCR-based allelic discrimination, bidirectional Sanger sequencing, and analysis of variable number tandem repeats (VNTR). Associations between albuminuria and genetic polymorphisms were tested using an additive model and correlation trend test. Linked WES data from the same patients were analyzed to identify other variants associated with albuminuria and GFR.

Results: Albuminuria was present in 13.1% of patients, including 16.3% in HUSTLE and 11.0% in TWiTCH. APOL1 genetic variants were common (G1 allele frequency = 21.9%, G2 allele = 16.0%, Table) and similar to published cohorts. Children with two APOL1 G1 alleles had an increased risk of albuminuria that approached statistical significance (p=0.053). Conversely, the presence of the DARC SNP that confers Duffy antigen expression had a protective effect (p=.038). WES analysis did not identify additional non-synonymous APOL1 variants linked with albuminuria. However, 93 non-synonymous variants were associated with DTPA GFR (p<0.001). Using patients with eGFR as a validation cohort, 7 variants in FUBP1, ZFAND4, CD163, GMFG and HLA-E maintained their association with kidney filtrative function (p<0.05). In particular, two variants in CD163, which is a macrophage scavenger receptor for hemoglobin-haptoglobin complexes, were strongly associated with increased GFR in both patient cohorts.

 

Gene

SNP

Location

MAF

Albuminuria

(Additive)

Variant frequency

Cases

Controls

APOL1  G1

rs73885319

22q12

21.9%

0.053

32.0%

20.3%

APOL1  G2

rs71785313

22q12

16.0%

0.445

10.0%

16.0%

DARC

rs2814778

1q23

13.6%

0.038

4.2%

15.7%

eNOS    4a

VNTR

7q35

31.2%

0.333*

31.5%

22.7%

eNOS

rs1799983

7q35

12.5%

0.663

14.6%

12.2%

eNOS

rs2070744

7q35

15.6%

0.299

10.4%

16.3%

CUBN

rs7918972

10p12

16.2%

0.457

12.5%

16.8%

CUBN

rs1801239

10p12

2.7%

0.199

0.0%

3.1%

Table. Candidate genes associated with microalbuminuria. All SNPs were tested with either an additive or recessive genetic model. *The eNOS VNTR was analyzed by the chi-square method.

Conclusion:  Genetic polymorphisms associated with chronic kidney disease in African American adults may influence the development of early-onset albuminuria among children with SCA, including an increased risk among children with ≥1 APOL1 G1 alleles and a decreased risk associated with the DARC SNP. Previously published eNOS and CUBN variants had no measureable effects. WES analysis suggests novel genetic variants including CD163 SNPs may influence the development of elevated GFR in children with SCA, and provide candidate genes for future research.

 

Disclosures: Off Label Use: Hydroxyurea is FDA approved for the treatment of sickle cell anemia in adults, but has not yet been approved in children. . Nottage: Janssen Pharmaceuticals: Employment . Ware: Biomedomics: Research Funding ; Bristol Myers Squibb: Research Funding ; Bayer Pharmaceuticals: Consultancy ; Eli Lilly: Other: DSMB membership .

*signifies non-member of ASH