ÁñÁ«ÊÓƵ

Object moved to here.

APOL1-Mediated Kidney Disease | Nephrology | JAMA | ÁñÁ«ÊÓƵ Network

ÁñÁ«ÊÓƵ

[Skip to Navigation]
Sign In
JAMA Insights
´¡±è°ù¾±±ôÌý25, 2024

APOL1-Mediated Kidney Disease

Author Affiliations
  • 1Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
  • 2Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
JAMA. Published online April 25, 2024. doi:10.1001/jama.2024.2667

Introduction

According to the 2020 Annual Data Report, 30% of patients with end-stage kidney disease (ESKD) in the US are Black individuals, although they comprise only 13% to 14% of the population. In 2010, researchers identified 2 common variants of the APOL1 (apolipoprotein L1) gene (G1 and G2), which account for much of the excess nondiabetic chronic kidney disease (CKD) risk among Black individuals in the US.1,2 This review explains the evolutionary origin of APOL1 high-risk genetic variants, defines APOL1-mediated kidney disease (AMKD), and discusses recommendations for AMKD screening and management.

APOL1 and Trypanosomiasis

The wild-type APOL1 gene (G0) was identified in 2003 as the component of human serum that confers resistance to Trypanosoma brucei, the cause of African trypanosomiasis (sleeping sickness). The APOL1 gene encodes an innate immune pore-forming protein that inserts into the trypanosome’s lysosomal membrane, inducing osmotic stress and ultimately lysing the trypanosome. Approximately 10 000 years ago, 2 novel T brucei strains emerged in Africa that were resistant to the G0 immune response and therefore caused African sleeping sickness. The G1 and G2 variants of the APOL1 gene restored immune protection against the novel T brucei strains and underwent positive selection in West Africa.

Genetics of APOL1

The risk of kidney disease associated with the APOL1 G1 and G2 alleles follows a recessive inheritance pattern. Individuals who are heterozygous for a G1 or G2 variant (G1/G0 or G2/G0) are resistant to African sleeping sickness but do not have increased risk of kidney disease. However, individuals with 2 alleles of APOL1 G1 or G2 variants (G1/G1, G2/G2, or G1/G2) have an increased risk of kidney disease and are collectively known as the APOL1 high-risk genotype.1

The allele frequencies of G1 and G2 vary across different populations worldwide. The prevalence of these high-risk alleles is highest in West African populations. In different populations in Nigeria, G1 allele frequency has been reported to be 37% to 45%, and G2 frequency has been estimated to be 7.5% to 17%.2 Among Black individuals in the US, the G1 allele frequency is 22% and G2 allele frequency is 13%.2 Approximately 13% of Black individuals in the US (more than 5 million people) have a high-risk APOL1 genotype.3 High-risk alleles are also found frequently in sub-Saharan African, Western African, Caribbean, Central American, and South American populations.4

APOL1-Mediated Kidney Disease

Recent experimental evidence suggests that the G1 and G2 variants may cause kidney injury by increased transport of monovalent cations—similar to the mechanism by which G1 and G2 provide resistance to T brucei.5,6

Individuals with a high-risk APOL1 genotype have been estimated to have a 15% to 30% lifetime risk of developing ESKD.3 The basis of this variable penetrance is currently unknown. AMKD is diagnosed when an individual with a high-risk APOL1 genotype develops nondiabetic kidney disease. There is no established glomerular filtration rate (GFR) cutoff to diagnose AMKD. Proteinuria (urine albumin to creatinine ratio >30 mg/g) is often present but is not a requirement for the diagnosis of AMKD.

Approximately 75% of Black individuals in the US with focal segmental glomerulosclerosis (FSGS) have been estimated to have a high-risk APOL1 genotype.4 Black individuals in the US with a high-risk APOL1 genotype are also more likely to develop hypertension-attributed ESKD, lupus nephritis–attributed ESKD, FSGS, and HIV-associated nephropathy compared with Black individuals who have a low-risk APOL1 genotype (Figure). A recent study reported that 25.8% of patients with COVID-19–associated kidney injury had collapsing glomerulopathy, and 91.7% of these patients had a high-risk APOL1 genotype.7

Figure.  APOL1-Mediated Kidney Disease in Black Individuals in the US

Bubble plot shows the odds ratio of APOL1-mediated kidney disease (AMKD) in Black individuals in the US with a high-risk APOL1 genotype compared with Black individuals with a low-risk APOL1 genotype. The area of each bubble represents the population attributable risk—the proportion of the incidence of each type of kidney disease that is attributed to high-risk APOL1 genotype.

Odds ratios from Friedman and Pollak.3 Estimated number of affected individuals was extrapolated from . Population attributable risk was calculated from odds ratio and the frequency of high-risk APOL1 genotype in the Black US population.

CKD indicates chronic kidney disease; HTN-ESKD, hypertension-attributed end-stage kidney disease; and FSGS, focal segmental glomerulosclerosis.

Screening for High-Risk APOL1 Genotype and AMKD

Screening of asymptomatic individuals in the US for high-risk APOL1 genotype is not currently recommended. Individuals identified as Black in clinical and epidemiological studies in the US represent a genetically diverse group, and the risk of a genetic variant is not evenly distributed in this population. However, individuals with recent West African heritage who have nondiabetic CKD, proteinuria, or a family history of kidney disease may be offered APOL1 screening. All individuals should receive genetic counseling before and after APOL1 genetic testing and be advised about potential for increased cost of health insurance that may be associated with detection of a high-risk APOL1 genotype. When individuals are noted to have a high-risk genotype, physicians may consider more frequent screening for reduced GFR and proteinuria to identify AMKD at an early stage.

Prospective kidney donors with recent West African heritage may be encouraged to undergo APOL1 genetic testing because preliminary data suggest that donors with a high-risk APOL1 genotype have a greater decrease in postdonation kidney function and are more likely to develop ESKD. A study of 136 Black living kidney donors who were evaluated at a median of 12 years after donation reported that compared with donors with a low-risk genotype, those with a high-risk APOL1 genotype had lower eGFR (mean [SD], 57 [18] vs 67 [15] mL/min per 1.73 m2; P = .02) and faster decline in eGFR after adjustment for predonation eGFR (1.19 [95% CI, 0-2.3] vs 0.4 [95% CI, 0.1-0.7 mL/min per 1.73 m2 per year, P = .02).8

A study of 478 deceased donor kidney transplants from Black donors reported that after adjusted analysis, transplanted kidneys from deceased donors with a high-risk APOL1 genotype had shorter allograft survival compared with a low-risk APOL1 genotype (absolute rates not reported; hazard ratio, 2.05; P = 3 ×â¶Ä‰10−4) but no difference in kidney recipient survival.9 A large ongoing National Institutes of Health–funded study, APOLLO () is evaluating the effects of APOL1 variants on outcomes for both kidney transplant donors and recipients.

Early identification of patients with a high-risk APOL1 genotype may be associated with beneficial lifestyle modifications and decreased blood pressure. A recent study10 randomized 2050 US adults with self-identified African ancestry and hypertension without CKD to immediate vs delayed reporting (12 months later) of APOL1 test results. At 3 months, patients identified with a high-risk APOL1 genotype had a significantly greater decrease in mean systolic blood pressure (6 mm Hg) compared with low-risk APOL1 genotypes (3 mm Hg) and the control group with delayed reporting (3 mm Hg). At 12 months, compared with individuals who had a low-risk genotype, patients with a high-risk APOL1 genotype self-reported significant improvements in diet and exercise (129/218 [59%] vs 547/1468 [37%]; P < .01) and increased use of blood pressure medication (21/218 [10%] vs 68/1468 [5%]; P = .005).

Treatment Options for AMKD

Although there is currently no approved treatment for AMKD, multiple therapeutic trials are ongoing. The JUSTICE trial () is testing baricitinib, a JAK-STAT inhibitor that reduces APOL1 gene expression. A phase 2/3 clinical trial () is evaluating the effect of inaxaplin, a small molecule APOL1 inhibitor, on proteinuria and the rate of GFR decline among individuals with a high-risk APOL1 genotype and proteinuric kidney disease.

Back to top
Article Information

Corresponding Author: Opeyemi A. Olabisi, MD, PhD, Duke Molecular Physiology Institute, Division of Nephrology, Department of Medicine, Duke University School of Medicine, 300 N Duke St, #50-104, Durham, NC 27701 (Opeyemi.Olabisi@duke.edu).

Published Online: April 25, 2024. doi:10.1001/jama.2024.2667

Conflict of Interest Disclosures: Dr Olabisi reported grants from NIH during the conduct of the study. No other disclosures were reported.

References
1.
Genovese  G, Friedman  DJ, Ross  MD,  et al.  Association of trypanolytic ApoL1 variants with kidney disease in African Americans.  Ìý³§³¦¾±±ð²Ô³¦±ð. 2010;329(5993):841-845. doi:
2.
Kopp  JB, Nelson  GW, Sampath  K,  et al.  APOL1 genetic variants in focal segmental glomerulosclerosis and HIV-associated nephropathy.   J Am Soc Nephrol. 2011;22(11):2129-2137. doi:
3.
Friedman  DJ, Pollak  MR.  APOL1 nephropathy: from genetics to clinical applications.   Clin J Am Soc Nephrol. 2021;16(2):294-303. doi:
4.
Nadkarni  GN, Gignoux  CR, Sorokin  EP,  et al.  Worldwide frequencies of APOL1 renal risk ±¹²¹°ù¾±²¹²Ô³Ù²õ.Ìý  N Engl J Med. 2018;379(26):2571-2572. doi:
5.
Egbuna  O, Zimmerman  B, Manos  G,  et al; VX19-147-101 Study Group.  Inaxaplin for proteinuric kidney disease in persons with two APOL1 ±¹²¹°ù¾±²¹²Ô³Ù²õ.Ìý  N Engl J Med. 2023;388(11):969-979. doi:
6.
Datta  S, Antonio  BM, Zahler  NH,  et al.  APOL1-mediated monovalent cation transport contributes to APOL1-mediated podocytopathy in kidney disease.   J Clin Invest. Published online January 16, 2024:e172262. doi:
7.
May  RM, Cassol  C, Hannoudi  A,  et al.  A multi-center retrospective cohort study DEFINES the spectrum of kidney pathology in Coronavirus 2019 disease (COVID-19).   Kidney Int. 2021;100(6):1303-1315. doi:
8.
Doshi  MD, Ortigosa-Goggins  M, Garg  AX,  et al.  APOL1 genotype and renal function of black living donors.   J Am Soc Nephrol. 2018;29(4):1309-1316. doi:
9.
Freedman  BI, Pastan  SO, Israni  AK,  et al.  APOL1 genotype and kidney transplantation outcomes from deceased African American donors.  Ìý°Õ°ù²¹²Ô²õ±è±ô²¹²Ô³Ù²¹³Ù¾±´Ç²Ô. 2016;100(1):194-202. doi:
10.
Nadkarni  GN, Fei  K, Ramos  MA,  et al.  Effects of testing and disclosing ancestry-specific genetic risk for kidney failure on patients and health care professionals: a randomized clinical trial.   ÁñÁ«ÊÓƵ Netw Open. 2022;5(3):e221048. doi:
×