Abstract
Apolipoprotein L1 (APOL1) high-risk variants confer an increased risk for the development and progression of kidney disease among individuals of recent African ancestry. Over the past several years, significant progress has been made in understanding the pathogenesis of APOL1-mediated kidney diseases (AMKD), including genetic regulation, environmental interactions, immunomodulatory, proinflammatory and apoptotic signaling processes, as well as the complex role of APOL1 as an ion channel. Collectively, these findings have paved the way for novel therapeutic strategies to mitigate APOL1-mediated kidney injury. Precision medicine approaches are being developed to identify subgroups of AMKD patients who may benefit from these targeted interventions, fueling hope for improved clinical outcomes. This review summarizes key mechanistic insights in the pathogenesis of AMKD, emergent therapies, and discusses future challenges.
Keywords: APOL1, cytokine, glomerular, interferon, podocyte, proteinuria
The APOL1 gene resides on chromosome 22 and is part of the APOL gene family, present in humans and certain primates. Although the lack of endogenous APOL1 expression in commonly used mouse and rat models has been a barrier to disease modeling and drug discovery efforts, significant progress has been made since the original seminal work by Pollak et al. in 2010.1 G1 and G2 risk alleles define the APOL1 risk genotypes (G1/G1, G2/G2, G1/G2), whereas the “non-risk” APOL1 allele is referred to as G0. The APOL1 G1 allele consists of 2 missense variants in high linkage disequilibrium (APOL1 p.S342G, rs73885319; APOL1 p.I384M, rs6091014), whereas the G2 allele is a 2–amino acid deletion (APOL1 p.delN388/Y389, rs60910145). G1 and G2 alleles exhibit a notably high prevalence in populations of recent African ancestry, likely reflecting the influence of natural selection in West and Central Africa to protect against African trypanosomiasis known as “sleeping sickness.” As a result of the slave trade and other migration patterns, APOL1 high-risk variants are widely disseminated, especially in the Americas.2 High-risk variants of APOL1 are found in approximately 10% to 15% of African Americans.3 In African Americans, carrying 2 high-risk alleles confers a 1.49-fold increased risk of chronic kidney disease and a 1.88-fold risk of end-stage kidney disease compared to those with 0 or 1 risk allele.4
AMKD encompasses diverse clinical manifestations characterized by kidney function decline, variable proteinuria levels, and hypertension. A recent large Phenome-Wide Association Study confirmed the association of APOL1 with primarily kidney and kidney-associated pathologies.5 Some of the most frequent conditions studied within the spectrum of AMKD include focal segmental glomerulosclerosis (FSGS), to virus-related forms such as HIV-associated nephropathy and COVID-19-associated nephropathy, and the syndrome of solidified or diffuse glomerulosclerosis with low level proteinuria (often mislabeled arterionephrosclerosis or hypertensive nephropathy).6, 7, 8, 9 The role of APOL1 in conditions such as preeclampsia, sickle cell disease and some autoimmune diseases is not fully understood. Improved knowledge of APOL1 biology and its therapeutic targeting offers a unique opportunity to treat clinical entities of important public health relevance. In this review, we summarize mechanistic pathways implicated in AMKD and provide an overview of promising emerging therapeutic options.
APOL1 Expression and Upstream Regulation
The mode of inheritance in AMKD pathogenesis has been a topic of controversy.10 Although most autosomal recessive diseases are characterized by loss-of-function mutations, APOL1 is atypical, having a recessive model of inheritance for chronic kidney disease and apparent gain-of-function characteristics.11 Recent data investigating the use of small molecule inhibitors and antisense oligonucleotides to reduce APOL1 expression and its downstream effects further support the gain-of-function model.12,13 The fact that a functional APOL1 gene is found only in some African primate species and that individuals lacking APOL1 can still maintain normal kidney function suggests that this gene is not essential for normal kidney development or functioning.14,15 In theory, inhibiting APOL1 toxicity should ameliorate kidney disease without major adverse effects. However, caution is warranted until the effects of APOL1 expression in other systems such as the endothelium, immune cells, and hepatocytes are fully understood. Furthermore, reducing APOL1 expression may result in deleterious implications in areas endemic for trypanosomiasis.
The expression of APOL1 is tightly controlled by a complex interplay between genetic and epigenetic factors. Numerous immune and inflammatory pathways have been identified that upregulate APOL1 expression, including the interferon family, the most extensively studied, interleukin-1β and proteins from the toll-like receptor family.16, 17, 18 The APOL1 promoter contains regulatory elements that interact with several transcription factors, including STAT2, STAT3, and interferon regulating factors 1, 2, and 3. Interferons and toll-like receptor agonists have been shown to increase APOL1 expression by up to 200-fold.16 Comparative promoter analyses have revealed differential effects on APOL1 expression. Toll-like receptor 3 activation exerts a more pronounced effect than toll-like receptor 4, whereas interferon γ has a greater impact on APOL1 expression compared to interferon β and α. These findings support the hypothesis that APOL1 is a cellular immune response gene.
The Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway, a highly conserved pathway that regulates immunological and adaptative responses, has been well studied in the context of APOL1 regulation. This pathway consists of 4 JAK members (JAK1–JAK3 and receptor tyrosine kinase 2) and 7 STAT members (STAT1–STAT4, STAT5a, STAT5b, and STAT6), and it is essential for cell maintenance, immune fitness, and tissue repair.19, 20, 21 Extensive activation of the JAK/STAT pathway has been recognized as a critical step in kidney diseases such as diabetic nephropathy and autosomal dominant polycystic kidney disease.22, 23, 24, 25, 26 Most recently, COVID-19-associated nephropathy has been identified as a condition where COVID-19-induced cytokines (predominantly interleukin-6, interleukin-1β, and interleukin-18) are sufficient to activate the JAK/STAT signaling pathway.26 Interestingly, the amplification of APOL1 expression in human podocytes and glomerular endothelial cells was seen even when interferon levels were negligible (indicating the presence of interferon-independent mechanisms), but still resulting in podocyte injury and loss. This was rescued by baricitinib (JAK inhibitor), providing a rationale for its use as a therapeutic agent in APOL1-induced cellular injury.
Host genetic factors may also affect the expression of APOL1. For example, gain-of-function mutations in TMEM173, the gene encoding stimulator of interferon genes (STING) can exacerbate the production of interferon, a condition that has been recently described as STING-associated vasculopathy with onset in infancy. Type I IFN triggers a positive feedback loop leading to activation of JAK1 and STAT1/STAT 2, and transcription of proinflammatory IFN-stimulated genes. The recently published case of a patient with APOL1 G1 and G2 risk alleles with a high interferon state due to STING-associated vasculopathy with onset in infancy, illustrated a human model where a high interferon state can lead to collapsing glomerulopathy.27
Several studies continue to explore the role of epigenetic factors, copy number variants and SNPs in APOL1 nephropathy.28 Genetic modifiers such as SMOC2, DEF1B, UBD, NUDT7, and GSTB1 have also been identified.29, 30, 31, 32, 33 In addition, environmental factors such as air pollution have been implicated in the development of AMKD in patients carrying APOL1 risk variants, probably due to cellular stress mechanisms.34, 35, 36, 37 In sum, AMKD has diverse clinical manifestations and it is likely that complex epistatic and environmental interactions result in differential pathological cellular programming.
APOL1 Mechanisms of Podocyte Injury
APOL1 as an Ion Channel
One of the most widely accepted proposed mechanisms of APOL1-mediated injury centers on its role as an ion channel.38,39 Conflicting findings have been reported regarding the nature of these channels, especially their anionic or cationic activity. This is related to variability in localization (cellular vs. organelle), model studied (trypanosomal activity vs. podocyte-specific toxicity), and structural or biochemical factors, derived from pH environment, among others.40, 41, 42 Subcellular localization of APOL1 and/or affinity for binding partners have been described but there is a lack of consensus across models.43, 44, 45
Several studies support the hypothesis that APOL1 forms distinct anion-selective pores in unilamellar vesicular membranes, promoting chloride influx facilitated by the initial influx of extracellular sodium following its concentration gradient.46,47 This osmotic imbalance leads to the passive entry of water into the cell, resulting in cell swelling and trypanosome lysis. However, other studies support the cationic nature of the channel, suggesting that trypanosome lysis requires an acidic pH for the initial steps to allow APOL-1 insertion into vacuolar lipid bilayers, to be subsequently transported to the plasma membrane, where it is exposed to a nonacidic pH allowing APOL1 to open pH-sensitive cationic channels, depolarizing the membrane and killing the trypanosome.48 Another publication supports this view demonstrating that mammalian cells expressing APOL1 risk variants exhibit increased nonselective cation permeability, resulting in a net efflux of intracellular potassium through the plasma membrane, thereby inducing cell damage through the activation of stress-activated protein kinases, p38, mitogen-activated mitogen kinase, and JNK.49 These seemingly discordant findings may be reconciled by the proposal that APOL1 ion-channel selectivity is pH-switchable. At pH of 5, APOL1 may promote chloride permeability through anionic channels, whereas at neutral pH, it facilitates potassium permeability.50 Collectively, the body of evidence suggests that APOL1 channel activity depends on 3 factors that allow APOL1 to associate with pore-forming vesicles: delicate pH fine tuning, presence of negatively charged phospholipids in vesicle membranes, and low ionic strength.
APOL1-Associated Mitochondrial Stress
APOL1-induced mitochondrial dysfunction is believed to contribute to AMKD through various mechanisms.51 The induction of APOL1 G1 and G2 expression results in a significant reduction in the maximum oxygen consumption rate and respiratory reserve capacity compared to cells expressing the APOL1 G0 variant.52 After being transported to the mitochondria by incompletely understood mechanisms, APOL1 risk variants form higher-order oligomers within the mitochondria and activate pore opening, resulting in cell toxicity by increasing fatty acid oxidation, and decreased redox homeostasis, disruption of the mitochondrial membrane potential, and cell toxicity.45 Another possible mechanism underlying APOL1-induced mitochondrial dysfunction is the increase in mitochondrial fragmentation (fission), as opposed to fusion. Physiologically, fission helps segregate the most severely damaged mitochondria to preserve the overall health of the mitochondrial network. However, when G1 and G2 variant-induced mitochondrial fission cannot be adequately compensated through mitophagy (possibly due to defective intracellular trafficking), cell death mechanisms are activated.53, 54, 55 Overexpression of APOL1 G1 and G2 in HEK293 cells promotes mitochondrial fragmentation thought active DRP1. The inhibition of mitochondrial fission using the DRP1 inhibitor, Mdivi-129, appears to preserve mitochondrial morphology, resulting in fewer fragmented mitochondria, and treatment restores cell viability in a dose-dependent manner.56,57
APOL1-Associated Endoplasmic Reticulum and Lysosomal Stress
Other organelles that have been implicated in APOL1-induced cell damage are lysosomes and the endoplasmic reticulum (ER). APOL1 risk variants are associated with increased lysosomal permeability and compromised endolysosomal trafficking, suggesting their involvement in cellular perturbations.58 Insertion of APOL1 into the lysosomal membrane triggers ion flux into the organelle, leading to osmotic damage and death.46 Notably, APOL1 causes lysosomal dysfunction in cultured human renal cells as well as in parasites. The G1 and G2 variants of APOL1 decrease the number of lysosomes in podocytes, leading to leakage of lysosomal enzymes into the cytoplasm.59,60
Recent studies have shown that APOL1 expression is localized to the ER. In their study using cultured HEK293 cells, Chun et al.61 uncovered a distinct pattern of localization for APOL1 risk variants, primarily within the ER, whereas wild type APOL1 localization predominantly to lipid droplets. Notably, when cells were subjected to treatments promoting lipid droplet formation, a notable shift in the localization of G1 and G2 variants occurred, moving from the ER to the lipid droplets, reducing autophagic flux and cytotoxicity. In addition, factors such as tissue hypoxia,62 oxidative stress, and chronic inflammation can further amplify ER stress, exacerbating kidney disease progression among those with high-risk APOL1 variants. Further research efforts are warranted to unravel the complex mechanisms of ER stress, its modulation by APOL1 variants, and its impact on disease pathogenesis. Consequently, therapeutic approaches targeting ER stress that hold promise for other clinical applications (e.g., cancer and metabolic diseases), could be an important opportunity for drug-repurposing.63
Inflammation and APOL1
As previously described, inflammatory mechanisms induce the expression of APOL1. However, these mechanisms also appear to be involved in the induction and maintenance of APOL1-mediated damage. STING is an adjuvant protein on the ER that recognizes the cyclic dinucleotides generated by cyclic GMP-AMP synthase, that recognizes foreign and host DNA (incremented in states of cellular stress).64 Once cyclic GMP-AMP synthase senses cytoplasmic dsDNA, it converts GTP and ATP into 2′,3′-cGAMP, which binds and activates STING, promoting the secretion of type I interferons and various proinflammatory cytokines.65 However, when cyclic GMP-AMP synthase-STING pathway activation exceeds a certain threshold, STING induces necroptosis, apoptosis, and lysosome fragmentation via NOD-like receptor pyrin domain-containing 3 inflammasome-dependent pyroptosis.66,67 The NOD-like receptor pyrin domain-containing 3 inflammasome is a multiprotein complex that forms part of the innate immune system and activates multiple inflammatory proteins such as interleukin 1β, interleukin-18, and the pore-forming gasdermin D, leading to cellular osmotic imbalance and the release of proinflammatory intracellular contents.66 Recent studies have identified the STING-NOD-like receptor pyrin domain-containing 3 pathway as a pivotal mechanism underlying APOL1-induced cytotoxicity.68 Emerging small-molecule-based strategies and biologics to therapeutically target cyclic GMP-AMP synthase-STING and NOD-like receptor pyrin domain-containing 3 signaling are gaining interest in several models of chronic inflammation (e.g., cancer and autoimmune disorders), and these findings support its potential value in the treatment of AMKD.
Other Mechanisms
APOL1-mediated cellular injury depends on complex molecular mechanisms that are intricately linked. In addition to those mentioned previously, other mechanisms implicated in AMKD that have been suggested include activation of protein kinase R,69 which appears during viral infections and inhibits protein synthesis, increased autophagic cell death (mediated by the BCL2- homology 3 domain within the pore-forming domain of APOL1),70 or the alteration of the ubiquitin-proteasome system, which reduces ubiquitin levels prolonging the intracellular retention of proteins (including APOL1 itself).31 In addition, APOL1 G1 and G2 variants have been shown to have high affinity for suPAR activated αvβ3 integrin on podocytes in the progression of chronic kidney disease.71 These and the aforementioned mechanisms are summarized in Figure 1.
Figure 1.
Purported mechanisms of APOL1-induced kidney injury. PKR, protein kinase R.
Emerging Therapies in AMKD
Drug discovery has traditionally relied on well-characterized disease targets, high-throughput screening methods to test large compound libraries, and prior experimental data.72,73 Nonetheless, the emergence of novel technologies such as genomics, proteomics, and bioinformatics to generate and analyze large-scale data sets, enabled the identification of new mechanistic pathways and therefore more precise options such as peptide-based inhibitors, oligonucleotides, or gene therapies.74, 75, 76 These modalities have been extensively described in other fields such as oncology and rheumatology. The rapid development of drug repurposing methods, systems biology, and artificial intelligence in nephrology is supporting the emergence of novel approaches to tackle AMKD.77, 78, 79 Given the number of potential mechanisms that contribute to APOL1-mediated renal injury, it is not surprising that different approaches are currently being explored. A summary of phase 1 to 3 clinical trials for AMKD since its discovery is presented in Table 1. A brief discussion of these emerging agents (Figure 2) will be discussed below.
Table 1.
Recent and ongoing APOL1 therapeutic trials
| NCT number | Drug | Mechanism of action | Status | Phase | Completion |
|---|---|---|---|---|---|
| NCT04340362 | VX-147 | APOL1 channel blocker (small molecule inhibitor) | Completed | Phase 2 | December 2021 |
| NCT05312879 | Recruiting | Phase 2/3 | June 2026 | ||
| NCT05324410 | VX-840 | APOL1 channel blocker (small molecule inhibitor) | Completed | Phase 1 | November 2022 |
| NCT04269031 | AZD2373 | APOL1 antisense oligonucleotide | Completed | Phase 1 | August 2021 |
| NCT05351047 | Active, not recruiting | Phase 1 | July 2023 | ||
| NCT05237388 | Baricitinib | Janus Kinase-STAT Inhibition | Recruiting | Phase 2 | March 2026 |
APOL1, apolipoprotein L1; STAT, signal transducer and activator of transcription.
Search of clinicaltrials.gov was performed on July 19, 2023.
Figure 2.
Mechanism of action of current APOL1 therapeutic agents in development.
APOL1 Small Molecule Inhibitors
Small molecules are a class of pharmacological agents that exhibit high specificity and potency in targeting enzymes or protein-protein interactions of disease-relevant pathways.78 With their relatively low molecular weight and favorable physicochemical properties, small molecule inhibitors possess the ability to penetrate cellular membranes and enable access to intracellular components. These inherent characteristics offer significant advantages for therapeutic intervention by mitigating off-target effects and minimizing toxicity to normal cells. In addition, harnessing the modulatory potential of small molecule inhibitors holds great promise in advancing personalized medicine, particularly in the context of AMKD, because individual patients may exhibit distinct molecular drivers of disease within the spectrum of AMKD.26,49,53,68 Moreover, the oral bioavailability of many small molecule inhibitors enables convenient administration.
A novel small molecule inhibitor on APOL1 channel called VX-147 or inaxaplin demonstrated promising results in reducing proteinuria in patients with APOL1-associated FSGS in a recently published phase 2a study.12 After demonstration of efficacy in preclinical models (a reduction of cationic influx in vitro and a reduction of proteinuria in a transgenic APOL1 mouse model), inaxaplin was administered during 13 weeks to 16 participants who had 3 APOL1 high-risk variants, biopsy-proven FSGS, and estimated glomerular filtration rate ≥27 ml/min per 1.73 m2. Among the 13 participants who were adherent to the treatment threshold, the mean change from the baseline urinary protein-to-creatinine ratio at week 13 was −47.6% (95% confidence interval, −60.0 to −31.3). Furthermore, in an analysis that included all 16 participants regardless of adherence to therapy, reductions were similar to those in the primary analysis in all but 1 participant. The drug showed a substantial decrease in proteinuria, even in patients receiving standard-of-care treatment. However, there are some limitations to note. In addition to statistical limitations and biases inherent to the single arm, small sample study design, most patients had FSGS with subnephrotic range proteinuria, and secondary FSGS could not be ruled out. Furthermore, baseline and concomitant therapy for FSGS were variable during the treatment phase and it remains unclear whether longer duration of inaxaplin would persistently suppress proteinuria. A larger phase 2/3 inaxaplin AMKD study (NCT05312879), not limited to FSGS, is currently actively enrolling. In addition, another small molecule inhibitor (VX-840) is currently being studied by the same sponsor (phase 1 completed in November 2022).
Recently, small molecule inhibitor, MZ-301 has been developed to potentially block APOL1 electrophysiological currents in response to a voltage ramp in HEK293 cells expressing APOL1 G2.80 APOL1-mediated ion currents measured in HEK293 cells were noted to decrease in the presence of incremental doses of MZ-301, eventually rescuing them from APOL1-mediated cytotoxicity. Furthermore, MZ-301 was described to inhibit APOL1-dependent cytotoxicity in vitro in human immortalized podocytes, which translated into urine albumin-to-creatinine ratio reductions in APOL1 G2 mutant mice.
Antisense Oligonucleotides
Oligonucleotide therapeutics, such as those based on antisense oligonucleotides (ASOs), small interfering RNA, microRNA, aptamers, and decoys, are promising agents that have gained importance during the past decades in nephrology and other fields.81, 82, 83 APOL1 ASOs are oligonucleotide analogs that modify expression of specific RNAs and can alter protein synthesis. ASOs bind to select mRNA sequences and can cause RNase H1-mediated degradation (ceasing synthesis of the protein), splicing defects, or interfere with gene expression. In a recent study, a generation 2.5 APOL1 ASO (IONIS-APOL1Rx) was selected as the APOL1 clinical candidate based on its consistent and potent activity in vitro as well as in vivo in genomic APOL1-transgenic mice. Subcutaneous administration of IONIS-APOL1Rx to APOL1 G1-transgenic mice resulted in dose-dependent reductions in kidney and liver APOL1 mRNA, preventing dose-dependent interferon-induced proteinuria.84 The agent was used in a first-in-human, single ascending dose, phase I study (NCT04269031), to evaluate the safety and assess the pharmacokinetics of escalating single doses of a subcutaneously administered ASO (ION532, also known as AZD2373), with results pending. Another phase I study (NCT05351047) is ongoing.
JAK/STAT Pathway Blockade
As described above, JAK/STAT plays a critical role in activating proinflammatory cell programs and its inhibition could efficiently decrease APOL1-associated cellular toxicity, which has been tested in preclinical models with promising results. A phase 2 trial (JUSTICE, NCT05237388) is currently recruiting to evaluate the efficacy of baricitinib, a JAK inhibitor approved for the treatment of rheumatoid arthritis and alopecia areata, in patients with AMKD. STAT3 inhibition using next-generation ASOs, including 2.5 ASO (e.g., AZD9150) has been explored for other clinical indications. STAT3 binds mRNA and silences gene expression through blocking translation or recruiting RNase H enzymes, which degrade the DNA-RNA heteroduplex.19,20 AZD9150 is currently being studied in leukemia and lymphoma due to its proapoptotic and cell regulatory effects.85,86
Future Directions and Considerations
Extensive studies have shed light on cellular pathways activated by APOL1 risk variants in cellular, animal, and human studies; however, several gaps remain. Evidence that less than 30% of individuals with 2 high-risk APOL1 variants develop AMKD warrants careful exploration to further define “second hits” and clarify patient populations that should undergo genetic screening. This will influence ongoing initiatives to potentially reduce adverse kidney and transplant outcomes, support the development of noninvasive biomarkers that could potentially anticipate the onset of kidney disease, provide risk-stratification algorithms and advance precision-based therapeutic approaches.
There are still other unresolved questions in developing APOL1 therapeutics. There is uncertainty regarding the impact of circulating APOL1 on the kidneys and consequently a lack of clarity regarding the relative efficacy of reducing systemic APOL1 levels versus inhibiting the function of mutant APOL1 protein. The multiple upstream regulatory and downstream pathogenic signaling cascades may also require potentially divergent therapeutic approaches in certain AMKD subpopulations. Safety will be a concern, especially in regions endemic for trypanosomiasis and where prolonged therapy may be required. Small molecule inhibitors can lead to off-target effects and prolonged genetic alterations. Advances are also required in improving kidney-specific drug delivery to reduce the potential for off-target effects of APOL1 inhibitors.
In summary, significant progress has been made in understanding the cellular injury mechanisms of APOL1 risk variants and the development of new therapies for AMKD. More work will be needed in molecular subphenotyping, precision-based targeted approaches, and careful investigation of the efficacy and safety profile of emerging therapies.
Disclosure
KNC reports consulting fees from Travere, Goldfinch, Chinook, ANI, and Aurinia and funds to his department for being a site principal investigator for studies sponsored by Vertex and Travere outside the submitted work. GVR and MC declare no competing interests.
References
- 1.Genovese G., Friedman D.J., Ross M.D., et al. Association of trypanolytic ApoL1 variants with kidney disease in African Americans. Science. 2010;329:841–845. doi: 10.1126/science.1193032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Nadkarni G.N., Gignoux C.R., Sorokin E.P., et al. Worldwide frequencies of APOL1 renal risk variants. N Engl J Med. 2018;379:2571–2572. doi: 10.1056/NEJMc1800748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Friedman D.J., Kozlitina J., Genovese G., Jog P., Pollak M.R. Population-based risk assessment of APOL1 on renal disease. J Am Soc Nephrol. 2011;22:2098–2105. doi: 10.1681/ASN.2011050519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Foster M.C., Coresh J., Fornage M., et al. APOL1 variants associate with increased risk of CKD among African Americans. J Am Soc Nephrol. 2013;24:1484–1491. doi: 10.1681/ASN.2013010113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bajaj A., Ihegword A., Qiu C., et al. Phenome-wide association analysis suggests the APOL1 linked disease spectrum primarily drives kidney-specific pathways. Kidney Int. 2020;97:1032–1041. doi: 10.1016/j.kint.2020.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Yusuf A.A., Govender M.A., Brandenburg J.T., Winkler C.A. Kidney disease and APOL1. Hum Mol Genet. 2021;30:R129–R137. doi: 10.1093/hmg/ddab024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wu H., Larsen C.P., Hernandez-Arroyo C.F., et al. AKI and collapsing glomerulopathy associated with COVID-19 and APOL 1 high-risk genotype. J Am Soc Nephrol. 2020;31:1688–1695. doi: 10.1681/ASN.2020050558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Pollak M.R., Genovese G., Friedman D.J. APOL1 and kidney disease. Curr Opin Nephrol Hypertens. 2012;21:179–182. doi: 10.1097/MNH.0b013e32835012ab. [DOI] [PubMed] [Google Scholar]
- 9.Friedman D.J., Pollak M.R. APOL1 nephropathy: from genetics to clinical applications. Clin J Am Soc Nephrol. 2021;16:294–303. doi: 10.2215/CJN.15161219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bruggeman L.A., O’Toole J.F., Sedor J.R. APOL1 polymorphisms and kidney disease: loss-of-function or gain-of-function? Am J Physiol Ren Physiol. 2019;316:F1–F8. doi: 10.1152/ajprenal.00426.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Dummer P.D., Limou S., Rosenberg A.Z., et al. APOL1 kidney disease risk variants: an evolving landscape. Semin Nephrol. 2015;35:222–236. doi: 10.1016/j.semnephrol.2015.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Egbuna O., Zimmerman B., Manos G., et al. Inaxaplin for proteinuric kidney disease in persons with two APOL1 variants. N Engl J Med. 2023;388:969–979. doi: 10.1056/NEJMoa2202396. [DOI] [PubMed] [Google Scholar]
- 13.Friedman D.J., Ma L., Freedman B.I. Treatment potential in APOL1-associated nephropathy. Curr Opin Nephrol Hypertens. 2022;31:442–448. doi: 10.1097/MNH.0000000000000816. [DOI] [PubMed] [Google Scholar]
- 14.Johnstone D.B., Shegokar V., Nihalani D., et al. APOL1 null alleles from a rural village in India do not correlate with glomerulosclerosis. PLoS One. 2012;7 doi: 10.1371/journal.pone.0051546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Vanhollebeke B., Truc P., Poelvoorde P., et al. Human Trypanosoma evansi infection linked to a lack of apolipoprotein L-I. N Engl J Med. 2006;355:2752–2756. doi: 10.1056/NEJMoa063265. [DOI] [PubMed] [Google Scholar]
- 16.Nichols B., Jog P., Lee J.H., et al. Innate immunity pathways regulate the nephropathy gene apolipoprotein L1. Kidney Int. 2015;87:332–342. doi: 10.1038/ki.2014.270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wang D.P., Yu Z.X., He Z.C., et al. Apolipoprotein L1 is transcriptionally regulated by SP1, IRF1 and IRF2 in hepatoma cells. FEBS Lett. 2020;594:3108–3121. doi: 10.1002/1873-3468.13887. [DOI] [PubMed] [Google Scholar]
- 18.O’Toole J.F., Bruggeman L.A., Madhavan S., Sedor J.R. The cell biology of APOL1. Semin Nephrol. 2017;37:538–545. doi: 10.1016/j.semnephrol.2017.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Pace J., Paladugu P., Das B., He J.C., Mallipattu S.K. Targeting STAT3 signaling in kidney disease. Am J Physiol Ren Physiol. 2019;316:F1151–F1161. doi: 10.1152/ajprenal.00034.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hu X., Li J., Fu M., Zhao X., Wang W. The JAK/STAT signaling pathway: from bench to clinic. Signal Transduct Target Ther. 2021;6:402. doi: 10.1038/s41392-021-00791-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Villarino A.V., Kanno Y., O’Shea J.J. Mechanisms and consequences of Jak-STAT signaling in the immune system. Nat Immunol. 2017;18:374–384. doi: 10.1038/ni.3691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Woroniecka K.I., Park A.S., Mohtat D., Thomas D.B., Pullman J.M., Susztak K. Transcriptome analysis of human diabetic kidney disease. Diabetes. 2011;60:2354–2369. doi: 10.2337/db10-1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lu T.C., Wang Z.H., Feng X., et al. Knockdown of Stat3 activity in vivo prevents diabetic glomerulopathy. Kidney Int. 2009;76:63–71. doi: 10.1038/ki.2009.98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Berthier C.C., Zhang H., Schin M., et al. Enhanced expression of Janus kinase-signal transducer and activator of transcription pathway members in human diabetic nephropathy. Diabetes. 2009;58:469–477. doi: 10.2337/db08-1328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Brosius F.C., 3rd, He J.C. JAK inhibition and progressive kidney disease. Curr Opin Nephrol Hypertens. 2015;24:88–95. doi: 10.1097/MNH.0000000000000079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Nystrom S.E., Li G., Datta S., et al. JAK inhibitor blocks COVID-19 cytokine-induced JAK/STAT/APOL1 signaling in glomerular cells and podocytopathy in human kidney organoids. JCI Insight. 2022;7 doi: 10.1172/jci.insight.157432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Abid Q., Best Rocha A., Larsen C.P., et al. APOL1-associated collapsing focal segmental glomerulosclerosis in a patient with stimulator of interferon genes (STING)-associated vasculopathy with onset in infancy (SAVI) Am J Kidney Dis. 2020;75:287–290. doi: 10.1053/j.ajkd.2019.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Langefeld C.D., Comeau M.E., Ng M.C.Y., et al. Genome-wide association studies suggest that APOL1-environment interactions more likely trigger kidney disease in African Americans with nondiabetic nephropathy than strong APOL1-second gene interactions. Kidney Int. 2018;94:599–607. doi: 10.1016/j.kint.2018.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Chaudhary N.S., Armstrong N.D., Hidalgo B.A., et al. SMOC2 gene interacts with APOL1 in the development of end-stage kidney disease: a genome-wide association study. Front Med (Lausanne) 2022;9 doi: 10.3389/fmed.2022.971297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Vy H.M.T., Lin B.M., Gulamali F.F., et al. Genome-wide epistatic interaction between DEF1B and APOL1 high-risk genotypes for chronic kidney disease. Clin J Am Soc Nephrol. 2022;17:1522–1525. doi: 10.2215/CJN.03610322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zhang J.Y., Wang M., Tian L., et al. UBD modifies APOL1-induced kidney disease risk. Proc Natl Acad Sci U S A. 2018;115:3446–3451. doi: 10.1073/pnas.1716113115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Divers J., Ma L., Brown W.M., et al. Genome-wide association study for time to failure of kidney transplants from African American deceased donors. Clin Transpl. 2020;34 doi: 10.1111/ctr.13827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Bodonyi-Kovacs G., Ma J.Z., Chang J., et al. Combined effects of GSTM1 null allele and APOL1 renal risk alleles in CKD progression in the African American study of kidney disease and hypertension trial. J Am Soc Nephrol. 2016;27:3140–3152. doi: 10.1681/ASN.2015050487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Chen T.K., Coresh J., Daya N., et al. Race, APOL1 risk variants, and clinical outcomes among older adults: the ARIC study. J Am Geriatr Soc. 2021;69:155–163. doi: 10.1111/jgs.16797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Tamrat R., Peralta C.A., Tajuddin S.M., Evans M.K., Zonderman A.B., Crews D.C. Apolipoprotein L1, income and early kidney damage. BMC Nephrol. 2015;16:14. doi: 10.1186/s12882-015-0008-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hung A.M., Shah S.C., Bick A.G., et al. APOL1 risk variants, acute kidney injury, and death in participants with African ancestry hospitalized with COVID-19 from the Million Veteran Program. JAMA Intern Med. 2022;182:386–395. doi: 10.1001/jamainternmed.2021.8538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Paranjpe I., Chaudhary K., Paranjpe M., et al. Association of APOL1 risk genotype and air pollution for kidney disease. Clin J Am Soc Nephrol. 2020;15:401–403. doi: 10.2215/CJN.11921019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Beckerman P., Bi-Karchin J., Park A.S., et al. Transgenic expression of human APOL1 risk variants in podocytes induces kidney disease in mice. Nat Med. 2017;23:429–438. doi: 10.1038/nm.4287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Datta S., Kataria R., Zhang J.Y., et al. Kidney disease-associated APOL1 variants have dose-dependent, dominant toxic gain-of-function. J Am Soc Nephrol. 2020;31:2083–2096. doi: 10.1681/ASN.2020010079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Daneshpajouhnejad P., Kopp J.B., Winkler C.A., Rosenberg A.Z. The evolving story of apolipoprotein L1 nephropathy: the end of the beginning. Nat Rev Nephrol. 2022;18:307–320. doi: 10.1038/s41581-022-00538-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Scales S.J., Gupta N., De Maziere A.M., et al. Apolipoprotein L1-specific antibodies detect endogenous APOL1 inside the endoplasmic reticulum and on the plasma membrane of podocytes. J Am Soc Nephrol. 2020;31:2044–2064. doi: 10.1681/ASN.2019080829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Vanwalleghem G., Fontaine F., Lecordier L., et al. Coupling of lysosomal and mitochondrial membrane permeabilization in trypanolysis by APOL1. Nat Commun. 2015;6:8078. doi: 10.1038/ncomms9078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Madhavan S.M., O’Toole J.F., Konieczkowski M., Ganesan S., Bruggeman L.A., Sedor J.R. APOL1 localization in normal kidney and nondiabetic kidney disease. J Am Soc Nephrol. 2011;22:2119–2128. doi: 10.1681/ASN.2011010069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ma L., Shelness G.S., Snipes J.A., et al. Localization of APOL1 protein and mRNA in the human kidney: nondiseased tissue, primary cells, and immortalized cell lines. J Am Soc Nephrol. 2015;26:339–348. doi: 10.1681/ASN.2013091017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Shah S.S., Lannon H., Dias L., et al. APOL1 kidney risk variants induce cell death via mitochondrial translocation and opening of the mitochondrial permeability transition pore. J Am Soc Nephrol. 2019;30:2355–2368. doi: 10.1681/ASN.2019020114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Perez-Morga D., Vanhollebeke B., Paturiaux-Hanocq F., et al. Apolipoprotein L-I promotes trypanosome lysis by forming pores in lysosomal membranes. Science. 2005;309:469–472. doi: 10.1126/science.1114566. [DOI] [PubMed] [Google Scholar]
- 47.Vanhollebeke B., Pays E. The function of apolipoproteins L. Cell Mol Life Sci. 2006;63:1937–1944. doi: 10.1007/s00018-006-6091-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Thomson R., Finkelstein A. Human trypanolytic factor APOL1 forms pH-gated cation-selective channels in planar lipid bilayers: relevance to trypanosome lysis. Proc Natl Acad Sci U S A. 2015;112:2894–2899. doi: 10.1073/pnas.1421953112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Olabisi O.A., Zhang J.Y., VerPlank L., et al. APOL1 kidney disease risk variants cause cytotoxicity by depleting cellular potassium and inducing stress-activated protein kinases. Proc Natl Acad Sci U S A. 2016;113:830–837. doi: 10.1073/pnas.1522913113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Bruno J., Pozzi N., Oliva J., Edwards J.C. Apolipoprotein L1 confers pH-switchable ion permeability to phospholipid vesicles. J Biol Chem. 2017;292:18344–18353. doi: 10.1074/jbc.M117.813444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Ma L., Divers J., Freedman B.I. Mechanisms of injury in APOL1-associated Kidney Disease. Kidney Dis Transplant. 2019;103:487–492. doi: 10.1097/TP.0000000000002509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ma L., Chou J.W., Snipes J.A., et al. APOL1 renal-risk variants induce mitochondrial dysfunction. J Am Soc Nephrol. 2017;28:1093–1105. doi: 10.1681/ASN.2016050567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ogata M., Hino S., Saito A., et al. Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol. 2006;26:9220–9231. doi: 10.1128/MCB.01453-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Youle R.J., van der Bliek A.M. Mitochondrial fission, fusion, and stress. Science. 2012;337:1062–1065. doi: 10.1126/science.1219855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Archer S.L. Mitochondrial dynamics--mitochondrial fission and fusion in human diseases. N Engl J Med. 2013;369:2236–2251. doi: 10.1056/NEJMra1215233. [DOI] [PubMed] [Google Scholar]
- 56.Ma L., Ainsworth H.C., Snipes J.A., et al. APOL1 kidney-risk variants induce mitochondrial fission. Kidney Int Rep. 2020;5:891–904. doi: 10.1016/j.ekir.2020.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kim H., Lee J.Y., Park K.J., Kim W.H., Roh G.S. A mitochondrial division inhibitor, Mdivi-1, inhibits mitochondrial fragmentation and attenuates kainic acid-induced hippocampal cell death. BMC Neurosci. 2016;17:33. doi: 10.1186/s12868-016-0270-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kruzel-Davila E., Shemer R., Ofir A., et al. APOL1-mediated cell injury involves disruption of conserved trafficking processes. J Am Soc Nephrol. 2017;28:1117–1130. doi: 10.1681/ASN.2016050546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lan X., Jhaveri A., Cheng K., et al. APOL1 risk variants enhance podocyte necrosis through compromising lysosomal membrane permeability. Am J Physiol Ren Physiol. 2014;307:F326–F336. doi: 10.1152/ajprenal.00647.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kruzel-Davila E., Bavli-Kertselli I., Ofir A., et al. Endoplasmic reticulum-translocation is essential for APOL1 cellular toxicity. iScience. 2022;25 doi: 10.1016/j.isci.2021.103717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chun J., Zhang J.Y., Wilkins M.S., et al. Recruitment of APOL1 kidney disease risk variants to lipid droplets attenuates cell toxicity. Proc Natl Acad Sci U S A. 2019;116:3712–3721. doi: 10.1073/pnas.1820414116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Grampp S., Kruger R., Lauer V., et al. Hypoxia hits APOL1 in the kidney. Kidney Int. 2023;104:53–60. doi: 10.1016/j.kint.2023.03.035. [DOI] [PubMed] [Google Scholar]
- 63.Cybulsky A.V. The intersecting roles of endoplasmic reticulum stress, ubiquitin- proteasome system, and autophagy in the pathogenesis of proteinuric kidney disease. Kidney Int. 2013;84:25–33. doi: 10.1038/ki.2012.390. [DOI] [PubMed] [Google Scholar]
- 64.Zhang H., You Q.D., Xu X.L. Targeting stimulator of interferon genes (STING): a medicinal chemistry perspective. J Med Chem. 2020;63:3785–3816. doi: 10.1021/acs.jmedchem.9b01039. [DOI] [PubMed] [Google Scholar]
- 65.Zhang S., Zheng R., Pan Y., Sun H. Potential therapeutic value of the STING inhibitors. Molecules. 2023;28 doi: 10.3390/molecules28073127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Couillin I., Riteau N. STING signaling and sterile inflammation. Front Immunol. 2021;12 doi: 10.3389/fimmu.2021.753789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Decout A., Katz J.D., Venkatraman S., Ablasser A. The cGAS-STING pathway as a therapeutic target in inflammatory diseases. Nat Rev Immunol. 2021;21:548–569. doi: 10.1038/s41577-021-00524-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wu J., Raman A., Coffey N.J., et al. The key role of NLRP3 and STING in APOL1-associated podocytopathy. J Clin Invest. 2021:131. doi: 10.1172/JCI136329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Okamoto K., Rausch J.W., Wakashin H., et al. APOL1 risk allele RNA contributes to renal toxicity by activating protein kinase R. Commun Biol. 2018;1:188. doi: 10.1038/s42003-018-0188-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Wan G., Zhaorigetu S., Liu Z., Kaini R., Jiang Z., Hu C.A. Apolipoprotein L1, a novel Bcl-2 homology domain 3-only lipid-binding protein, induces autophagic cell death. J Biol Chem. 2008;283:21540–21549. doi: 10.1074/jbc.M800214200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Hayek S.S., Koh K.H., Grams M.E., et al. A tripartite complex of suPAR, APOL1 risk variants and alphavbeta3 integrin on podocytes mediates chronic kidney disease. Nat Med. 2017;23:945–953. doi: 10.1038/nm.4362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Hughes J.P., Rees S., Kalindjian S.B., Philpott K.L. Principles of early drug discovery. Br J Pharmacol. 2011;162:1239–1249. doi: 10.1111/j.1476-5381.2010.01127.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Jenwitheesuk E., Horst J.A., Rivas K.L., Van Voorhis W.C., Samudrala R. Novel paradigms for drug discovery: computational multitarget screening. Trends Pharmacol Sci. 2008;29:62–71. doi: 10.1016/j.tips.2007.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Moffat J.G., Vincent F., Lee J.A., Eder J., Prunotto M. Opportunities and challenges in phenotypic drug discovery: an industry perspective. Nat Rev Drug Discov. 2017;16:531–543. doi: 10.1038/nrd.2017.111. [DOI] [PubMed] [Google Scholar]
- 75.Plenge R.M., Scolnick E.M., Altshuler D. Validating therapeutic targets through human genetics. Nat Rev Drug Discov. 2013;12:581–594. doi: 10.1038/nrd4051. [DOI] [PubMed] [Google Scholar]
- 76.Tautermann C.S. Current and future challenges in modern drug discovery. Methods Mol Biol. 2020;2114:1–17. doi: 10.1007/978-1-0716-0282-9_1. [DOI] [PubMed] [Google Scholar]
- 77.Reilly D.F., Breyer M.D. The use of genomics to drive kidney disease drug discovery and development. Clin J Am Soc Nephrol. 2020;15:1342–1351. doi: 10.2215/CJN.11070919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Liu G.H., Chen T., Zhang X., Ma X., Shi H. Small molecule inhibitors targeting the cancers. MedCommComm (2020) 2022;3:e181. doi: 10.1002/mco2.181. 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Zhou Y., Castonguay P., Sidhom E.H., et al. A small-molecule inhibitor of TRPC5 ion channels suppresses progressive kidney disease in animal models. Science. 2017;358:1332–1336. doi: 10.1126/science.aal4178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Assimon V., Bronner S., Yu C., et al. MZ-301 Is a Small Molecule Inhibitor of APOL1 Pore Function That Attenuates Albuminuria in a Mouse Model of APOL1-Mediated Kidney Disease. ASN Kidney Week 2022 Poster FR-PO318. https://www.asn-online.org/education/kidneyweek/archives/KW22Abstracts.pdf
- 81.Carton-Garcia F., Saande C.J., Meraviglia-Crivelli D., Aldabe R., Pastor F. Oligonucleotide-based therapies for renal diseases. Biomedicines. 2021;9:303. doi: 10.3390/biomedicines9030303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Dhuri K., Bechtold C., Quijano E., et al. Antisense oligonucleotides: an emerging area in drug discovery and development. J Clin Med. 2020;9:2004. doi: 10.3390/jcm9062004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Li H., Wang C., Che R., et al. A potential therapy using antisense oligonucleotides to treat autosomal recessive polycystic kidney disease. J Clin Med. 2023:12. doi: 10.3390/jcm12041428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Aghajan M., Booten S.L., Althage M., et al. Antisense oligonucleotide treatment ameliorates IFN-gamma-induced proteinuria in APOL1-transgenic mice. JCI Insight. 2019;4 doi: 10.1172/jci.insight.126124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Shastri A., Choudhary G., Teixeira M., et al. Antisense STAT3 inhibitor decreases viability of myelodysplastic and leukemic stem cells. J Clin Invest. 2018;128:5479–5488. doi: 10.1172/JCI120156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Hong D., Kurzrock R., Kim Y., et al. AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3 with early evidence of clinical activity in lymphoma and lung cancer. Sci Transl Med. 2015;7:314ra185. doi: 10.1126/scitranslmed.aac5272. [DOI] [PMC free article] [PubMed] [Google Scholar]


