[HTML][HTML] Metabolic reprogramming in a slowly developing orthologous model of polycystic kidney disease

K Hopp, EK Kleczko, BY Gitomer… - American Journal …, 2022 - journals.physiology.org
K Hopp, EK Kleczko, BY Gitomer, M Chonchol, J Klawitter, U Christians, J Klawitter
American Journal of Physiology-Renal Physiology, 2022journals.physiology.org
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited
kidney disease and affects 1 in 1,000 individuals. There is accumulating evidence
suggesting that there are shared cellular mechanisms responsible for cystogenesis in
human and murine PKD and that reprogramming of metabolism is a key disease feature. In
this study, we used a targeted metabolomics approach in an orthologous mouse model of
PKD (Pkd1 RC/RC) to investigate the metabolic modifications a cystic kidney undergoes …
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease and affects 1 in 1,000 individuals. There is accumulating evidence suggesting that there are shared cellular mechanisms responsible for cystogenesis in human and murine PKD and that reprogramming of metabolism is a key disease feature. In this study, we used a targeted metabolomics approach in an orthologous mouse model of PKD (Pkd1 RC/RC) to investigate the metabolic modifications a cystic kidney undergoes during disease progression. Using the Kyoto Encyclopedia of Genes and Genomes pathway database, we identified several biologically relevant metabolic pathways that were altered early in this disease (in 3-mo-old Pkd1 RC/RC mice), the most highly represented being arginine biosynthesis and metabolism and tryptophan and phenylalanine metabolism. During the next 6 mo of disease progression, multiple uremic solutes accumulated in the kidney of cystic mice, including several established markers of oxidative stress and endothelial dysfunction (allantoin, asymmetric dimethylarginine, homocysteine, malondialdehyde, methionine sulfoxide, and S-adenosylhomocysteine). Levels of kynurenines and polyamines were also augmented in kidneys of Pkd1 RC/RC versus wild-type mice, as were the levels of bacteria-produced indoles, whose increase within PKD kidneys suggests microbial dysbiosis. In summary, we confirmed previously published and identified novel metabolic markers and pathways of PKD progression that may prove helpful for diagnosis and monitoring of cystic kidney disease in patients. Furthermore, they provide targets for novel therapeutic approaches that deserve further study and hint toward currently understudied pathomechanisms.
NEW & NOTEWORTHY This report delineates the evolution of metabolic changes occurring during autosomal dominant polycystic kidney disease (ADPKD) progression. Using an orthologous model, we performed kidney metabolomics and confirmed dysregulation of metabolic pathways previously found altered in nonorthologous or rapidly-progressive PKD models. Importantly, we identified novel alterations, including augmentation of kynurenines, polyamines, and indoles, suggesting increased inflammation and microbial dysbiosis that provide insights into PKD pathomechanisms and may prove helpful for diagnosing, monitoring, and treating ADPKD.
American Physiological Society