Application of Exfoliated Podocytes from Urine in CKD: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Chronic kidney disease (CKD) is a global health issue, affecting more than 10% of the worldwide population. It is defined by structural and functional changes to the kidney. Urinary exfoliated podocytes and podocyte-specific markers have demonstrated value for the early diagnosis of CKD and prognosticating CKD progression.

  • exfoliated kidney cells
  • chronic kidney disease
  • non-invasive
  • early diagnosis

1. Introduction

Chronic kidney disease (CKD) is a progressive disease that is defined by structural and functional changes to the kidney [1]. CKD is considered to be a global issue, one with a substantial public health burden which is exponentially growing [2]. With more than 10% of the adult population currently affected by CKD, it is projected to become the fifth leading cause of mortality worldwide by 2040 [3]. There are multiple causes of CKD, some of which are more common and clearly defined (e.g., diabetes mellitus, hypertension, glomerulonephritis and polycystic kidney disease), whilst others are not fully understood (e.g., Mesoamerican nephropathy) [4][5][6][7]. CKD progresses differently in each individual, depending on the primary cause of CKD, as well as other co-morbidities [8]. CKD is typically identified by a reduction in kidney function, an estimated glomerular filtration rate (eGFR) of less than 60 mL/min/1.73 m2, and supported by markers of kidney tissue damage (albuminuria and hematuria), as well as other laboratory-based and imaging investigations that are present for at least 3 months [9]. The identification of early CKD, particularly in younger patients, remains challenging. Asymptomatic individuals living with CKD can lose up to 90% of their kidney function, at which point CKD is irreversible, given the advanced pathological damage [10]. Early diagnosis of CKD is clinically important, given that therapies are now available to stabilize kidney function from an early stage of the disease [11][12].
Histopathological examination of the kidney is the gold standard for the diagnosis and prognostication of CKD [13]. However, the risk of adverse events for patients after kidney biopsy has been well-documented. Post-biopsy risks include bleeding, excess pain and occasionally nephrectomy. For most individuals, bleeding usually resolves spontaneously following kidney biopsy, although for a small percentage of individuals, blood transfusion may be required [14][15]. Recently, the stress for the physician of routinely performing kidney biopsies has been addressed, with the increasing workload and time pressures of the modern-day clinical environment contributing to this issue [16]. There is a suggestion that the quality of training in kidney biopsy has reduced in recent years [17]. With the continuous development of non-invasive diagnostic and prognostication tools in CKD, it is questioned whether other diagnostic methods can complement or replace kidney biopsy in the near future.

2. Application of Exfoliated Podocytes in CKD

Urinary exfoliated podocytes and podocyte-specific markers have demonstrated value for the early diagnosis of CKD and prognosticating CKD progression (Table 1). Diabetic kidney disease (DKD) is the most common cause of CKD worldwide. In a post-hoc exploratory analysis comparing archived urine samples from normoalbuminuric patients with uncomplicated type 1 diabetes and healthy controls, urinary podocyte microparticle levels were found to be higher in the cohort with type 1 diabetes [18]. Interestingly, the elevation of urinary podocyte microparticle levels was well in advance of changes to other more well-established biomarkers of CKD such as albuminuria and nephrin, suggesting its potential utility as an early biomarker of glomerular injury in uncomplicated type 1 diabetes [18]. There is evidence demonstrating significant differences in urinary podocyte mRNA levels of nephrin, podocin, synaptopodin, Wilms Tumor-1 (WT-1) and α-actinin-4 between DKD and non-DKD patients [19]. These markers were found to precede the clinical appearance of microalbuminuria in patients with type 2 diabetes [20]. Urinary synaptopodocin mRNA levels were used to measure therapeutic response to angiotensin-converting enzyme inhibitor and angiotensin-receptor blocker treatment in DKD [21]. Urinary podocyte-derived indices such as podocin mRNA-to-creatinine ratio are a strong marker of podocyte detachment from GBM and are shown to project the rate of kidney functional decline in DKD [19]. For minimal change disease (MCN) and focal segmental glomerulosclerosis (FSGS), urinary nephrin and podocin mRNA levels were lower in patients with MCN and FSGS compared to healthy controls, and urinary nephrin and podocin mRNA levels correlated with the degree of proteinuria within this context [22]. Urinary synaptopodin mRNA levels were found to correlate with kidney function decline in FSGS [22]. In membranous nephropathy (MN), the number of urinary podocyte-derived microparticles displayed an inverse relationship with clinical parameters of MN, decreasing with improving clinical parameters following immunosuppression treatment [23]. The urinary podocyte mRNA levels of nephrin, podocin, and synaptopodin were all elevated in MN, with these levels clearly differentiating MN from other causes of nephrotic syndrome [24].
Table 1. Clinical utility of urinary exfoliated podocytes and podocyte-specific markers for early diagnosis and prognostication of CKD.
The role of exfoliated podocytes from urine as markers to prognosticate the progression of mesangial diseases such as IgA nephropathy has been explored. Previous studies noted that urinary podocyte counts correlated with serum creatinine and proteinuria in IgA nephropathy [25]. Those with segmental sclerosis, which was confirmed histologically, had greater numbers of urinary podocytes compared to those without segmental sclerosis [26]. Evidence is incomplete regarding the use of podocyte-specific mRNA and miRNA levels in IgA nephropathy as biomarkers for early diagnosis and prognostication, and this requires further study. For other mesangial glomerulopathies outside of IgA nephropathy, urinary podocyte counts are increased in hereditary and acquired diffuse mesangial sclerosis [26]. The application of non-invasive risk assessment techniques is not as well studied for these conditions, likely explained by a lack of clarity in pathological classification.
Utilizing urinary podocytes as markers of disease activity in lupus nephritis has been evaluated. Studies have noted that the majority of urinary podocytes in patients with lupus nephritis are viable but dedifferentiated, with a greater proportion of apoptotic urinary podocytes being much lower in patients without kidney disease [27]. Urinary podocyte-specific mRNA markers such as podocalyxin, synaptopodin, podocin, nephrin, as well as WT-1 levels, are significantly elevated in patients with active lupus nephritis compared to those without systemic lupus or active lupus nephritis [28]. Within this context, urinary nephrin mRNA levels are shown to correlate with the degree of proteinuria and systemic lupus disease activity, but not with the histological progression of lupus nephritis [32]. Meanwhile, urinary podocin mRNA levels are demonstrated to be an independent predictor of kidney function decline in lupus nephritis [27][28][32].
There is minimal data regarding the use of exfoliated podocytes from urine for anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis. It was previously found that the rate of podocyte detachment to urine predicted kidney function loss in ANCA-associated vasculitis [33]. Reports have also suggested that urinary podocin-to-nephrin mRNA ratio, a surrogate marker of intra-glomerular podocyte stress, correlated with the extent of crescent formation [29]. Paradoxically, patients with higher urinary podocyte-specific mRNA levels achieved better outcomes, as this indicated a stronger glomerular podocyte reserve for the reversibility of vasculitic disease [29].
Ultimately, there are urinary podocyte-specific biomarkers which have shown universal prognostic value for all forms of CKD. Urine synaptopodin levels are described as a generic marker of podocyte damage. Synaptopodin protein expression, determined by Western blot, has demonstrated significant correlations with kidney function in CKD, regardless of the degree of albuminuria [34]. Urinary podocyte-specific mRNA targets such as urinary brain-derived neurotrophic factor mRNA level had the best correlation with urinary kidney injury molecule-1 (KIM-1), which is recognized as a generic marker to prognosticate CKD progression [30].
The recent development of single-cell RNA sequencing (scRNA-seq) is a revolutionary technique in providing an unbiased genome-wide characterization of individual exfoliated cells from urine at scale [35]. Both animal and human studies have demonstrated that scRNA-seq is able to generate an initial map of gene expression for most kidney cells, thereby allowing us to advance from a morphology-based cell characterization through cell shape, color and location to the more objective method of cellular definition through transcriptomics [36][37]. In CKD, scRNA-seq may be able to define cell type-specific changes, cell fractions and cell-to-cell interactions [31][38][39]. This information can be useful for the diagnosis and risk stratification of CKD. In a combined analysis between urinary exfoliated podocytes, bladder single cells and human kidney tissue nucleus (extracted from DKD and control patients) scRNA-seq datasets, a strong correlative relationship was found between exfoliated podocyte and kidney tissue nucleus scRNA-seq datasets, where together they formed a strong cluster [31]. Urinary scRNA-seq has particularly shown a strong expression of monogenic nephrotic syndrome genes in podocytes [31]. Nevertheless, these results were obtained in a pilot study with few patients and controls. Abedini and colleagues ensured urine was collected at different time points and through different methods to confirm the reproducibility and feasibility of this approach [31]. Other confounding factors include the differences of scRNA-seq in capturing efficiency between the male and female urine samples, and perhaps a subgroup analyses between them is required to reduce sex-associated bias in the interpretation of results. Furthermore, there was significantly higher ambient RNA contamination in 24-h urine collections [31]. Meticulous arrangement to optimize the environment of urinary cell storage may mitigate the risks of cellular degradation during extended storage. As the authors noted, large prospective cohort studies are needed to validate the diagnostic and prognostic utility of urinary scRNA-seq in CKD.

This entry is adapted from the peer-reviewed paper 10.3390/ijms23147610

References

  1. Romagnani, P.; Remuzzi, G.; Glassock, R.; Levin, A.; Jager, K.J.; Tonelli, M.; Massy, Z.; Wanner, C.; Anders, H.J. Chronic kidney disease. Nat. Rev. Dis. Primers 2017, 3, 1–24.
  2. Kalantar-Zadeh, K.; Jafar, T.H.; Nitsch, D.; Neuen, B.L.; Perkovic, V. Chronic kidney disease. Lancet 2021, 398, 786–802.
  3. Foreman, K.J.; Marquez, N.; Dolgert, A.; Fukutaki, K.; Fullman, N.; McGaughey, M.; Pletcher, M.A.; Smith, A.E.; Tang, K.; Yuan, C.W.; et al. Forecasting life expectancy, years of life lost, and all-cause and cause-specific mortality for 250 causes of death: Reference and alternative scenarios for 2016–40 for 195 countries and territories. Lancet 2018, 392, 2052–2090.
  4. Anders, H.J.; Huber, T.B.; Isermann, B.; Schiffer, M. CKD in diabetes: Diabetic kidney disease versus nondiabetic kidney disease. Nat Rev. Nephrol. 2018, 14, 361–377.
  5. Couser, W.G. Glomerulonephritis. Lancet 1999, 353, 1509–1515.
  6. Alan, S.L.; Shen, C.; Landsittel, D.P.; Harris, P.C.; Torres, V.E.; Mrug, M.; Bae, K.T.; Grantham, J.J.; Rahbari-Oskoui, F.F.; Flessner, M.F.; et al. Baseline total kidney volume and the rate of kidney growth are associated with chronic kidney disease progression in Autosomal Dominant Polycystic Kidney Disease. Kidney Int. 2018, 93, 691–699.
  7. Ku, E.; Lee, B.J.; Wei, J.; Weir, M.R. Hypertension in CKD: Core curriculum 2019. Am. J. Kidney Dis. 2019, 74, 120–131.
  8. Ku, E.; Johansen, K.L.; McCulloch, C.E. Time-centered approach to understanding risk factors for the progression of CKD. Clin. J. Am. Soc. Nephrol. 2018, 13, 693–701.
  9. Levin, A.; Stevens, P.E.; Bilous, R.W.; Coresh, J.; De Francisco, A.L.; De Jong, P.E.; Griffith, K.E.; Hemmelgarn, B.R.; Iseki, K.; Lamb, E.J.; et al. Kidney Disease: Improving Global Outcomes (KDIGO) CKD Work Group. KDIGO 2012 clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int. Suppl. 2013, 3, 1–50.
  10. John, R.; Webb, M.; Young, A.; Stevens, P.E. Unreferred chronic kidney disease: A longitudinal study. Am. J. Kidney Dis. 2004, 43, 825–835.
  11. Neuen, B.L.; Ohkuma, T.; Neal, B.; Matthews, D.R.; De Zeeuw, D.; Mahaffey, K.W.; Fulcher, G.; Desai, M.; Li, Q.; Deng, H.; et al. Cardiovascular and renal outcomes with canagliflozin according to baseline kidney function: Data from the CANVAS Program. Circulation 2018, 138, 1537–1550.
  12. Butler, J.; Zannad, F.; Fitchett, D.; Zinman, B.; Koitka-Weber, A.; von Eynatten, M.; Zwiener, I.; George, J.; Brueckmann, M.; Cheung, A.K.; et al. Empagliflozin Improves Kidney Outcomes in Patients With or Without Heart Failure: Insights From the EMPA-REG OUTCOME Trial. Circ Heart Fail. 2019, 12, e005875.
  13. Berchtold, L.; Friedli, I.; Vallée, J.P.; Moll, S.; Martin, P.Y.; De Seigneux Matthey, S. Diagnosis and assessment of renal fibrosis: The state of the art. Swiss Med Wkly. 2017, 147, w14442.
  14. Poggio, E.D.; McClelland, R.L.; Blank, K.N.; Hansen, S.; Bansal, S.; Bomback, A.S.; Canetta, P.A.; Khairallah, P.; Kiryluk, K.; Lecker, S.H.; et al. Systematic review and meta-analysis of native kidney biopsy complications. Clin. J. Am. Soc. Nephrol. 2020, 15, 1595–1602.
  15. Corapi, K.M.; Chen, J.L.; Balk, E.M.; Gordon, C.E. Bleeding complications of native kidney biopsy: A systematic review and meta-analysis. Am. J. Kidney Dis. 2012, 60, 62–73.
  16. Gilbert, S.J. Does the Kidney Biopsy Portend the Future of Nephrology? Clin. J. Am. Soc. Nephrol. 2018, 13, 681–682.
  17. Rodby, R.A. Kidney biopsy should remain a required procedure for nephrology training programs: CON. Kidney360 2022. in print.
  18. Lytvyn, Y.; Xiao, F.; Kennedy, C.R.; Perkins, B.A.; Reich, H.N.; Scholey, J.W.; Cherney, D.Z.; Burger, D. Assessment of urinary microparticles in normotensive patients with type 1 diabetes. Diabetologia 2017, 60, 581–584.
  19. Wang, G.; Lai, F.M.; Lai, K.B.; Chow, K.M.; Li, P.K.T.; Szeto, C.C. Messenger RNA expression of podocyte-associated molecules in the urinary sediment of patients with diabetic nephropathy. Nephron Clin. Pract. 2007, 106, c169–c179.
  20. Fukuda, A.; Minakawa, A.; Kikuchi, M.; Sato, Y.; Nagatomo, M.; Nakamura, S.; Mizoguchi, T.; Fukunaga, N.; Shibata, H.; Naik, A.S.; et al. Urinary podocyte mRNAs precede microalbuminuria as a progression risk marker in human type 2 diabetic nephropathy. Sci. Rep. 2020, 10, 18209.
  21. Wang, G.; Lai, F.M.; Lai, K.B.; Chow, K.M.; Kwan, B.C.; Li, P.K.; Szeto, C.C. Urinary messenger RNA expression of podocyte-associated molecules in patients with diabetic nephropathy treated by angiotensin-converting enzyme inhibitor and angiotensin receptor blocker. Eur. J. Endocrinol. 2008, 158, 317–322.
  22. Szeto, C.C.; Wang, G.; Chow, K.M.; Lai, F.M.; Ma, T.K.; Kwan, B.C.; Luk, C.C.; Li, P.K.T. Podocyte mRNA in the urinary sediment of minimal change nephropathy and focal segmental glomerulosclerosis. Clin. Nephrol. 2015, 84, 198–205.
  23. Lu, J.; Hu, Z.B.; Chen, P.P.; Lu, C.C.; Zhang, J.X.; Li, X.Q.; Yuan, B.Y.; Huang, S.J.; Ma, K.L. Urinary levels of podocyte-derived microparticles are associated with the progression of chronic kidney disease. Ann. Transl. Med. 2019, 7, 445.
  24. Szeto, C.C.; Lai, K.B.; Chow, K.M.; Szeto, C.Y.; Yip, T.W.; Woo, K.S.; Li, P.K.; Lai, F.M. Messenger RNA expression of glomerular podocyte markers in the urinary sediment of acquired proteinuric diseases. Clin. Chim. Acta 2005, 361, 182–190.
  25. Shen, P.; Shen, J.; Li, W.; He, L. Urinary podocyte can be an indicator for the pathogenetic condition of patients with IgA nephropathy. Clin. Lab. 2014, 60, 1709–1715.
  26. Asao, R.; Asanuma, K.; Kodama, F.; Akiba-Takagi, M.; Nagai-Hosoe, Y.; Seki, T.; Takeda, Y.; Ohsawa, I.; Mano, S.; Matsuoka, K.; et al. Relationships between levels of urinary podocalyxin, number of urinary podocytes, and histologic injury in adult patients with IgA nephropathy. Clin. J. Am. Soc. Nephrol. 2012, 7, 1385–1393.
  27. Perez-Hernandez, J.; Olivares, M.D.; Forner, M.J.; Chaves, F.J.; Cortes, R.; Redon, J. Urinary dedifferentiated podocytes as a non-invasive biomarker of lupus nephritis. Nephrol. Dial Transplant. 2016, 31, 780–789.
  28. Kwon, S.H.; Woollard, J.R.; Saad, A.; Garovic, V.D.; Zand, L.; Jordan, K.L.; Textor, S.C.; Lerman, L.O. Elevated urinary podocyte-derived extracellular microvesicles in renovascular hypertensive patients. Nephrol. Dial. Transplant. 2017, 32, 800–807.
  29. Minakawa, A.; Fukuda, A.; Kikuchi, M.; Sato, Y.; Sato, Y.; Kitamura, K.; Fujimoto, S. Urinary podocyte mRNA is a potent biomarker of anti-neutrophil cytoplasmic antibody-associated glomerulonephritis. Clin. Exp. Nephrol. 2020, 24, 242–252.
  30. Endlich, N.; Lange, T.; Kuhn, J.; Klemm, P.; Kotb, A.M.; Siegerist, F.; Kindt, F.; Lindenmeyer, M.T.; Cohen, C.D.; Kuss, A.W.; et al. BDNF: mRNA expression in urine cells of patients with chronic kidney disease and its role in kidney function. J. Cell. Mol. Med. 2018, 22, 5265–5277.
  31. Abedini, A.; Zhu, Y.O.; Chatterjee, S.; Halasz, G.; Devalaraja-Narashimha, K.; Shrestha, R.; Balzer, M.S.; Park, J.; Zhou, T.; Ma, Z.; et al. Urinary single-cell profiling captures the cellular diversity of the kidney. J. Am. Soc. Nephrol. 2021, 32, 614–627.
  32. Wang, G.; Lai, F.M.; Tam, L.S.; Li, K.M.; Lai, K.B.; Chow, K.M.; Li, K.T.; Szeto, C.C. Messenger RNA expression of podocyte-associated molecules in urinary sediment of patients with lupus nephritis. J. Rheumatol. 2007, 34, 2358–2364.
  33. Zou, R.; Wang, S.X.; Liu, G.; Yu, F.; Chen, M.; Zhao, M.H. Podocyte detachment is associated with renal prognosis in ANCA-associated glomerulonephritis: A retrospective cohort study. Medicine 2016, 95, e3294.
  34. Kwon, S.K.; Kim, S.J.; Kim, H.Y. Urine synaptopodin excretion is an important marker of glomerular disease progression. Korean J. Intern. Med. 2016, 31, 938–943.
  35. Wang, Y.J.; Kaestner, K.H. Single-cell RNA-seq of the pancreatic islets—A promise not yet fulfilled? Cell Metab. 2019, 29, 539–544.
  36. Park, J.; Shrestha, R.; Qiu, C.; Kondo, A.; Huang, S.; Werth, M.; Li, M.; Barasch, J.; Suszták, K. Single-cell transcriptomics of the mouse kidney reveals potential cellular targets of kidney disease. Science 2018, 360, 758–763.
  37. Park, J.; Liu, C.L.; Kim, J.; Susztak, K. Understanding the kidney one cell at a time. Kidney Int. 2019, 96, 862–870.
  38. Arazi, A.; Rao, D.A.; Berthier, C.C.; Davidson, A.; Liu, Y.; Hoover, P.J.; Chicoine, A.; Eisenhaure, T.M.; Jonsson, A.H.; Li, S.; et al. The immune cell landscape in kidneys of patients with lupus nephritis. Nat Immunol. 2019, 20, 902–914.
  39. Menon, R.; Otto, E.A.; Sealfon, R.; Nair, V.; Wong, A.K.; Theesfeld, C.L.; Chen, X.; Wang, Y.; Boppana, A.S.; Luo, J.; et al. SARS-CoV-2 receptor networks in diabetic and COVID-19–associated kidney disease. Kidney Int. 2020, 98, 1502–1518.
More
This entry is offline, you can click here to edit this entry!
Video Production Service