Modeling heritable kidney disease using human kidney iPSC-derived organoids

2022 ◽  
pp. 275-296
Author(s):  
Thomas A. Forbes ◽  
Melissa H. Little
Keyword(s):  
2021 ◽  
Vol 22 (14) ◽  
pp. 7642
Author(s):  
Zoran V. Popovic ◽  
Felix Bestvater ◽  
Damir Krunic ◽  
Bernhard K. Krämer ◽  
Raoul Bergner ◽  
...  

The CD73 pathway is an important anti-inflammatory mechanism in various disease settings. Observations in mouse models suggested that CD73 might have a protective role in kidney damage; however, no direct evidence of its role in human kidney disease has been described to date. Here, we hypothesized that podocyte injury in human kidney diseases alters CD73 expression that may facilitate the diagnosis of podocytopathies. We assessed the expression of CD73 and one of its functionally important targets, the C-C chemokine receptor type 2 (CCR2), in podocytes from kidney biopsies of 39 patients with podocytopathy (including focal segmental glomerulosclerosis (FSGS), minimal change disease (MCD), membranous glomerulonephritis (MGN) and amyloidosis) and a control group. Podocyte CD73 expression in each of the disease groups was significantly increased in comparison to controls (p < 0.001–p < 0.0001). Moreover, there was a marked negative correlation between CD73 and CCR2 expression, as confirmed by immunohistochemistry and immunofluorescence (Pearson r = −0.5068, p = 0.0031; Pearson r = −0.4705, p = 0.0313, respectively), thus suggesting a protective role of CD73 in kidney injury. Finally, we identify CD73 as a novel potential diagnostic marker of human podocytopathies, particularly of MCD that has been notorious for the lack of pathological features recognizable by light microscopy and immunohistochemistry.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Yuting Guan ◽  
Xiujie Liang ◽  
Ziyuan Ma ◽  
Hailong Hu ◽  
Hongbo Liu ◽  
...  

AbstractGenome-wide association studies (GWAS) have identified loci for kidney disease, but the causal variants, genes, and pathways remain unknown. Here we identify two kidney disease genes Dipeptidase 1 (DPEP1) and Charged Multivesicular Body Protein 1 A (CHMP1A) via the triangulation of kidney function GWAS, human kidney expression, and methylation quantitative trait loci. Using single-cell chromatin accessibility and genome editing, we fine map the region that controls the expression of both genes. Mouse genetic models demonstrate the causal roles of both genes in kidney disease. Cellular studies indicate that both Dpep1 and Chmp1a are important regulators of a single pathway, ferroptosis and lead to kidney disease development via altering cellular iron trafficking.


2021 ◽  
Vol 36 (Supplement_1) ◽  
Author(s):  
Sandrine Lemoine ◽  
Alireza Akbari ◽  
Taylor Marcus ◽  
Christopher McIntyre

Abstract Background and Aims Maintenance of a cortico-medullary concentration gradient (CMG) required for urine concentration, is one of most important tubular function. However, we are lacking of functional tubular parameters to explore this function. The only tool available to assess it currently, is urinary osmolarity that is an indirect and nonspecific maker of CMG. In this study, we explore the ability of 23NaMRI in measuring 1) the dynamics of CMG for the first time compared to urinary osmolarity after a water load 2) the CMG in kidney disease. Method We conducted an exploratory pilot study for 10 healthy controls with water load then 5 cardiorenal patients with kidney disease. 1) Healthy controls were asked to be fasting since midnight. Urines sample were collected to measure fasting osmolarity and a first MRIscan were performed to acquire baseline anatomical and sodium images. Once the baseline was completed, healthy participants were asked to ingest water (15 mL/kg) within 15 minutes. Four subsequent sodium pictures were acquired an hour after water ingestion. Urine samples were obtained after each sodium acquisition every 15 min during one hours. 2) Cardiorenal patients underwent an MRI scan, provided a spot urine sample and have blood work collected. All MR experiments were carried out on a GE MR750 3T (GE Healthcare, WI). A custom-built two-loop (18cm in diameter) butterfly radiofrequency surface coil tuned for 23Na frequency (33.786 MHz) was used to acquire renal 23Na images. Results Mean age of the 10 healthy controls was 41.8 ± 15.3 years, mean body mass index (BMI) was 24.3 ± 3.8 kg/m2. Mean water intake was 1092 ± 233 mL, total water excreted was 1250 ± 301 mL . Mean age of the 5 cardiorenal patients was 76.6 ± 12.2 years, mean BMI was 28.1 ± 6.9 kg/m2. eGFR was 54 ± 37 mL/min/1.73m2. Urinary osmolarity was 498 ± 145 mosm/L and medulla/cortex ratio was 1.35 ± 0.11. Sodium imaging was successfully acquired in all volunteers. In the morning fasting, medulla/cortex ratio was 1.55 ± 0.11 regarding to a urinary osmolarity to 814 ± 121 mosm/L. Mean ± SD fasting urinary osmolarity dropped significantly to 73 ± 14 mosm/L for maximal dilution, p=0.001. Mean medulla/cortex ratio dropped significantly to 1.31 ± 0.09 mosm/L for maximal dilution, p=0.002. Figure 1 displays changes of 23NaMRI pictures before (A) then 1h (B), 1H15 (C), 1h30 (D) and 1h45 (E) after a water load. Urinary osmolarity and medulla/cortex ratio are significantly correlated, r=0.54, p=0.0001. We measured corticomedullary gradient in cardiorenal patient with different level of eGFR to show the ability and feasibility to measure this gradient in pathological settings. We were able to measure medulla/cortex ratio in patients with CKD with a mean SNR of 20.45 ± 9.45. Conclusion We explored CMG dynamically every 15 min and we were able to discriminate significant changes after a water load. We were also able to provide efficient 23NaMRI pictures in cardiorenal patients with kidney disease. CMG exploration would provide a relevant assessment of tubular dysfunction independently of glomerular alteration and thus could be of prognostic value.


2020 ◽  
Vol 319 (2) ◽  
pp. F284-F291 ◽  
Author(s):  
Di Feng

Podocyte dysfunction contributes to proteinuric chronic kidney disease. A number of key proteins are essential for podocyte function, including nephrin, podocin, CD2-associated protein (CD2AP), synaptopodin, and α-actinin-4 (ACTN4). Although most of these proteins were first identified through genetic studies associated with human kidney disease, subsequent studies have identified phosphorylation of these proteins as an important posttranslational event that regulates their function. In this review, a brief overview of the function of these key podocyte proteins is provided. Second, the role of phosphorylation in regulating the function of these proteins is described. Third, the association between these phosphorylation pathways and kidney disease is reviewed. Finally, challenges and future directions in studying phosphorylation are discussed. Better characterization of these phosphorylation pathways and others yet to be discovered holds promise for translating this knowledge into new therapies for patients with proteinuric chronic kidney disease.


2018 ◽  
Author(s):  
Jennifer L. Harder ◽  
Rajasree Menon ◽  
Edgar A. Otto ◽  
Jian Zhou ◽  
Sean Eddy ◽  
...  

ABSTRACTPodocyte injury is central to many forms of kidney disease, but transcriptional signatures reflecting podocyte injury and compensation mechanisms are challenging to analyze in vivo. Human kidney organoids derived from pluripotent stem cells (PSCs), a new model for disease and regeneration, present an opportunity to explore the transcriptional plasticity of podocytes. Here, transcriptional profiling of over 12,000 single cells from human PSC-derived kidney organoid cultures was used to identify robust and reproducible cell-lineage gene expression signatures shared with developing human kidneys based on trajectory analysis. Surprisingly, the gene expression signature characteristic of developing glomerular epithelial cells was also observed in glomerular tissue from a kidney disease cohort. This signature correlated with proteinuria and inverse eGFR, and was confirmed in an independent podocytopathy cohort. Three genes in particular were further identified as critical components of the glomerular disease signature. We conclude that cells in human PSC-derived kidney organoids reliably recapitulate the developmental transcriptional program of podocytes and other cell lineages in the human kidney, and that the early transcriptional profile seen in developing podocytes is reactivated in glomerular disease. Our findings demonstrate an innovative approach to identifying novel molecular programs involved in the pathogenesis of glomerulopathies.


2021 ◽  
Vol 4 (1) ◽  
Author(s):  
Benjamin Freije ◽  
Ricardo Melo Ferreira ◽  
Ying-Hua Cheng ◽  
Samir Parikh ◽  
Michael Eadon

Background: Worldwide, one in eleven adults have diabetes mellitus and 30% to 40% will develop diabetic kidney disease (DKD). A mechanistic understanding of DKD is crucial to develop treatment strategies. To unravel DKD’s pathogenesis, single cell (scRNA) sequencing has proven a powerful tool, but is limited by a lack of localization. Spatial transcriptomics allows the mapping of scRNA sequencing data back to histology. Methods: Frozen human nephrectomy and biopsy samples were processed according to Visium spatial gene expression protocols, stained with H&E, and imaged. Samples were permeabilized for RNA capture, reverse transcribed and sequenced on an Illumina NovaSeq 6000. Mapping and counting were completed in Space Ranger and data was processed in Seurat. Samples were laser microdissected, protein was isolated, and protein was quantified by HPLC-MS. Results: Clusters from scRNAseq were mapped upon reference and DKD spatial transcriptomic images (N=4 reference, 2 DKD). Differentially expressed genes were identified in diabetic kidneys, including the upregulation of Adipocyte Enhancer Binding Protein (AEBP1).  Pathway analysis revealed enrichment of extracellular matrix organization and immune process pathways. To increase the confidence of these findings, glomeruli and the tubulointerstitium were laser microdissected (N=7 diseased, 4 reference) for proteomic analysis. AEBP1 was upregulated in the tubular interstitium of diseased kidneys and selectively upregulated in the glomeruli of Diabetic Nephropathy samples (N=2). AEBP1 localized to the interstitium by spatial transcriptomics and was expressed in highly fibrotic regions. Glomerular expression was not observed due to glomerulosclerosis. Conclusion: AEBP1 upregulation is a marker of interstitial fibrosis, with specific expression in the glomeruli of diabetic nephropathy specimens with glomerulosclerosis. Impact: This is the first study utilizing spatial transcriptomics to define and localize markers of human kidney disease. Confirmatory studies are required in larger sample sizes. AEBP1 is a previously unidentified marker of DKD previously associated with fibrosis in other organ-specific diseases.


2021 ◽  
pp. ASN.2020101442
Author(s):  
Michael Randles ◽  
Franziska Lausecker ◽  
Qing Kong ◽  
Hani Suleiman ◽  
Graeme Reid ◽  
...  

Background: Accumulation of extracellular matrix in organs and tissues is a feature of both aging and disease. In the kidney, glomerulosclerosis and tubulointerstitial fibrosis accompany the decline in function, which current therapies cannot address, leading to organ failure. Whilst histological and ultrastructural patterns of excess matrix form the basis of human disease classifications, comprehensive molecular resolution of abnormal matrix is lacking. Methods: Using mass spectrometry-based proteomics we resolved matrix composition over age in mouse models of kidney disease. We compared the changes in mice with a global characterization of human kidney matrix during aging and to existing kidney disease datasets to identify common molecular features. Results: Ultrastructural changes in basement membranes are associated with altered cell adhesion and metabolic processes and with distinct matrix proteomes during aging and kidney disease progression in mice. Within the altered matrix, basement membrane components (laminins, type IV collagen, type XVIII collagen) were reduced and interstitial matrix proteins (collagens I, III, VI, XV, fibrinogens and nephronectin) were increased, a pattern also seen in human kidney aging. Indeed, this signature of matrix proteins was consistently modulated across all age and disease comparisons and the increase in interstitial matrix was also observed in human kidney disease datasets. Conclusions: This study provides deep molecular resolution of matrix accumulation in kidney aging and disease and identifies a common signature of proteins that provides insight into mechanisms of response to kidney injury and repair.


Metabolites ◽  
2021 ◽  
Vol 11 (7) ◽  
pp. 460
Author(s):  
Ulla T. Schultheiss ◽  
Robin Kosch ◽  
Fruzsina Kotsis ◽  
Michael Altenbuchinger ◽  
Helena U. Zacharias

Kidney diseases still pose one of the biggest challenges for global health, and their heterogeneity and often high comorbidity load seriously hinders the unraveling of their underlying pathomechanisms and the delivery of optimal patient care. Metabolomics, the quantitative study of small organic compounds, called metabolites, in a biological specimen, is gaining more and more importance in nephrology research. Conducting a metabolomics study in human kidney disease cohorts, however, requires thorough knowledge about the key workflow steps: study planning, sample collection, metabolomics data acquisition and preprocessing, statistical/bioinformatics data analysis, and results interpretation within a biomedical context. This review provides a guide for future metabolomics studies in human kidney disease cohorts. We will offer an overview of important a priori considerations for metabolomics cohort studies, available analytical as well as statistical/bioinformatics data analysis techniques, and subsequent interpretation of metabolic findings. We will further point out potential research questions for metabolomics studies in the context of kidney diseases and summarize the main results and data availability of important studies already conducted in this field.


2020 ◽  
Vol 472 (4) ◽  
pp. 449-460 ◽  
Author(s):  
Sarah E. Motta ◽  
Pedro Henrique Imenez Silva ◽  
Arezoo Daryadel ◽  
Betül Haykir ◽  
Eva Maria Pastor-Arroyo ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document