Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Stem Cell Reports ; 19(6): 859-876, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38788724

ABSTRACT

Hepatocyte nuclear factor 1B (HNF1B) encodes a transcription factor expressed in developing human kidney epithelia. Heterozygous HNF1B mutations are the commonest monogenic cause of dysplastic kidney malformations (DKMs). To understand their pathobiology, we generated heterozygous HNF1B mutant kidney organoids from CRISPR-Cas9 gene-edited human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) reprogrammed from a family with HNF1B-associated DKMs. Mutant organoids contained enlarged malformed tubules displaying deregulated cell turnover. Numerous genes implicated in Mendelian kidney tubulopathies were downregulated, and mutant tubules resisted the cyclic AMP (cAMP)-mediated dilatation seen in controls. Bulk and single-cell RNA sequencing (scRNA-seq) analyses indicated abnormal Wingless/Integrated (WNT), calcium, and glutamatergic pathways, the latter hitherto unstudied in developing kidneys. Glutamate ionotropic receptor kainate type subunit 3 (GRIK3) was upregulated in malformed mutant nephron tubules and prominent in HNF1B mutant fetal human dysplastic kidney epithelia. These results reveal morphological, molecular, and physiological roles for HNF1B in human kidney tubule differentiation and morphogenesis illuminating the developmental origin of mutant-HNF1B-causing kidney disease.


Subject(s)
Hepatocyte Nuclear Factor 1-beta , Induced Pluripotent Stem Cells , Organoids , Humans , Hepatocyte Nuclear Factor 1-beta/genetics , Hepatocyte Nuclear Factor 1-beta/metabolism , Organoids/metabolism , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation/genetics , Heterozygote , Kidney Tubules/pathology , Kidney Tubules/metabolism , Mutation , Kidney/pathology , Kidney/metabolism , Kidney/abnormalities , CRISPR-Cas Systems , Pluripotent Stem Cells/metabolism , Gene Editing
2.
Cell Rep ; 42(12): 113502, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38032796

ABSTRACT

Optogenetics is a rapidly advancing technology combining photochemical, optical, and synthetic biology to control cellular behavior. Together, sensitive light-responsive optogenetic tools and human pluripotent stem cell differentiation models have the potential to fine-tune differentiation and unpick the processes by which cell specification and tissue patterning are controlled by morphogens. We used an optogenetic bone morphogenetic protein (BMP) signaling system (optoBMP) to drive chondrogenic differentiation of human embryonic stem cells (hESCs). We engineered light-sensitive hESCs through CRISPR-Cas9-mediated integration of the optoBMP system into the AAVS1 locus. The activation of optoBMP with blue light, in lieu of BMP growth factors, resulted in the activation of BMP signaling mechanisms and upregulation of a chondrogenic phenotype, with significant transcriptional differences compared to cells in the dark. Furthermore, cells differentiated with light could form chondrogenic pellets consisting of a hyaline-like cartilaginous matrix. Our findings indicate the applicability of optogenetics for understanding human development and tissue engineering.


Subject(s)
Optogenetics , Pluripotent Stem Cells , Humans , Chondrocytes , Cell Differentiation/genetics , Cartilage/metabolism , Chondrogenesis/genetics , Bone Morphogenetic Protein 2/metabolism , Cells, Cultured
3.
Nephron ; 145(3): 285-296, 2021.
Article in English | MEDLINE | ID: mdl-33774632

ABSTRACT

BACKGROUND: Kidney disease causes major suffering and premature mortality worldwide. With no cure for kidney failure currently available, and with limited options for treatment, there is an urgent need to develop effective pharmaceutical interventions to slow or prevent kidney disease progression. SUMMARY: In this review, we consider the feasibility of using human pluripotent stem cell-derived kidney tissues, or organoids, to model genetic kidney disease. Notable successes have been made in modelling genetic tubular diseases (e.g., cystinosis), polycystic kidney disease, and medullary cystic kidney disease. Organoid models have also been used to test novel therapies that ameliorate aberrant cell biology. Some progress has been made in modelling congenital glomerular disease, even though glomeruli within organoids are developmentally immature. Less progress has been made in modelling structural kidney malformations, perhaps because sufficiently mature metanephric mesenchyme-derived nephrons, ureteric bud-derived branching collecting ducts, and a prominent stromal cell population are not generated together within a single protocol. Key Messages: We predict that the field will advance significantly if organoids can be generated with a full complement of cell lineages and with kidney components displaying key physiological functions, such as glomerular filtration. The future economic upscaling of reproducible organoid generation will facilitate more widespread research applications, including the potential therapeutic application of these stem cell-based technologies.


Subject(s)
Kidney Diseases/genetics , Pluripotent Stem Cells/metabolism , Genetic Predisposition to Disease , Humans , Kidney Diseases/congenital , Kidney Diseases/pathology
4.
Stem Cell Reports ; 16(1): 134-148, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33306987

ABSTRACT

MicroRNAs (miRNAs) are gene expression regulators and they have been implicated in acquired kidney diseases and in renal development, mostly through animal studies. We hypothesized that the miR-199a/214 cluster regulates human kidney development. We detected its expression in human embryonic kidneys by in situ hybridization. To mechanistically study the cluster, we used 2D and 3D human embryonic stem cell (hESC) models of kidney development. After confirming expression in each model, we inhibited the miRNAs using lentivirally transduced miRNA sponges. This reduced the WT1+ metanephric mesenchyme domain in 2D cultures. Sponges did not prevent the formation of 3D kidney-like organoids. These organoids, however, contained dysmorphic glomeruli, downregulated WT1, aberrant proximal tubules, and increased interstitial capillaries. Thus, the miR-199a/214 cluster fine-tunes differentiation of both metanephric mesenchymal-derived nephrons and kidney endothelia. While clinical implications require further study, it is noted that patients with heterozygous deletions encompassing this miRNA locus can have malformed kidneys.


Subject(s)
Cell Differentiation , MicroRNAs/metabolism , Antagomirs/metabolism , Capillaries/pathology , Cell Culture Techniques , Down-Regulation , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Kidney Tubules, Proximal/blood supply , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Models, Biological , Neovascularization, Physiologic , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...