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.
Cell Rep ; 42(12): 113431, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38039961

ABSTRACT

In autosomal dominant polycystic kidney disease (ADPKD), renal cyst lesions predominantly arise from collecting ducts (CDs). However, relevant CD cyst models using human cells are lacking. Although previous reports have generated in vitro renal tubule cyst models from human induced pluripotent stem cells (hiPSCs), therapeutic drug candidates for ADPKD have not been identified. Here, by establishing expansion cultures of hiPSC-derived ureteric bud tip cells, an embryonic precursor that gives rise to CDs, we succeed in advancing the developmental stage of CD organoids and show that all CD organoids derived from PKD1-/- hiPSCs spontaneously develop multiple cysts, clarifying the initiation mechanisms of cystogenesis. Moreover, we identify retinoic acid receptor (RAR) agonists as candidate drugs that suppress in vitro cystogenesis and confirm the therapeutic effects on an ADPKD mouse model in vivo. Therefore, our in vitro CD cyst model contributes to understanding disease mechanisms and drug discovery for ADPKD.


Subject(s)
Cysts , Induced Pluripotent Stem Cells , Kidney Neoplasms , Polycystic Kidney, Autosomal Dominant , Mice , Animals , Humans , Induced Pluripotent Stem Cells/pathology , Kidney/pathology , Kidney Neoplasms/pathology , Organoids/pathology , Cysts/pathology , TRPP Cation Channels
2.
PLoS One ; 17(11): e0275600, 2022.
Article in English | MEDLINE | ID: mdl-36378656

ABSTRACT

Cell therapies using human induced pluripotent stem cell (hiPSC)-derived nephron progenitor cells (NPCs) are expected to ameliorate acute kidney injury (AKI). However, using hiPSC-derived NPCs clinically is a challenge because hiPSCs themselves are tumorigenic. LIN28A, ESRG, CNMD and SFRP2 transcripts have been used as a marker of residual hiPSCs for a variety of cell types undergoing clinical trials. In this study, by reanalyzing public databases, we found a baseline expression of LIN28A, ESRG, CNMD and SFRP2 in hiPSC-derived NPCs and several other cell types, suggesting LIN28A, ESRG, CNMD and SFRP2 are not always reliable markers for iPSC detection. As an alternative, we discovered a lncRNA marker gene, MIR302CHG, among many known and unknown iPSC markers, as highly differentially expressed between hiPSCs and NPCs, by RNA sequencing and quantitative RT-PCR (qRT-PCR) analyses. Using MIR302CHG as an hiPSC marker, we constructed two assay methods, a combination of magnetic bead-based enrichment and qRT-PCR and digital droplet PCR alone, to detect a small number of residual hiPSCs in NPC populations. The use of these in vitro assays could contribute to patient safety in treatments using hiPSC-derived cells.


Subject(s)
Induced Pluripotent Stem Cells , Neuroblastoma , RNA, Long Noncoding , Humans , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation/genetics , In Vitro Techniques , Nephrons , RNA, Long Noncoding/metabolism , Neuroblastoma/metabolism
3.
Sci Rep ; 11(1): 3936, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33594180

ABSTRACT

Erythropoietin (EPO) is a crucial hormone for erythropoiesis and produced by adult kidneys. Insufficient EPO production in chronic kidney disease (CKD) can cause renal anemia. Although hypoxia-inducible factors (HIFs) are known as a main regulator, the mechanisms of EPO production have not been fully elucidated. In this study, we aimed to examine the roles of retinoic acid (RA) in EPO production using EPO-producing cells derived from human induced pluripotent stem cells (hiPSC-EPO cells) that we previously established. RA augmented EPO production by hiPSC-EPO cells under hypoxia or by treatment with prolyl hydroxylase domain-containing protein (PHD) inhibitors that upregulate HIF signals. Combination treatment with RA and a PHD inhibitor improved renal anemia in vitamin A-depleted CKD model mice. Our findings using hiPSC-EPO cells and CKD model mice may contribute to clarifying the EPO production mechanism and developing efficient therapies for renal anemia.


Subject(s)
Anemia/drug therapy , Erythropoietin/biosynthesis , Glycine/analogs & derivatives , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Isoquinolines/therapeutic use , Tretinoin/therapeutic use , Anemia/etiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Drug Evaluation, Preclinical , Drug Therapy, Combination , Glycine/therapeutic use , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Induced Pluripotent Stem Cells , Kidney Diseases/complications , Male , Mice , Mice, Inbred C57BL , Tretinoin/pharmacology
4.
Sci Transl Med ; 9(409)2017 Sep 27.
Article in English | MEDLINE | ID: mdl-28954928

ABSTRACT

The production of erythropoietin (EPO) by the kidneys, a principal hormone for the hematopoietic system, is reduced in patients with chronic kidney disease (CKD), eventually resulting in severe anemia. Although recombinant human EPO treatment improves anemia in patients with CKD, returning to full red blood cell production without fluctuations does not always occur. We established a method to generate EPO-producing cells from human induced pluripotent stem cells (hiPSCs) by modifying previously reported hepatic differentiation protocols. These cells showed increased EPO expression and secretion in response to low oxygen conditions, prolyl hydroxylase domain-containing enzyme inhibitors, and insulin-like growth factor 1. The EPO protein secreted from hiPSC-derived EPO-producing (hiPSC-EPO) cells induced the erythropoietic differentiation of human umbilical cord blood progenitor cells in vitro. Furthermore, transplantation of hiPSC-EPO cells into mice with CKD induced by adenine treatment improved renal anemia. Thus, hiPSC-EPO cells may be a useful tool for clarifying the mechanisms of EPO production and may be useful as a therapeutic strategy for treating renal anemia.


Subject(s)
Anemia/therapy , Erythropoietin/biosynthesis , Kidney/pathology , Pluripotent Stem Cells/cytology , Stem Cell Transplantation , Anemia/pathology , Animals , Cell Differentiation/drug effects , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Erythropoiesis/drug effects , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/ultrastructure , Insulin-Like Growth Factor I/pharmacology , Mice , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/ultrastructure , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...