Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(2): 113602, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38237600

ABSTRACT

Recent regenerative studies using human pluripotent stem cells (hPSCs) have developed multiple kidney-lineage cells and organoids. However, to further form functional segments of the kidney, interactions of epithelial and interstitial cells are required. Here we describe a selective differentiation of renal interstitial progenitor-like cells (IPLCs) from human induced pluripotent stem cells (hiPSCs) by modifying our previous induction method for nephron progenitor cells (NPCs) and analyzing mouse embryonic interstitial progenitor cell (IPC) development. Our IPLCs combined with hiPSC-derived NPCs and nephric duct cells form nephrogenic niche- and mesangium-like structures in vitro. Furthermore, we successfully induce hiPSC-derived IPLCs to differentiate into mesangial and erythropoietin-producing cell lineages in vitro by screening differentiation-inducing factors and confirm that p38 MAPK, hypoxia, and VEGF signaling pathways are involved in the differentiation of mesangial-lineage cells. These findings indicate that our IPC-lineage induction method contributes to kidney regeneration and developmental research.


Subject(s)
Erythropoietin , Induced Pluripotent Stem Cells , Humans , Animals , Mice , Kidney , Cell Lineage , Regeneration
2.
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
3.
Commun Biol ; 6(1): 854, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37770589

ABSTRACT

Alport syndrome (AS) is a hereditary glomerulonephritis caused by COL4A3, COL4A4 or COL4A5 gene mutations and characterized by abnormalities of glomerular basement membranes (GBMs). Due to a lack of curative treatments, the condition proceeds to end-stage renal disease even in adolescents. Hampering drug discovery is the absence of effective in vitro methods for testing the restoration of normal GBMs. Here, we aimed to develop kidney organoid models from AS patient iPSCs for this purpose. We established iPSC-derived collagen α5(IV)-expressing kidney organoids and confirmed that kidney organoids from COL4A5 mutation-corrected iPSCs restore collagen α5(IV) protein expression. Importantly, our model recapitulates the differences in collagen composition between iPSC-derived kidney organoids from mild and severe AS cases. Furthermore, we demonstrate that a chemical chaperone, 4-phenyl butyric acid, has the potential to correct GBM abnormalities in kidney organoids showing mild AS phenotypes. This iPSC-derived kidney organoid model will contribute to drug discovery for AS.


Subject(s)
Induced Pluripotent Stem Cells , Nephritis, Hereditary , Adolescent , Humans , Nephritis, Hereditary/genetics , Nephritis, Hereditary/metabolism , Nephritis, Hereditary/therapy , Collagen Type IV/genetics , Collagen Type IV/metabolism , Induced Pluripotent Stem Cells/metabolism , Kidney/metabolism , Glomerular Basement Membrane
4.
Drug Metab Dispos ; 51(9): 1177-1187, 2023 09.
Article in English | MEDLINE | ID: mdl-37385755

ABSTRACT

The proximal tubule plays an important role in the kidney and is a major site of drug interaction and toxicity. Analysis of kidney toxicity via in vitro assays is challenging, because only a few assays that reflect functions of drug transporters in renal proximal tubular epithelial cells (RPTECs) are available. In this study, we aimed to develop a simple and reproducible method for culturing RPTECs by monitoring organic anion transporter 1 (OAT1) as a selection marker. Culturing RPTECs in spherical cellular aggregates increased OAT1 protein expression, which was low in the conventional two-dimensional (2D) culture, to a level similar to that in human renal cortices. By proteome analysis, it was revealed that the expression of representative two proximal tubule markers was maintained and 3D spheroid culture improved the protein expression of approximately 7% of the 139 transporter proteins detected, and the expression of 2.3% of the 4,800 proteins detected increased by approximately fivefold that in human renal cortices. Furthermore, the expression levels of approximately 4,800 proteins in three-dimensional (3D) RPTEC spheroids (for 12 days) were maintained for over 20 days. Cisplatin and adefovir exhibited transporter-dependent ATP decreases in 3D RPTEC spheroids. These results indicate that the 3D RPTEC spheroids developed by monitoring OAT1 gene expression are a simple and reproducible in vitro experimental system with improved gene and protein expressions compared with 2D RPTECs and were more similar to that in human kidney cortices. Therefore, it can potentially be used for evaluating human renal proximal tubular toxicity and drug disposition. SIGNIFICANCE STATEMENT: This study developed a simple and reproducible spheroidal culture method with acceptable throughput using commercially available RPTECs by monitoring OAT1 gene expression. RPTECs cultured using this new method showed improved mRNA/protein expression profiles to those in 2D RPTECs and were more similar to those of human kidney cortices. This study provides a potential in vitro proximal tubule system for pharmacokinetic and toxicological evaluations during drug development.


Subject(s)
Kidney , Organic Anion Transport Protein 1 , Humans , Kidney/metabolism , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Kidney Tubules, Proximal/metabolism , Membrane Transport Proteins/metabolism , Gene Expression , Epithelial Cells/metabolism
5.
Sci Rep ; 13(1): 8659, 2023 05 29.
Article in English | MEDLINE | ID: mdl-37248264

ABSTRACT

For pluripotent stem cell (PSC)-based regenerative therapy against diabetes, the differentiation efficiency to pancreatic lineage cells needs to be improved based on the mechanistic understanding of pancreatic differentiation. Here, we aimed to elucidate the molecular mechanisms underlying pancreatic endoderm differentiation by searching for factors that regulate a crucial pancreatic endoderm marker gene, NKX6.1. Unbiasedly screening an siRNA knockdown library, we identified a candidate transcription factor, HHEX. HHEX knockdown suppressed the expression of another pancreatic endoderm marker gene, PTF1A, as well as NKX6.1, independently of PDX1, a known regulator of NKX6.1 expression. In contrast, the overexpression of HHEX upregulated the expressions of NKX6.1 and PTF1A. RNA-seq analysis showed decreased expressions of several genes related to pancreatic development, such as NKX6.1, PTF1A, ONECUT1 and ONECUT3, in HHEX knockdown pancreatic endoderm. These results suggest that HHEX plays a key role in pancreatic endoderm differentiation.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Endoderm , Trans-Activators/genetics , Trans-Activators/metabolism , Cell Differentiation/genetics , Pancreas/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
6.
STAR Protoc ; 3(3): 101484, 2022 09 16.
Article in English | MEDLINE | ID: mdl-35769929

ABSTRACT

The ureteric bud (UB) is a kidney precursor tissue that repeats branching morphogenesis and gives rise to the collecting ducts (CDs) and lower urinary tract. Here, we describe protocols to generate iUB organoids from human iPSCs; iUB organoids repeat branching morphogenesis. We describe how to expand iUB-organoid-derived tip colonies and how to induce CD progenitors from iUB organoids. These organoids can be used to study CD development and potentially as a model of kidney and urinary tract diseases. For complete details on the use and execution of this protocol, please refer to Mae et al. (2020).


Subject(s)
Induced Pluripotent Stem Cells , Organoids , Humans , Kidney , Morphogenesis
7.
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
8.
Biochem Biophys Res Commun ; 529(4): 1186-1194, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32819584

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is a hereditary disorder which manifests progressive renal cyst formation and leads to end-stage kidney disease. Around 85% of cases are caused by PKD1 heterozygous mutations, exhibiting relatively poorer renal outcomes than those with mutations in other causative gene PKD2. Although many disease models have been proposed for ADPKD, the pre-symptomatic pathology of the human disease remains unknown. To unveil the mechanisms of early cytogenesis, robust and genetically relevant human models are needed. Here, we report a novel ADPKD model using kidney organoids derived from disease-specific human induced pluripotent stem cells (hiPSCs). Importantly, we found that kidney organoids differentiated from gene-edited heterozygous PKD1-mutant as well as ADPKD patient-derived hiPSCs can reproduce renal cysts. Further, we demonstrated the possibility of ADPKD kidney organoids serving as drug screening platforms. This newly developed model will contribute to identifying novel therapeutic targets, extending the field of ADPKD research.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Kidney/pathology , Models, Biological , Organoids/pathology , Polycystic Kidney, Autosomal Dominant/pathology , Amino Acid Sequence , Base Sequence , Cell Differentiation/drug effects , Cell Line , Colforsin/pharmacology , Drug Evaluation, Preclinical , Humans , Induced Pluripotent Stem Cells/drug effects , Mutation/genetics , Phenotype , TRPP Cation Channels/chemistry , TRPP Cation Channels/genetics
9.
Cell Rep ; 32(4): 107963, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32726627

ABSTRACT

Ureteric bud (UB) is the embryonic kidney progenitor tissue that gives rise to the collecting duct and lower urinary tract. UB-like structures generated from human pluripotent stem cells by previously reported methods show limited developmental ability and limited branching. Here we report a method to generate UB organoids that possess epithelial polarity and tubular lumen and repeat branching morphogenesis. We also succeed in monitoring UB tip cells by utilizing the ability of tip cells to uptake very-low-density lipoprotein, cryopreserving UB progenitor cells, and expanding UB tip cells that can reconstitute the organoids and differentiate into collecting duct progenitors. Moreover, we successfully reproduce some phenotypes of multicystic dysplastic kidney (MCDK) using the UB organoids. These methods will help elucidate the developmental mechanisms of UB branching and develop a selective differentiation method for collecting duct cells, contributing to the creation of disease models for congenital renal abnormalities.


Subject(s)
Kidney Tubules, Collecting/embryology , Tissue Culture Techniques/methods , Urinary Tract/embryology , Cell Differentiation/physiology , Humans , Induced Pluripotent Stem Cells/metabolism , Kidney/embryology , Morphogenesis , Organogenesis/physiology , Organoids/metabolism , Pluripotent Stem Cells/metabolism
10.
Cell Rep ; 31(1): 107476, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32268094

ABSTRACT

Recent studies using human pluripotent stem cells (hPSCs) have developed protocols to induce kidney-lineage cells and reconstruct kidney organoids. However, the separate generation of metanephric nephron progenitors (NPs), mesonephric NPs, and ureteric bud (UB) cells, which constitute embryonic kidneys, in in vitro differentiation culture systems has not been fully investigated. Here, we create a culture system in which these mesoderm-like cell types and paraxial and lateral plate mesoderm-like cells are separately generated from hPSCs. We recapitulate nephrogenic niches from separately induced metanephric NP-like and UB-like cells, which are subsequently differentiated into glomeruli, renal tubules, and collecting ducts in vitro and further vascularized in vivo. Our selective differentiation protocols should contribute to understanding the mechanisms underlying human kidney development and disease and also supply cell sources for regenerative therapies.


Subject(s)
Cell Culture Techniques/methods , Cell Lineage/physiology , Pluripotent Stem Cells/cytology , Cell Differentiation/physiology , Cells, Cultured , Epithelial Cells , Humans , Kidney/cytology , Mesoderm , Nephrons , Organogenesis/physiology , Organoids/cytology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology
11.
Sci Rep ; 9(1): 10701, 2019 Jul 18.
Article in English | MEDLINE | ID: mdl-31316087

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

12.
Nihon Yakurigaku Zasshi ; 153(2): 61-66, 2019.
Article in Japanese | MEDLINE | ID: mdl-30745515

ABSTRACT

Each of the billions of the cells in our body exhibits their identity with unique gene expression profile. Recent advances in single cell transcriptomics enable to conduct cell taxonomy identifying new cell types and to re-arrange cells in order of pseudo-time course describing differentiation status of each cell. Even though the cost is still high, the single cell transcriptomics now becomes one of the conventional assays. We have applied the single cell gene expression analysis to dissect human development. In this article, we show our recent progress on a study describing early development of the kidney using human iPS cells by the single cell transcriptomics.


Subject(s)
Gene Expression Profiling , Induced Pluripotent Stem Cells/cytology , Kidney/embryology , Single-Cell Analysis , Cell Differentiation , Humans , Transcriptome
13.
Methods Mol Biol ; 1926: 117-123, 2019.
Article in English | MEDLINE | ID: mdl-30742267

ABSTRACT

The generation of ureteric bud (UB), which is the renal progenitor that gives rise to renal collecting ducts and lower urinary tract, from human-induced pluripotent stem cells (hiPSCs) provides a cell source for studying the development of UB and kidney disease. Here we describe a stepwise and efficient two-dimensional differentiation method of hiPSCs into Wolffian duct (WD) cells. We also describe how to generate three-dimensional WD epithelial structures that can differentiate into UB-like structures.


Subject(s)
Embryo, Mammalian/cytology , Induced Pluripotent Stem Cells/cytology , Kidney/cytology , Ureter/cytology , Animals , Cell Differentiation/physiology , Gene Expression Regulation, Developmental , Humans , Mesoderm/cytology
14.
Stem Cell Res ; 35: 101400, 2019 03.
Article in English | MEDLINE | ID: mdl-30735882

ABSTRACT

Cholangiocytes are the epithelial cells that line bile ducts, and ductal plate malformation is a developmental anomaly of bile ducts that causes severe congenital biliary disorders. However, because of a lack of specific marker genes, methods for the stepwise differentiation and isolation of human induced pluripotent stem cell (hiPSC)-derived cholangiocyte progenitors at ductal plate stages have not been established. We herein generated an AQP1-GFP reporter hiPSC line and developed a combination treatment with transforming growth factor (TGF) ß2 and epidermal growth factor (EGF) to induce hiPSC-derived hepatoblasts into AQP1+ cells in vitro. By confirming that the isolated AQP1+ cells showed similar gene expression patterns to cholangiocyte progenitors at the remodeling ductal plate stage around gestational week (GW) 20, we established a differentiation protocol from hiPSCs through SOX9+CK19+AQP1- ductal plate-like cells into SOX9+CK19+AQP1+ remodeling ductal plate-like cells. We further generated 3D bile duct-like structures from the induced ductal plate-like cells. These results suggest that AQP1 is a useful marker for the generation of remodeling ductal plate cells from hiPSCs. Our methods may contribute to elucidating the differentiation mechanisms of ductal plate cells and the pathogenesis of ductal plate malformation.


Subject(s)
Aquaporin 1 , Bile Ducts , Epithelial Cells , Green Fluorescent Proteins , Induced Pluripotent Stem Cells , Aquaporin 1/biosynthesis , Aquaporin 1/genetics , Bile Ducts/abnormalities , Bile Ducts/metabolism , Bile Ducts/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology
15.
Sci Rep ; 8(1): 6375, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29686294

ABSTRACT

Cell therapy using renal progenitors differentiated from human embryonic stem cells (hESCs) or induced pluripotent stem cells (hiPSCs) has the potential to significantly reduce the number of patients receiving dialysis therapy. However, the differentiation cultures may contain undifferentiated or undesired cell types that cause unwanted side effects, such as neoplastic formation, when transplanted into a body. Moreover, the hESCs/iPSCs are often genetically modified in order to isolate the derived renal progenitors, hampering clinical applications. To establish an isolation method for renal progenitors induced from hESCs/iPSCs without genetic modifications, we screened antibodies against cell surface markers. We identified the combination of four markers, CD9-CD140a+CD140b+CD271+, which could enrich OSR1+SIX2+ renal progenitors. Furthermore, these isolated cells ameliorated renal injury in an acute kidney injury (AKI) mouse model when used for cell therapy. These cells could contribute to the development of hiPSC-based cell therapy and disease modeling against kidney diseases.


Subject(s)
Acute Kidney Injury/therapy , Biomarkers/metabolism , Cell Separation/methods , Cell- and Tissue-Based Therapy/methods , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Stem Cell Transplantation/methods , Acute Kidney Injury/metabolism , Animals , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice
16.
Biochem Biophys Res Commun ; 495(1): 954-961, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29158085

ABSTRACT

Recent progress in kidney regeneration research is noteworthy. However, the selective and robust differentiation of the ureteric bud (UB), an embryonic renal progenitor, from human pluripotent stem cells (hPSCs) remains to be established. The present study aimed to establish a robust induction method for branching UB tissue from hPSCs towards the creation of renal disease models. Here, we found that anterior intermediate mesoderm (IM) differentiates from anterior primitive streak, which allowed us to successfully develop an efficient two-dimensional differentiation method of hPSCs into Wolffian duct (WD) cells. We also established a simplified procedure to generate three-dimensional WD epithelial structures that can form branching UB tissues. This system may contribute to hPSC-based regenerative therapies and disease models for intractable disorders arising in the kidney and lower urinary tract.


Subject(s)
Cell Differentiation/physiology , Pluripotent Stem Cells/physiology , Regeneration/physiology , Tissue Engineering/methods , Ureter/cytology , Ureter/growth & development , Cells, Cultured , Humans , Pluripotent Stem Cells/cytology
17.
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
18.
Circ J ; 81(1): 110-118, 2016 Dec 22.
Article in English | MEDLINE | ID: mdl-27867156

ABSTRACT

BACKGROUND: Approximately 10-20% of Kawasaki disease (KD) patients are resistant to intravenous immunoglobulin (IVIG) treatment. Further, these patients are at a particularly high risk of having coronary artery abnormalities. The mechanisms of IVIG resistance in KD have been analyzed using patient leukocytes, but not patient vascular endothelial cells (ECs). The present study clarifies the mechanisms of IVIG resistance in KD using an induced pluripotent stem cell (iPSC) disease model.Methods and Results:Dermal fibroblasts or peripheral blood mononuclear cells from 2 IVIG-resistant and 2 IVIG-responsive KD patients were reprogrammed by the episomal vector-mediated transduction of 6 reprogramming factors. KD patient-derived iPSCs were differentiated into ECs (iPSC-ECs). The gene expression profiles of iPSC-ECs generated from IVIG-resistant and IVIG-responsive KD patients were compared by RNA-sequencing analyses. We found that the expression ofCXCL12was significantly upregulated in iPSC-ECs from IVIG-resistant KD patients. Additionally, Gene Set Enrichment Analysis (GSEA) revealed that gene sets involved in interleukin (IL)-6 signaling were also upregulated. CONCLUSIONS: The first iPSC-based model for KD is reported here. Our mechanistic analyses suggest thatCXCL12, which plays a role in leukocyte transmigration, is a key molecule candidate for IVIG resistance and KD severity. They also indicate that an upregulation of IL-6-related genes may be involved in this pathogenesis.


Subject(s)
Drug Resistance , Immunoglobulins, Intravenous , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Mucocutaneous Lymph Node Syndrome/metabolism , Transcription, Genetic , Adolescent , Cells, Cultured , Chemokine CXCL12/biosynthesis , Child , Female , Humans , Induced Pluripotent Stem Cells/pathology , Interleukin-6/biosynthesis , Male , Mucocutaneous Lymph Node Syndrome/pathology
19.
Sci Rep ; 6: 30013, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27418197

ABSTRACT

Cardiovascular complications are the leading cause of death in autosomal dominant polycystic kidney disease (ADPKD), and intracranial aneurysm (ICA) causing subarachnoid hemorrhage is among the most serious complications. The diagnostic and therapeutic strategies for ICAs in ADPKD have not been fully established. We here generated induced pluripotent stem cells (iPSCs) from seven ADPKD patients, including four with ICAs. The vascular cells differentiated from ADPKD-iPSCs showed altered Ca(2+) entry and gene expression profiles compared with those of iPSCs from non-ADPKD subjects. We found that the expression level of a metalloenzyme gene, matrix metalloproteinase (MMP) 1, was specifically elevated in iPSC-derived endothelia from ADPKD patients with ICAs. Furthermore, we confirmed the correlation between the serum MMP1 levels and the development of ICAs in 354 ADPKD patients, indicating that high serum MMP1 levels may be a novel risk factor. These results suggest that cellular disease models with ADPKD-specific iPSCs can be used to study the disease mechanisms and to identify novel disease-related molecules or risk factors.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Intracranial Aneurysm/pathology , Matrix Metalloproteinase 1/blood , Polycystic Kidney, Autosomal Dominant/pathology , Subarachnoid Hemorrhage/pathology , Aged , Animals , Biomarkers/blood , Cell Differentiation , Cells, Cultured , DNA Methylation/genetics , Female , Humans , Intracranial Aneurysm/blood , Male , Matrix Metalloproteinase 1/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Polycystic Kidney, Autosomal Dominant/mortality , Risk Factors , TRPP Cation Channels/genetics
20.
Differentiation ; 92(5): 281-290, 2016 12.
Article in English | MEDLINE | ID: mdl-27087651

ABSTRACT

Many reports have described methods that induce definitive endoderm (DE) cells from human pluripotent stem cells (hPSCs). However, it is unclear whether the differentiation propensity of these DE cells is uniform. This uncertainty is due to the different developmental stages that give rise to anterior and posterior DE from anterior primitive streak (APS). Therefore, these DE cell populations might be generated from the different stages of APS cells, which affect the DE cell differentiation potential. Here, we succeeded in selectively differentiating early and late APS cells from human induced pluripotent stem cells (hiPSCs) using different concentrations of CHIR99021, a small molecule Wnt/ß-catenin pathway activator. We also established novel differentiation systems from hiPSCs into three types of DE cells: anterior and posterior domains of anterior DE cells through early APS cells and posterior DE cells through late APS cells. These different DE cell populations could differentiate into distinct endodermal lineages in vitro, such as lung, liver or small intestine progenitors. These results indicate that different APS cells can produce distinct types of DE cells that have proper developmental potency and suggest a method to evaluate the quality of endodermal cell induction from hPSCs.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Endoderm/growth & development , Induced Pluripotent Stem Cells/drug effects , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Lineage/genetics , Embryonic Stem Cells/drug effects , Endoderm/drug effects , Gene Expression Regulation, Developmental/drug effects , Humans , Induced Pluripotent Stem Cells/cytology , Intestine, Small/growth & development , Liver/growth & development , Lung/growth & development , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Wnt Signaling Pathway/drug effects , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...