Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 129
Filtrer
1.
Eur J Cell Biol ; 103(4): 151450, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39137450

RÉSUMÉ

Over the past decade, the induction protocols for the two types of kidney organoids (nephron organoids and ureteric bud organoids) from pluripotent stem cells (PSCs) have been established based on the knowledge gained in developmental nephrology. Kidney organoids are now used for disease modeling and drug screening, but they also have potential as tools for clinical transplantation therapy. One of the options to achieve this goal would be to assemble multiple renal progenitor cells (nephron progenitor, ureteric bud, stromal progenitor) to reproduce the organotypic kidney structure from PSCs. At least from mouse PSCs, all the three progenitors have been induced and assembled into such "higher order" kidney organoids. We will provide an overview of the developmental nephrology required for the induction of renal progenitors and discuss recent advances and remaining challenges of kidney organoids for clinical transplantation therapy.

2.
Proc Natl Acad Sci U S A ; 121(31): e2404193121, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39042698

RÉSUMÉ

Hematopoietic stem cells (HSCs) develop from hemogenic endothelial cells (HECs) in vivo during mouse embryogenesis. When cultured in vitro, cells from the embryo phenotypically defined as pre-HSC-I and pre-HSC-II have the potential to differentiate into HSCs. However, minimal factors required for HSC induction from HECs have not yet been determined. In this study, we demonstrated that stem cell factor (SCF) and thrombopoietin (TPO) induced engrafting HSCs from embryonic day (E) 11.5 pre-HSC-I in a serum-free and feeder-free culture condition. In contrast, E10.5 pre-HSC-I and HECs required an endothelial cell layer in addition to SCF and TPO to differentiate into HSCs. A single-cell RNA sequencing analysis of E10.5 to 11.5 dorsal aortae with surrounding tissues and fetal livers detected TPO expression confined in hepatoblasts, while SCF was expressed in various tissues, including endothelial cells and hepatoblasts. Our results suggest a transition of signal requirement during HSC development from HECs. The differentiation of E10.5 HECs to E11.5 pre-HSC-I in the aorta-gonad-mesonephros region depends on SCF and endothelial cell-derived factors. Subsequently, SCF and TPO drive the differentiation of E11.5 pre-HSC-I to pre-HSC-II/HSCs in the fetal liver. The culture system established in this study provides a beneficial tool for exploring the molecular mechanisms underlying the development of HSCs from HECs.


Sujet(s)
Différenciation cellulaire , Hémangioblastes , Cellules souches hématopoïétiques , Facteur de croissance des cellules souches , Thrombopoïétine , Animaux , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/cytologie , Souris , Thrombopoïétine/métabolisme , Facteur de croissance des cellules souches/métabolisme , Hémangioblastes/métabolisme , Hémangioblastes/cytologie , Cellules endothéliales/métabolisme , Cellules endothéliales/cytologie , Transduction du signal , Hématopoïèse/physiologie , Développement embryonnaire , Embryon de mammifère/métabolisme , Embryon de mammifère/cytologie , Foie/embryologie , Foie/métabolisme , Foie/cytologie
3.
Development ; 151(5)2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38345319

RÉSUMÉ

The trunk axial skeleton develops from paraxial mesoderm cells. Our recent study demonstrated that conditional knockout of the stem cell factor Sall4 in mice by TCre caused tail truncation and a disorganized axial skeleton posterior to the lumbar level. Based on this phenotype, we hypothesized that, in addition to the previously reported role of Sall4 in neuromesodermal progenitors, Sall4 is involved in the development of the paraxial mesoderm tissue. Analysis of gene expression and SALL4 binding suggests that Sall4 directly or indirectly regulates genes involved in presomitic mesoderm differentiation, somite formation and somite differentiation. Furthermore, ATAC-seq in TCre; Sall4 mutant posterior trunk mesoderm shows that Sall4 knockout reduces chromatin accessibility. We found that Sall4-dependent open chromatin status drives activation and repression of WNT signaling activators and repressors, respectively, to promote WNT signaling. Moreover, footprinting analysis of ATAC-seq data suggests that Sall4-dependent chromatin accessibility facilitates CTCF binding, which contributes to the repression of neural genes within the mesoderm. This study unveils multiple mechanisms by which Sall4 regulates paraxial mesoderm development by directing activation of mesodermal genes and repression of neural genes.


Sujet(s)
Protéines de liaison à l'ADN , Régulation de l'expression des gènes au cours du développement , Mésoderme , Facteurs de transcription , Animaux , Souris , Différenciation cellulaire , Chromatine/métabolisme , Expression des gènes , Mésoderme/métabolisme , Somites/métabolisme , Protéines de liaison à l'ADN/métabolisme , Facteurs de transcription/métabolisme
4.
Genetics ; 227(1)2024 05 07.
Article de Anglais | MEDLINE | ID: mdl-38386912

RÉSUMÉ

Vertebrate limbs start to develop as paired protrusions from the lateral plate mesoderm at specific locations of the body with forelimb buds developing anteriorly and hindlimb buds posteriorly. During the initiation process, limb progenitor cells maintain active proliferation to form protrusions and start to express Fgf10, which triggers molecular processes for outgrowth and patterning. Although both processes occur in both types of limbs, forelimbs (Tbx5), and hindlimbs (Isl1) utilize distinct transcriptional systems to trigger their development. Here, we report that Sall1 and Sall4, zinc finger transcription factor genes, regulate hindlimb initiation in mouse embryos. Compared to the 100% frequency loss of hindlimb buds in TCre; Isl1 conditional knockouts, Hoxb6Cre; Isl1 conditional knockout causes a hypomorphic phenotype with only approximately 5% of mutants lacking the hindlimb. Our previous study of SALL4 ChIP-seq showed SALL4 enrichment in an Isl1 enhancer, suggesting that SALL4 acts upstream of Isl1. Removing 1 allele of Sall4 from the hypomorphic Hoxb6Cre; Isl1 mutant background caused loss of hindlimbs, but removing both alleles caused an even higher frequency of loss of hindlimbs, suggesting a genetic interaction between Sall4 and Isl1. Furthermore, TCre-mediated conditional double knockouts of Sall1 and Sall4 displayed a loss of expression of hindlimb progenitor markers (Isl1, Pitx1, Tbx4) and failed to develop hindlimbs, demonstrating functional redundancy between Sall1 and Sall4. Our data provides genetic evidence that Sall1 and Sall4 act as master regulators of hindlimb initiation.


Sujet(s)
Protéines de liaison à l'ADN , Régulation de l'expression des gènes au cours du développement , Membre pelvien , Protéines à homéodomaine LIM , Facteurs de transcription , Animaux , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Souris , Membre pelvien/embryologie , Membre pelvien/métabolisme , Protéines à homéodomaine LIM/génétique , Protéines à homéodomaine LIM/métabolisme , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/métabolisme , Bourgeons de membre/métabolisme , Bourgeons de membre/embryologie , Souris knockout , Embryon de mammifère/métabolisme , Facteur de croissance fibroblastique de type 10/génétique , Facteur de croissance fibroblastique de type 10/métabolisme , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme
5.
Cell Stem Cell ; 30(8): 1017-1027, 2023 08 03.
Article de Anglais | MEDLINE | ID: mdl-37541208

RÉSUMÉ

Kidney organoids have enabled modeling of human development and disease. While methods of generating the nephron lineage are well established, new protocols to induce another lineage, the ureteric bud/collecting duct, have been reported in the past 5 years. Many reports have described modeling of various hereditary kidney diseases, with polycystic kidney disease serving as the archetypal disease, by using patient-derived or genome-edited kidney organoids. The generation of more organotypic kidneys is also becoming feasible. In this review, I also discuss the significant challenges for more sophisticated disease modeling and for realizing the ambitious goal of generating transplantable synthetic kidneys.


Sujet(s)
Rein , Organoïdes , Humains , Différenciation cellulaire
6.
Dev Biol ; 501: 28-38, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37301463

RÉSUMÉ

Recent studies illustrate the importance of regulation of cellular metabolism, especially glycolysis and pathways branching from glycolysis, during vertebrate embryo development. For example, glycolysis generates cellular energy ATP. Glucose carbons are also directed to the pentose phosphate pathway, which is needed to sustain anabolic processes in the rapidly growing embryos. However, our understanding of the exact status of glycolytic metabolism as well as genes that regulate glycolytic metabolism are still incomplete. Sall4 is a zinc finger transcription factor that is highly expressed in undifferentiated cells in developing mouse embryos, such as blastocysts and the post-implantation epiblast. TCre; Sall4 conditional knockout mouse embryos exhibit various defects in the posterior part of the body, including hindlimbs. Using transcriptomics approaches, we found that many genes encoding glycolytic enzymes are upregulated in the posterior trunk, including the hindlimb-forming region, of Sall4 conditional knockout mouse embryos. In situ hybridization and qRT-PCR also confirmed upregulation of expression of several glycolytic genes in hindlimb buds. A fraction of those genes are bound by SALL4 at the promoters, gene bodies or distantly-located regions, suggesting that Sall4 directly regulates expression of several glycolytic enzyme genes in hindlimb buds. To further gain insight into the metabolic status associated with the observed changes at the transcriptional level, we performed a comprehensive analysis of metabolite levels in limb buds in wild type and Sall4 conditional knockout embryos by high-resolution mass spectrometry. We found that the levels of metabolic intermediates of glycolysis are lower, but glycolytic end-products pyruvate and lactate did not exhibit differences in Sall4 conditional knockout hindlimb buds. The increased expression of glycolytic genes would have caused accelerated glycolytic flow, resulting in low levels of intermediates. This condition may have prevented intermediates from being re-directed to other pathways, such as the pentose phosphate pathway. Indeed, the change in glycolytic metabolite levels is associated with reduced levels of ATP and metabolites of the pentose phosphate pathway. To further test whether glycolysis regulates limb patterning downstream of Sall4, we conditionally inactivated Hk2, which encodes a rate-limiting enzyme gene in glycolysis and is regulated by Sall4. The TCre; Hk2 conditional knockout hindlimb exhibited a short femur, and a lack of tibia and anterior digits in hindlimbs, which are defects similarly found in the TCre; Sall4 conditional knockout. The similarity of skeletal defects in Sall4 mutants and Hk2 mutants suggests that regulation of glycolysis plays a role in hindlimb patterning. These data suggest that Sall4 restricts glycolysis in limb buds and contributes to patterning and regulation of glucose carbon flow during development of limb buds.


Sujet(s)
Régulation de l'expression des gènes au cours du développement , Bourgeons de membre , Animaux , Souris , Adénosine triphosphate/métabolisme , Glucose/métabolisme , Glycolyse/génétique , Bourgeons de membre/métabolisme , Souris knockout
8.
Front Med (Lausanne) ; 10: 1089159, 2023.
Article de Anglais | MEDLINE | ID: mdl-37035301

RÉSUMÉ

Introduction: Mutations in ADAMTS9 cause nephronophthisis-related ciliopathies (NPHP-RC), which are characterized by multiple developmental defects and kidney diseases. Patients with NPHP-RC usually have normal glomeruli and negligible or no proteinuria. Herein, we identified novel compound-heterozygous ADAMTS9 variants in two siblings with NPHP-RC who had glomerular manifestations, including proteinuria. Methods: To investigate whether ADAMTS9 dysfunction causes NPHP and glomerulopathy, we differentiated ADAMTS9 knockout human induced pluripotent stem cells (hiPSCs) into kidney organoids. Single-cell RNA sequencing was utilized to elucidate the gene expression profiles from the ADAMTS9 knockout kidney organoids. Results: ADAMTS9 knockout had no effect on nephron differentiation; however, it reduced the number of primary cilia, thereby recapitulating renal ciliopathy. Single-cell transcriptomics revealed that podocyte clusters express the highest levels of ADAMTS9, followed by the proximal tubules. Loss of ADAMTS9 increased the activity of multiple signaling pathways, including the Wnt/PCP signaling pathway, in podocyte clusters. Conclusions: Mutations in ADMATS9 cause a glomerulotubular nephropathy in kidney and our study provides insights into the functional roles of ADMATS9 in glomeruli and tubules.

9.
Nat Commun ; 14(1): 762, 2023 02 10.
Article de Anglais | MEDLINE | ID: mdl-36765089

RÉSUMÉ

MYC-driven medulloblastomas are highly aggressive childhood brain tumors, however, the molecular and genetic events triggering MYC amplification and malignant transformation remain elusive. Here we report that mutations in CTDNEP1, a CTD nuclear-envelope-phosphatase, are the most significantly enriched recurrent alterations in MYC-driven medulloblastomas, and define high-risk subsets with poorer prognosis. Ctdnep1 ablation promotes the transformation of murine cerebellar progenitors into Myc-amplified medulloblastomas, resembling their human counterparts. CTDNEP1 deficiency stabilizes and activates MYC activity by elevating MYC serine-62 phosphorylation, and triggers chromosomal instability to induce p53 loss and Myc amplifications. Further, phosphoproteomics reveals that CTDNEP1 post-translationally modulates the activities of key regulators for chromosome segregation and mitotic checkpoint regulators including topoisomerase TOP2A and checkpoint kinase CHEK1. Co-targeting MYC and CHEK1 activities synergistically inhibits CTDNEP1-deficient MYC-amplified tumor growth and prolongs animal survival. Together, our studies demonstrate that CTDNEP1 is a tumor suppressor in highly aggressive MYC-driven medulloblastomas by controlling MYC activity and mitotic fidelity, pointing to a CTDNEP1-dependent targetable therapeutic vulnerability.


Sujet(s)
Tumeurs du cerveau , Tumeurs du cervelet , Médulloblastome , Humains , Souris , Animaux , Enfant , Médulloblastome/anatomopathologie , Phosphoric monoester hydrolases/génétique , Tumeurs du cervelet/anatomopathologie , Transformation cellulaire néoplasique/génétique , Instabilité du génome , Protéines proto-oncogènes c-myc/génétique , Phosphoprotein Phosphatases/génétique
10.
Transplantation ; 107(9): 1883-1894, 2023 09 01.
Article de Anglais | MEDLINE | ID: mdl-36717963

RÉSUMÉ

The kidney is an important organ for maintenance of homeostasis in the human body. As renal failure progresses, renal replacement therapy becomes necessary. However, there is a chronic shortage of kidney donors, creating a major problem for transplantation. To solve this problem, many strategies for the generation of transplantable kidneys are under investigation. Since the first reports describing that nephron progenitors could be induced from human induced pluripotent stem cells, kidney organoids have been attracting attention as tools for studying human kidney development and diseases. Because the kidney is formed through the interactions of multiple renal progenitors, current studies are investigating ways to combine these progenitors derived from human induced pluripotent stem cells for the generation of transplantable kidney organoids. Other bioengineering strategies, such as decellularization and recellularization of scaffolds, 3-dimensional bioprinting, interspecies blastocyst complementation and progenitor replacement, and xenotransplantation, also have the potential to generate whole kidneys, although each of these strategies has its own challenges. Combinations of these approaches will lead to the generation of bioengineered kidneys that are transplantable into humans.


Sujet(s)
Cellules souches pluripotentes induites , Transplantation rénale , Humains , Rein , Bioingénierie , Néphrons , Transplantation rénale/effets indésirables , Structures d'échafaudage tissulaires
12.
Cell Stem Cell ; 29(11): 1510-1512, 2022 Nov 03.
Article de Anglais | MEDLINE | ID: mdl-36332567

RÉSUMÉ

Collecting ducts in the kidney control the balance of water and electrolytes, as well as pH of the urine to maintain body homeostasis. Shi et al.1 recently reported in Nature Biotechnology a protocol to generate functional collecting duct cells from human pluripotent stem cells.


Sujet(s)
Tubules collecteurs rénaux , Humains , Tubules collecteurs rénaux/physiologie , Rein , Homéostasie
13.
FASEB J ; 36(11): e22606, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-36250931

RÉSUMÉ

Kinesin family member 26b (Kif26b) is essential for kidney development, and its deletion in mice leads to kidney agenesis. However, the roles of this gene in adult settings remain elusive. Thus, this study aims to investigate the role of Kif26b in the progression of renal fibrosis. A renal fibrosis model with adenine administration using Kif26b heterozygous mice and wild-type mice was established. Renal fibrosis and the underlying mechanism were investigated. The underlying pathways and functions of Kif26b were evaluated in an in vitro model using primary renal fibroblasts. Kif26b heterozygous mice were protected from renal fibrosis with adenine administration. Renal expressions of connective tissue growth factor (CTGF) and myofibroblast accumulation were reduced in Kif26b heterozygous mice. The expression of nonmuscle myosin heavy chain II (NMHCII), which binds to the C-terminus of Kif26b protein, was also suppressed in Kif26b heterozygous mice. The in vitro study revealed reduced expressions of CTGF, α-smooth muscle actin, and myosin heavy chain 9 (Myh9) via transfection with siRNAs targeting Kif26b in renal fibroblasts (RFB). RFBs, which were transfected by the expression vector of Kif26b, demonstrated higher expressions of these genes than non-transfected cells. Finally, Kif26b suppression and NMHCII blockage led to reduced abilities of migration and collagen gel contraction in renal fibroblasts. Taken together, Kif26b contributes to the progression of interstitial fibrosis via migration and myofibroblast differentiation through Myh9 in the renal fibrosis model. Blockage of this pathway at appropriate timing might be a therapeutic approach for renal fibrosis.


Sujet(s)
Rein , Kinésine , Myofibroblastes , Animaux , Souris , Actines/génétique , Actines/métabolisme , Adénine/métabolisme , Collagène/métabolisme , Facteur de croissance du tissu conjonctif/génétique , Facteur de croissance du tissu conjonctif/métabolisme , Fibroblastes/métabolisme , Fibrose , Rein/métabolisme , Kinésine/génétique , Myofibroblastes/métabolisme , Chaînes lourdes de myosine/génétique , Chaînes lourdes de myosine/métabolisme , Différenciation cellulaire , Mouvement cellulaire
14.
Curr Opin Nephrol Hypertens ; 31(4): 367-373, 2022 07 01.
Article de Anglais | MEDLINE | ID: mdl-35727170

RÉSUMÉ

PURPOSE OF REVIEW: During embryogenesis, the kidney is mainly generated from three progenitor cells; nephron progenitors, ureteric bud progenitors and stromal progenitors. Mutual interactions of the all three progenitor populations are essential to form a functional kidney with the higher-order structure. Pluripotent stem cells have potential to differentiate into all cell types of the animal body, including the kidney. In this review, we will summarize recent advances in reconstructing kidney organoids from pluripotent stem cells. RECENT FINDINGS: In the past years, major advances were reported to induce nephron and ureteric bud progenitors from pluripotent stem cells in mice and humans, and to create kidney organoids of nephron and/or ureteric bud-derived collecting duct tissues in vitro. These kidney organoid technologies were applied to high-throughput genetic screenings and small chemical screenings to identify key factors for kidney development and disease. Furthermore, a novel method was established to induce stromal progenitors from pluripotent stem cells, leading to creation of kidney organoids with the higher-order structures completely derived from pluripotent stem cells. SUMMARY: These advances in kidney organoids from pluripotent stem cells should lay a foundation to establish a novel therapy for kidney disease, which ultimately eliminate the need of dialysis and kidney transplantation for patients with kidney disease in the future.


Sujet(s)
Maladies du rein , Cellules souches pluripotentes , Animaux , Différenciation cellulaire , Humains , Rein/métabolisme , Maladies du rein/métabolisme , Souris , Organoïdes , Dialyse rénale
15.
Glia ; 70(9): 1720-1733, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35567352

RÉSUMÉ

Microglia play many critical roles in neural development. Recent single-cell RNA-sequencing studies have found diversity of microglia both across different stages and within the same stage in the developing brain. However, how such diversity is controlled during development is poorly understood. In this study, we first found the expression of the macrophage mannose receptor CD206 in early-stage embryonic microglia on mouse brain sections. This expression showed a sharp decline between E12.5 and E13.5 across the central nervous system. We next tested the roles of the microglia-expressed zinc finger transcription factor SALL1 in this early transition of gene expression. By deleting Sall1 specifically in microglia, we found that many microglia continued to express CD206 when it is normally downregulated. In addition, the mutant microglia continued to show less ramified morphology in comparison with controls even into postnatal stages. Thus, SALL1 is required for early microglia to transition into a more mature status during development.


Sujet(s)
Régulation de l'expression des gènes au cours du développement , Microglie , Neurogenèse , Facteurs de transcription , Doigts de zinc , Animaux , Régulation de l'expression des gènes au cours du développement/génétique , Souris , Microglie/métabolisme , Neurogenèse/génétique , Neurogenèse/physiologie , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Doigts de zinc/génétique , Doigts de zinc/physiologie
16.
Nat Metab ; 4(2): 180-189, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-35228746

RÉSUMÉ

Adult skeletal muscle is a highly plastic tissue that readily reduces or gains its mass in response to mechanical and metabolic stimulation; however, the upstream mechanisms that control muscle mass remain unclear. Notch signalling is highly conserved, and regulates many cellular events, including proliferation and differentiation of various types of tissue stem cell via cell-cell contact. Here we reveal that multinucleated myofibres express Notch2, which plays a crucial role in disuse- or diabetes-induced muscle atrophy. Mechanistically, in both atrophic conditions, the microvascular endothelium upregulates and releases the Notch ligand, Dll4, which then activates muscular Notch2 without direct cell-cell contact. Inhibition of the Dll4-Notch2 axis substantively prevents these muscle atrophy and promotes mechanical overloading-induced muscle hypertrophy in mice. Our results illuminate a tissue-specific function of the endothelium in controlling tissue plasticity and highlight the endothelial Dll4-muscular Notch2 axis as a central upstream mechanism that regulates catabolic signals from mechanical and metabolic stimulation, providing a therapeutic target for muscle-wasting diseases.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Protéines de liaison au calcium , Amyotrophie , Animaux , Endothélium , Souris , Muscles squelettiques , Récepteur Notch2
18.
Nat Commun ; 13(1): 611, 2022 02 01.
Article de Anglais | MEDLINE | ID: mdl-35105870

RÉSUMÉ

Organs consist of the parenchyma and stroma, the latter of which coordinates the generation of organotypic structures. Despite recent advances in organoid technology, induction of organ-specific stroma and recapitulation of complex organ configurations from pluripotent stem cells (PSCs) have remained challenging. By elucidating the in vivo molecular features of the renal stromal lineage at a single-cell resolution level, we herein establish an in vitro induction protocol for stromal progenitors (SPs) from mouse PSCs. When the induced SPs are assembled with two differentially induced parenchymal progenitors (nephron progenitors and ureteric buds), the completely PSC-derived organoids reproduce the complex kidney structure, with multiple types of stromal cells distributed along differentiating nephrons and branching ureteric buds. Thus, integration of PSC-derived lineage-specific stroma into parenchymal organoids will pave the way toward recapitulation of the organotypic architecture and functions.


Sujet(s)
Rein/cytologie , Rein/physiologie , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/physiologie , Animaux , Différenciation cellulaire , Femelle , Mâle , Souris , Souris de lignée C57BL , Souris de lignée ICR , Néphrons , Organogenèse/génétique , Organogenèse/physiologie , Organoïdes/cytologie , Transcriptome
19.
Science ; 373(6552)2021 07 16.
Article de Anglais | MEDLINE | ID: mdl-34437124

RÉSUMÉ

Oocytes mature in a specialized fluid-filled sac, the ovarian follicle, which provides signals needed for meiosis and germ cell growth. Methods have been developed to generate functional oocytes from pluripotent stem cell-derived primordial germ cell-like cells (PGCLCs) when placed in culture with embryonic ovarian somatic cells. In this study, we developed culture conditions to recreate the stepwise differentiation process from pluripotent cells to fetal ovarian somatic cell-like cells (FOSLCs). When FOSLCs were aggregated with PGCLCs derived from mouse embryonic stem cells, the PGCLCs entered meiosis to generate functional oocytes capable of fertilization and development to live offspring. Generating functional mouse oocytes in a reconstituted ovarian environment provides a method for in vitro oocyte production and follicle generation for a better understanding of mammalian reproduction.


Sujet(s)
Cellules souches embryonnaires de souris/physiologie , Ovocytes/physiologie , Ovogenèse , Follicule ovarique/cytologie , Animaux , Techniques de culture cellulaire , Différenciation cellulaire , Développement embryonnaire , Femelle , Fécondation in vitro , Mâle , Mésoderme/cytologie , Mésoderme/physiologie , Souris , Souris de lignée ICR , Cellules souches embryonnaires de souris/cytologie , Ovocytes/cytologie , Follicule ovarique/embryologie , Follicule ovarique/physiologie , RNA-Seq , Facteur stéroïdogène-1/génétique , Facteur stéroïdogène-1/métabolisme , Transcriptome
20.
Sci Rep ; 11(1): 3982, 2021 02 17.
Article de Anglais | MEDLINE | ID: mdl-33597637

RÉSUMÉ

Mutations in the NPHS1 gene, which encodes NEPHRIN, cause congenital nephrotic syndrome, resulting from impaired slit diaphragm (SD) formation in glomerular podocytes. We previously reported NEPHRIN and SD abnormalities in the podocytes of kidney organoids generated from patient-derived induced pluripotent stem cells (iPSCs) with an NPHS1 missense mutation (E725D). However, the mechanisms underlying the disease may vary depending on the mutations involved, and thus generation of iPSCs from multiple patients is warranted. Here we established iPSCs from two additional patients with different NPHS1 mutations and examined the podocyte abnormalities in kidney organoids derived from these cells. One patient had truncating mutations, and NEPHRIN was undetectable in the resulting organoids. The other patient had a missense mutation (R460Q), and the mutant NEPHRIN in the organoids failed to accumulate on the podocyte surface to form SD precursors. However, the same mutant protein behaved normally when overexpressed in heterologous cells, suggesting that NEPHRIN localization is cell context-dependent. The localization of another SD-associated protein, PODOCIN, was impaired in both types of mutant organoids in a cell domain-specific manner. Thus, the new iPSC lines and resultant kidney organoids will be useful resources for dissecting the disease mechanisms, as well as for drug development for therapies.


Sujet(s)
Cellules souches pluripotentes induites/métabolisme , Protéines membranaires/métabolisme , Protéines mutantes/métabolisme , Syndrome néphrotique/physiopathologie , Organoïdes/métabolisme , Séquence d'acides aminés , Cellules HEK293 , Humains , Protéines et peptides de signalisation intracellulaire/métabolisme , Rein , Mâle , Protéines membranaires/génétique , Protéines mutantes/génétique , Mutation faux-sens
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE