Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
2.
Nat Commun ; 13(1): 611, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35105870

RESUMO

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.


Assuntos
Rim/citologia , Rim/fisiologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Animais , Diferenciação Celular , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Néfrons , Organogênese/genética , Organogênese/fisiologia , Organoides/citologia , Transcriptoma
3.
J Am Soc Nephrol ; 31(10): 2355-2371, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32747355

RESUMO

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disease leading to renal failure, wherein multiple cysts form in renal tubules and collecting ducts derived from distinct precursors: the nephron progenitor and ureteric bud (UB), respectively. Recent progress in induced pluripotent stem cell (iPSC) biology has enabled cyst formation in nephron progenitor-derived human kidney organoids in which PKD1 or PKD2, the major causative genes for ADPKD, are deleted. However, cysts have not been generated in UB organoids, despite the prevalence of collecting duct cysts in patients with ADPKD. METHODS: CRISPR-Cas9 technology deleted PKD1 in human iPSCs and the cells induced to differentiate along pathways leading to formation of either nephron progenitor or UB organoids. Cyst formation was investigated in both types of kidney organoid derived from PKD1-deleted iPSCs and in UB organoids generated from iPSCs from a patient with ADPKD who had a missense mutation. RESULTS: Cysts formed in UB organoids with homozygous PKD1 mutations upon cAMP stimulation and, to a lesser extent, in heterozygous mutant organoids. Furthermore, UB organoids generated from iPSCs from a patient with ADPKD who had a heterozygous missense mutation developed cysts upon cAMP stimulation. CONCLUSIONS: Cysts form in PKD1 mutant UB organoids as well as in iPSCs derived from a patient with ADPKD. The organoids provide a robust model of the genesis of ADPKD.


Assuntos
Células-Tronco Pluripotentes Induzidas/patologia , Néfrons/patologia , Organoides/patologia , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , Ureter/patologia , Técnicas de Cultura de Células , Humanos , Mutação de Sentido Incorreto/genética , Rim Policístico Autossômico Dominante/patologia
4.
Stem Cell Reports ; 13(2): 322-337, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31378669

RESUMO

Kidney formation is regulated by the balance between maintenance and differentiation of nephron progenitor cells (NPCs). Now that directed differentiation of NPCs from human induced pluripotent stem cells (iPSCs) can be achieved, maintenance and propagation of NPCs in vitro should be beneficial for regenerative medicine. Although WNT and FGF signals were previously shown to be essential for NPC propagation, the requirement for BMP/TGFß signaling remains controversial. Here we reveal that activin has superior effects to BMP7 on maintenance efficiency of human iPSC-derived NPCs. Activin expanded ITGA8+/PDGFRA-/SIX2-GFP+ NPCs by 5-fold per week at 80%-90% efficiency, and the propagated cells possessed robust capacity for nephron formation both in vitro and in vivo. The expanded cells also maintained their nephron-forming potential after freezing. Furthermore, the protocol was applicable to multiple non-GFP-tagged iPSC lines. Thus, our activin-based protocol will be applicable to a variety of research fields including disease modeling and drug screening.


Assuntos
Ativinas/farmacologia , Proteína Morfogenética Óssea 7/farmacologia , Proliferação de Células/efeitos dos fármacos , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Diferenciação Celular , Reprogramação Celular , Edição de Genes , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Camundongos , Néfrons/citologia , Néfrons/metabolismo , Néfrons/patologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fosforilação , Podócitos/metabolismo , Podócitos/patologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Transplante de Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo
5.
Methods Mol Biol ; 1926: 87-102, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30742265

RESUMO

Nephrons, the functional units of the kidney, are derived from nephron progenitor cells (NPCs). Here, we describe methods to reconstruct nephron tissue via induction of NPCs from mouse and human pluripotent stem cells, which mimic multistep developmental signals in vivo. Induced NPCs differentiate into three-dimensional nephron structures, including glomerular podocytes and nephric tubules, which are useful for studying early stages of kidney specification and morphogenetic processes in the context of normal development or disease.


Assuntos
Túbulos Renais/citologia , Rim/citologia , Néfrons/citologia , Células-Tronco Pluripotentes/citologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Camundongos
6.
J Am Soc Nephrol ; 30(2): 304-321, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30635375

RESUMO

BACKGROUND: Previous research has elucidated the signals required to induce nephron progenitor cells (NPCs) from pluripotent stem cells (PSCs), enabling the generation of kidney organoids. However, selectively controlling differentiation of NPCs to podocytes has been a challenge. METHODS: We investigated the effects of various growth factors in cultured mouse embryonic NPCs during three distinct steps of nephron patterning: from NPC to pretubular aggregate, from the latter to epithelial renal vesicle (RV), and from RV to podocyte. We then applied the findings to human PSC-derived NPCs to establish a method for selective induction of human podocytes. RESULTS: Mouse NPC differentiation experiments revealed that phase-specific manipulation of Wnt and Tgf-ß signaling is critical for podocyte differentiation. First, optimal timing and intensity of Wnt signaling were essential for mesenchymal-to-epithelial transition and podocyte differentiation. Then, inhibition of Tgf-ß signaling supported domination of the RV proximal domain. Inhibition of Tgf-ß signaling in the third phase enriched the podocyte fraction by suppressing development of other nephron lineages. The resultant protocol enabled successful induction of human podocytes from PSCs with >90% purity. The induced podocytes exhibited global gene expression signatures comparable to those of adult human podocytes, had podocyte morphologic features (including foot process-like and slit diaphragm-like structures), and showed functional responsiveness to drug-induced injury. CONCLUSIONS: Elucidation of signals that induce podocytes during the nephron-patterning process enabled us to establish a highly efficient method for selective induction of human podocytes from PSCs. These PSC-derived podocytes show molecular, morphologic, and functional characteristics of podocytes, and offer a new resource for disease modeling and nephrotoxicity testing.


Assuntos
Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes/transplante , Podócitos/metabolismo , Animais , Técnicas de Cultura de Células , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Glomérulos Renais/metabolismo , Túbulos Renais/metabolismo , Camundongos , Camundongos Transgênicos , Néfrons/citologia , Organoides/metabolismo , Células-Tronco Pluripotentes/citologia , Transdução de Sinais
7.
Sci Rep ; 8(1): 14919, 2018 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-30297790

RESUMO

Kidney regeneration from pluripotent stem cells is receiving a lot of attention because limited treatments are currently available for chronic kidney disease (CKD). It has been shown that uremic state in CKD is toxic to somatic stem/progenitor cells, such as endothelial progenitor and mesenchymal stem cells, affecting their differentiation and angiogenic potential. Recent studies reported that specific abnormalities caused by the non-inherited disease are often retained in induced pluripotent stem cell (iPSC)-derived products obtained from patients. Thus, it is indispensable to first assess whether iPSCs derived from patients with CKD due to non-inherited disease (CKD-iPSCs) have the ability to generate kidneys. In this study, we generated iPSCs from patients undergoing haemodialysis due to diabetes nephropathy and glomerulonephritis (HD-iPSCs) as representatives of CKD-iPSCs or from healthy controls (HC-iPSCs). HD-iPSCs differentiated into nephron progenitor cells (NPCs) with similar efficiency to HC-iPSCs. Additionally, HD-iPSC-derived NPCs expressed comparable levels of NPC markers and differentiated into vascularised glomeruli upon transplantation into mice, as HC-iPSC-derived NPCs. Our results indicate the potential of HD-iPSCs as a feasible cell source for kidney regeneration. This is the first study paving the way for CKD patient-stem cell-derived kidney regeneration, emphasising the potential of CKD-iPSCs.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Rim/citologia , Rim/fisiologia , Regeneração , Insuficiência Renal Crônica/terapia , Animais , Diferenciação Celular , Células Cultivadas , Nefropatias Diabéticas/fisiopatologia , Nefropatias Diabéticas/terapia , Glomerulonefrite/fisiopatologia , Glomerulonefrite/terapia , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Rim/fisiopatologia , Camundongos , Néfrons/citologia , Néfrons/fisiologia , Néfrons/fisiopatologia , Diálise Renal , Insuficiência Renal Crônica/fisiopatologia
8.
Stem Cell Reports ; 11(3): 727-740, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30174315

RESUMO

Mutations in the NPHS1 gene, which encodes NEPHRIN, cause congenital nephrotic syndrome, resulting from impaired slit diaphragm (SD) formation in glomerular podocytes. However, methods for SD reconstitution have been unavailable, thereby limiting studies in the field. In the present study, we established human induced pluripotent stem cells (iPSCs) from a patient with an NPHS1 missense mutation, and reproduced the SD formation process using iPSC-derived kidney organoids. The mutant NEPHRIN failed to become localized on the cell surface for pre-SD domain formation in the induced podocytes. Upon transplantation, the mutant podocytes developed foot processes, but exhibited impaired SD formation. Genetic correction of the single amino acid mutation restored NEPHRIN localization and phosphorylation, colocalization of other SD-associated proteins, and SD formation. Thus, these kidney organoids from patient-derived iPSCs identified SD abnormalities in the podocytes at the initial phase of congenital nephrotic disease.


Assuntos
Células-Tronco Pluripotentes Induzidas/patologia , Proteínas de Membrana/análise , Síndrome Nefrótica/patologia , Organoides/patologia , Podócitos/patologia , Animais , Células Cultivadas , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Rim/metabolismo , Rim/patologia , Proteínas de Membrana/genética , Camundongos SCID , Mutação de Sentido Incorreto , Síndrome Nefrótica/genética , Organoides/metabolismo , Podócitos/metabolismo
9.
Cell Stem Cell ; 21(6): 730-746.e6, 2017 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-29129523

RESUMO

Organogenesis generates higher-order structures containing functional subunits, connective components, and progenitor niches. Despite recent advances in organoid-based modeling of tissue development, recapitulating these complex configurations from pluripotent stem cells (PSCs) has remained challenging. In this study, we report assembly of kidney organoids that recapitulate embryonic branching morphogenesis. By studying the distinct origins and developmental processes of the ureteric bud, which contains epithelial kidney progenitors that undergo branching morphogenesis and thereby plays a central role in orchestrating organ geometry, and neighboring mesenchymal nephron progenitors, we established a protocol for differential induction of each lineage from mouse and human PSCs. Importantly, reassembled organoids developed the inherent architectures of the embryonic kidney, including the peripheral progenitor niche and internally differentiated nephrons that were interconnected by a ramified ureteric epithelium. This selective induction and reassembly strategy will be a powerful approach to recapitulate organotypic architecture in PSC-derived organoids.


Assuntos
Rim/metabolismo , Organogênese , Células-Tronco Pluripotentes/metabolismo , Animais , Células Cultivadas , Humanos , Rim/embriologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Células-Tronco Pluripotentes/citologia
10.
Sci Rep ; 7(1): 4554, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28674456

RESUMO

The kidney is formed by reciprocal interactions between the nephron progenitor and the ureteric bud, the former of which gives rise to the epithelia of nephrons consisting of glomeruli and renal tubules. The transcription factor PAX2 is essential for this mesenchymal-to-epithelial transition of nephron progenitors, as well as ureteric bud lineage development, in mice. PAX2 mutations in humans cause renal coloboma syndrome. We previously reported the induction of nephron progenitors and three-dimensional nephron structures from human induced pluripotent stem (iPS) cells. Here we generate iPS cells lacking PAX2, and address the role of PAX2 in our in vitro induction protocol. While PAX2-null human nephron progenitors were properly formed, they unexpectedly became epithelialised to form glomeruli and renal tubules. However, the mutant glomerular parietal epithelial cells failed to transit to the squamous morphology, retaining the shape and markers of columnar epithelia. Therefore, PAX2 is dispensable for mesenchymal-to-epithelial transition of nephron progenitors, but is required for morphological development of glomerular parietal epithelial cells, during nephron formation from human iPS cells in vitro.


Assuntos
Diferenciação Celular/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Néfrons/citologia , Néfrons/metabolismo , Organogênese/genética , Fator de Transcrição PAX2/genética , Biomarcadores , Transição Epitelial-Mesenquimal/genética , Imunofluorescência , Expressão Gênica , Técnicas de Inativação de Genes , Genótipo , Humanos , Técnicas In Vitro , Glomérulos Renais/citologia , Glomérulos Renais/metabolismo , Túbulos Renais/citologia , Túbulos Renais/metabolismo , Fator de Transcrição PAX2/metabolismo
11.
Methods Mol Biol ; 1597: 179-193, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28361318

RESUMO

The kidney is a vital organ that has an important role in the maintenance of homeostasis by fluid volume regulation and waste product excretion. This role cannot be performed without the three-dimensional (3D) structure of the kidney. Therefore, it is important to generate the 3D structure of the kidney when inducing functional kidney tissue or the whole organ from pluripotent stem cells. In this chapter, we describe the detailed methods to induce kidney progenitor cells from pluripotent stem cells, which are based on embryological development. We also provide a method to generate 3D kidney tissue with vascularized glomeruli upon transplantation.


Assuntos
Rim/citologia , Células-Tronco Pluripotentes/citologia , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID
12.
Pediatr Nephrol ; 32(2): 195-200, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-26868670

RESUMO

Studies of kidney regeneration using stem cells have progressed rapidly in recent years. Our group has developed a protocol to induce nephron progenitors from both mouse and human pluripotent stem cells which is based on a revised model of early stage kidney specification. The induced progenitors readily reconstitute three-dimensional nephron structures, including glomeruli and renal tubules, in vitro. We can further generate human induced pluripotent stem cells (iPSCs), in which nephrin-expressing glomerular podocytes are tagged with green fluorescent protein (GFP). The sorted GFP-positive cells retain the podocyte-specific molecular and structural features. Upon transplantation, mouse endothelial cells of the host animals are integrated into the human iPSC-derived glomeruli, and the podocytes show further maturation. Other laboratories have reported different protocols to induce nephron structures from human iPSCs in vitro. These findings will accelerate our understanding of kidney development and diseases in humans.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Glomérulos Renais/citologia , Túbulos Renais/citologia , Animais , Técnicas de Cultura de Células , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Glomérulos Renais/metabolismo , Túbulos Renais/metabolismo , Camundongos
13.
Cell Rep ; 15(4): 801-813, 2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27149838

RESUMO

Nephron progenitors in the embryonic kidney propagate while generating differentiated nephrons. However, in mice, the progenitors terminally differentiate shortly after birth. Here, we report a method for selectively expanding nephron progenitors in vitro in an undifferentiated state. Combinatorial and concentration-dependent stimulation with LIF, FGF2/9, BMP7, and a WNT agonist is critical for expansion. The purified progenitors proliferated beyond the physiological limits observed in vivo, both for cell numbers and lifespan. Neonatal progenitors were maintained for a week, while progenitors from embryonic day 11.5 expanded 1,800-fold for nearly 20 days and still reconstituted 3D nephrons containing glomeruli and renal tubules. Furthermore, progenitors generated from mouse embryonic stem cells and human induced pluripotent cells could be expanded with retained nephron-forming potential. Thus, we have established in vitro conditions for promoting the propagation of nephron progenitors, which will be essential for dissecting the mechanisms of kidney organogenesis and for regenerative medicine.

14.
J Am Soc Nephrol ; 27(6): 1778-91, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26586691

RESUMO

Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator-like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in vitro These induced human podocytes exhibited apicobasal polarity, with nephrin proteins accumulated close to the basal domain, and possessed primary processes that were connected with slit diaphragm-like structures. Microarray analysis of sorted iPS cell-derived podocytes identified well conserved marker gene expression previously shown in mouse and human podocytes in vivo Furthermore, we developed a novel transplantation method using spacers that release the tension of host kidney capsules, thereby allowing the effective formation of glomeruli from human iPS cell-derived nephron progenitors. The human glomeruli were vascularized with the host mouse endothelial cells, and iPS cell-derived podocytes with numerous cell processes accumulated around the fenestrated endothelial cells. Therefore, the podocytes generated from iPS cells retain the podocyte-specific molecular and structural features, which will be useful for dissecting human glomerular development and diseases.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Glomérulos Renais/irrigação sanguínea , Podócitos/fisiologia , Animais , Transplante de Células , Células Cultivadas , Humanos , Camundongos , Podócitos/ultraestrutura
16.
Kidney Int ; 87(5): 894-900, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25469851

RESUMO

It has been a challenge in developmental biology and regenerative medicine to generate nephron progenitors that reconstitute the three-dimensional (3D) nephron structure in vitro. Many studies have tried to induce nephron progenitors from pluripotent stem cells by mimicking the developmental processes in vivo. However, the current developmental model does not precisely address the spatiotemporal origin of nephron progenitors, hampering our understanding of cell fate decisions in the kidney. Here, we present a revised model of early-stage kidney specification, suggesting distinct origins of the two major kidney components: the ureteric bud and metanephric mesenchyme. This model enables the induction of metanephric nephron progenitors from both mouse and human pluripotent stem cells. The induced cells self-organize in the presence of Wnt signaling and reconstitute 3D nephron structures including both nephric tubules with a clear lumina and glomeruli with podocytes. The engrafted kidney tissue develops vascularized glomeruli and nephric tubules, but it does not produce urine, suggesting the requirement for further maturation. Nevertheless, the generation of nephron components from human-induced pluripotent stem cells will be useful for future application in regenerative therapy and modeling of congenital kidney diseases in vitro. This review discusses the possibility of de novo organogenesis of a functional kidney from pluripotent stem cells and the future direction toward clinical applications.


Assuntos
Néfrons/citologia , Células-Tronco Pluripotentes/fisiologia , Animais , Humanos , Néfrons/embriologia , Néfrons/fisiologia , Regeneração
17.
J Am Soc Nephrol ; 26(5): 1081-91, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25168025

RESUMO

The kidney develops from reciprocal interactions between the metanephric mesenchyme and ureteric bud. The mesenchyme transforms into epithelia and forms complicated nephron structures, whereas the ureteric bud extends its pre-existing epithelial ducts. Although the roles are well established for extracellular stimuli, such as Wnt and Notch, it is unclear how the intracellular cytoskeleton regulates these morphogenetic processes. Myh9 and Myh10 encode nonmuscle myosin II heavy chains, and Myh9 mutations in humans are implicated in congenital kidney diseases and focal segmental glomerulosclerosis in adults. Here, we analyzed the roles of Myh9 and Myh10 in the developing kidney. Ureteric bud-specific depletion of Myh9 resulted in no apparent phenotypes, whereas mesenchyme-specific Myh9 deletion caused proximal tubule dilations and renal failure. Mesenchyme-specific Myh9/Myh10 mutant mice died shortly after birth and showed a severe defect in nephron formation. The nascent mutant nephrons failed to form a continuous lumen, which likely resulted from impaired apical constriction of the elongating tubules. In addition, nephron progenitors lacking Myh9/Myh10 or the possible interactor Kif26b were less condensed at midgestation and reduced at birth. Taken together, nonmuscle myosin II regulates the morphogenesis of immature nephrons derived from the metanephric mesenchyme and the maintenance of nephron progenitors. Our data also suggest that Myh9 deletion in mice results in failure to maintain renal tubules but not in glomerulosclerosis.


Assuntos
Morfogênese , Cadeias Pesadas de Miosina/fisiologia , Néfrons/embriologia , Miosina não Muscular Tipo IIA/fisiologia , Miosina não Muscular Tipo IIB/fisiologia , Animais , Animais Recém-Nascidos , Mesoderma/fisiologia , Camundongos Endogâmicos C57BL , Néfrons/metabolismo , Isoformas de Proteínas/metabolismo
18.
J Am Soc Nephrol ; 25(11): 2584-95, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24744442

RESUMO

The balanced self-renewal and differentiation of nephron progenitors are critical for kidney development and controlled, in part, by the transcription factor Six2, which antagonizes canonical Wnt signaling-mediated differentiation. A nuclear factor, Sall1, is expressed in Six2-positive progenitors as well as differentiating nascent nephrons, and it is essential for kidney formation. However, the molecular functions and targets of Sall1, especially the functions and targets in the nephron progenitors, remain unknown. Here, we report that Sall1 deletion in Six2-positive nephron progenitors results in severe progenitor depletion and apoptosis of the differentiating nephrons in mice. Analysis of mice with an inducible Sall1 deletion revealed that Sall1 activates genes expressed in progenitors while repressing genes expressed in differentiating nephrons. Sall1 and Six2 co-occupied many progenitor-related gene loci, and Sall1 bound to Six2 biochemically. In contrast, Sall1 did not bind to the Wnt4 locus suppressed by Six2. Sall1-mediated repression was also independent of its binding to DNA. Thus, Sall1 maintains nephron progenitors and their derivatives by a unique mechanism, which partly overlaps but is distinct from that of Six2: Sall1 activates progenitor-related genes in Six2-positive nephron progenitors and represses gene expression in Six2-negative differentiating nascent nephrons.


Assuntos
Rim/embriologia , Néfrons/embriologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Feminino , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Rim/citologia , Rim/fisiologia , Camundongos Transgênicos , Néfrons/citologia , Néfrons/fisiologia , Técnicas de Cultura de Órgãos , Gravidez , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia
19.
Cell Stem Cell ; 14(1): 53-67, 2014 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24332837

RESUMO

Recapitulating three-dimensional (3D) structures of complex organs, such as the kidney, from pluripotent stem cells (PSCs) is a major challenge. Here, we define the developmental origins of the metanephric mesenchyme (MM), which generates most kidney components. Unexpectedly, we find that posteriorly located T(+) MM precursors are developmentally distinct from Osr1(+) ureteric bud progenitors during the postgastrulation stage, and we identify phasic Wnt stimulation and stage-specific growth factor addition as molecular cues that promote their development into the MM. We then use this information to derive MM from PSCs. These progenitors reconstitute the 3D structures of the kidney in vitro, including glomeruli with podocytes and renal tubules with proximal and distal regions and clear lumina. Furthermore, the glomeruli are efficiently vascularized upon transplantation. Thus, by reevaluating the developmental origins of metanephric progenitors, we have provided key insights into kidney specification in vivo and taken important steps toward kidney organogenesis in vitro.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Rim/citologia , Mesoderma/citologia , Néfrons/citologia , Organogênese/fisiologia , Animais , Biomarcadores/metabolismo , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Células-Tronco Embrionárias/metabolismo , Humanos , Immunoblotting , Células-Tronco Pluripotentes Induzidas/metabolismo , Rim/química , Rim/metabolismo , Mesoderma/metabolismo , Néfrons/metabolismo , Técnicas de Cultura de Órgãos , Transdução de Sinais
20.
PLoS One ; 8(6): e68508, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23825698

RESUMO

The kidney, the metanephros, is formed by reciprocal interactions between the metanephric mesenchyme and the ureteric bud, the latter of which is derived from the Wolffian duct that elongates in the rostral-to-caudal direction. Sall1 expressed in the metanephric mesenchyme is essential for ureteric bud attraction in kidney development. Sall4, another member of the Sall gene family, is required for maintenance of embryonic stem cells and establishment of induced pluripotent stem cells, and is thus considered to be one of the stemness genes. Sall4 is also a causative gene for Okihiro syndrome and is essential for the formation of many organs in both humans and mice. However, its expression and role in kidney development remain unknown, despite the essential role of Sall1 in the metanephric mesenchyme. Here, we report that mouse Sall4 is expressed transiently in the Wolffian duct-derived lineage, and is nearly complementary to Sall1 expression. While Sall4 expression is excluded from the Wolffian duct at embryonic (E) day 9.5, Sall4 is expressed in the Wolffian duct weakly in the mesonephric region at E10.5 and more abundantly in the caudal metanephric region where ureteric budding occurs. Sall4 expression is highest at E11.5 in the Wolffian duct and ureteric bud, but disappears by E13.5. We further demonstrate that Sall4 deletion in the Wolffian duct and ureteric bud does not cause any apparent kidney phenotypes. Therefore, Sall4 is expressed transiently in the caudal Wolffian duct and the ureteric bud, but is dispensable for kidney development in mice.


Assuntos
Rim/embriologia , Fatores de Transcrição/metabolismo , Ureter/embriologia , Animais , Humanos , Camundongos , Ductos Mesonéfricos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA