Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cell Rep ; 43(2): 113602, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38237600

RESUMO

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.


Assuntos
Eritropoetina , Células-Tronco Pluripotentes Induzidas , Humanos , Animais , Camundongos , Rim , Linhagem da Célula , Regeneração
2.
Commun Biol ; 6(1): 854, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37770589

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas , Nefrite Hereditária , Adolescente , Humanos , Nefrite Hereditária/genética , Nefrite Hereditária/metabolismo , Nefrite Hereditária/terapia , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Rim/metabolismo , Membrana Basal Glomerular
3.
Commun Biol ; 6(1): 483, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37142732

RESUMO

Of late, numerous microphysiological systems have been employed to model the renal proximal tubule. Yet there is lack of research on refining the functions of the proximal tubule epithelial layer-selective filtration and reabsorption. In this report, pseudo proximal tubule cells extracted from human-induced pluripotent stem cell-derived kidney organoids are combined and cultured with immortalized proximal tubule cells. It is shown that the cocultured tissue is an impervious epithelium that offers improved levels of certain transporters, extracellular matrix proteins collagen and laminin, and superior glucose transport and P-glycoprotein activity. mRNA expression levels higher than those obtained from each cell type were detected, suggesting an anomalous synergistic crosstalk between the two. Alongside, the improvements in morphological characteristics and performance of the immortalized proximal tubule tissue layer exposed, upon maturation, to human umbilical vein endothelial cells are thoroughly quantified and compared. Glucose and albumin reabsorption, as well as xenobiotic efflux rates through P-glycoprotein were all improved. The data presented abreast highlight the advantages of the cocultured epithelial layer and the non-iPSC-based bilayer. The in vitro models presented herein can be helpful in personalized nephrotoxicity studies.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Endoteliais/metabolismo , Rim/metabolismo , Organoides/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP , Glucose/metabolismo
4.
Am J Physiol Renal Physiol ; 323(5): F515-F526, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36049065

RESUMO

Although apoptosis of podocytes has been widely reported in in vitro studies, it has been less frequently and less definitively documented in in vivo situations. To investigate this discrepancy, we analyzed the dying process of podocytes in vitro and in vivo using LMB2, a human (h)CD25-directed immunotoxin. LMB2 induced cell death within 2 days in 56.8 ± 13.6% of cultured podocytes expressing hCD25 in a caspase-3, Bak1, and Bax-dependent manner. LMB2 induced typical apoptotic features, including TUNEL staining and fragmented nuclei without lactate dehydrogenase leakage. In vivo, LMB2 effectively eliminated hCD25-expressing podocytes in NEP25 mice. Podocytes injured by LMB2 were occasionally stained for cleaved caspase-3 and cleaved lamin A but never for TUNEL. Urinary sediment contained TUNEL-positive podocytes. To examine the effect of glomerular filtration, we performed unilateral ureteral obstruction in NEP25 mice treated with LMB2 1 day before euthanasia. In the obstructed kidney, glomeruli contained significantly more cleaved lamin A-positive podocytes than those in the contralateral kidney (50.1 ± 5.4% vs. 29.3 ± 4.1%, P < 0.001). To further examine the dying process without glomerular filtration, we treated kidney organoids generated from nephron progenitor cells of NEP25 mice with LMB2. Podocytes showed TUNEL staining and nuclear fragmentation. These results indicate that on activation of apoptotic caspases, podocytes are detached and lost in the urine before nuclear fragmentation and that the physical force of glomerular filtration facilitates detachment. This phenomenon may be the reason why definitive apoptosis is not observed in podocytes in vivo.NEW & NOTEWORTHY This report clarifies why morphologically definitive apoptosis is not observed in podocytes in vivo. When caspase-3 is activated in podocytes, these cells are immediately detached from the glomerulus and lost in the urine before DNA fragmentation occurs. Detachment is facilitated by glomerular filtration. This phenomenon explains why podocytes in vivo rarely show TUNEL staining and never apoptotic bodies.


Assuntos
Imunotoxinas , Podócitos , Camundongos , Humanos , Animais , Podócitos/metabolismo , Caspase 3/metabolismo , Lamina Tipo A/metabolismo , Lamina Tipo A/farmacologia , Proteína X Associada a bcl-2/metabolismo , Apoptose , Lactato Desidrogenases/metabolismo
5.
Biochem Biophys Res Commun ; 558: 231-238, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32113685

RESUMO

Several groups have developed in vitro expansion cultures for mouse metanephric nephron progenitor cells (NPCs) using cocktails of small molecules and growth factors including BMP7. However, the detailed mechanisms by which BMP7 acts in the NPC expansion remain to be elucidated. Here, by performing chemical screening for BMP substitutes, we identified a small molecule, TCS21311, that can replace BMP7 and revealed a novel inhibitory role of BMP7 in JAK3-STAT3 signaling in NPC expansion culture. Further, we found that TCS21311 facilitates the proliferation of mouse embryonic NPCs and human induced pluripotent stem cell-derived NPCs when added to the expansion culture. These results will contribute to understanding the mechanisms of action of BMP7 in NPC proliferation in vitro and in vivo and to the stable supply of NPCs for regenerative therapy, disease modeling and drug discovery for kidney diseases.


Assuntos
Proteína Morfogenética Óssea 7/metabolismo , Inibidores de Janus Quinases/farmacologia , Néfrons/citologia , Néfrons/efeitos dos fármacos , Animais , Proteína Morfogenética Óssea 7/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Meios de Cultura , Avaliação Pré-Clínica de Medicamentos , Humanos , Técnicas In Vitro , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Janus Quinase 3/antagonistas & inibidores , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/metabolismo , Néfrons/metabolismo , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas
6.
Biochem Biophys Res Commun ; 529(4): 1186-1194, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32819584

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas/patologia , Rim/patologia , Modelos Biológicos , Organoides/patologia , Rim Policístico Autossômico Dominante/patologia , Sequência de Aminoácidos , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Colforsina/farmacologia , Avaliação Pré-Clínica de Medicamentos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Mutação/genética , Fenótipo , Canais de Cátion TRPP/química , Canais de Cátion TRPP/genética
7.
Cell Rep ; 31(1): 107476, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32268094

RESUMO

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.


Assuntos
Técnicas de Cultura de Células/métodos , Linhagem da Célula/fisiologia , Células-Tronco Pluripotentes/citologia , Diferenciação Celular/fisiologia , Células Cultivadas , Células Epiteliais , Humanos , Rim/citologia , Mesoderma , Néfrons , Organogênese/fisiologia , Organoides/citologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/fisiologia
8.
Sci Rep ; 9(1): 10701, 2019 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-31316087

RESUMO

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.

9.
Methods Mol Biol ; 1926: 151-159, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30742270

RESUMO

During kidney development, Six2+ nephron progenitor cells (NPCs) generate nephrons, the functional units of the kidney. Isolating and expanding NPCs in vitro have enabled wide applications in both basic and translational research. Here we describe the methods to derive NPC lines from mouse embryonic kidneys in a 3D culture format as well as gene editing in the NPC lines.


Assuntos
Néfrons/citologia , Células-Tronco/citologia , Animais , Linhagem Celular , Células Cultivadas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Edição de Genes/métodos , Camundongos
10.
Nature ; 561(7722): 243-247, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30185909

RESUMO

Large cutaneous ulcers are, in severe cases, life threatening1,2. As the global population ages, non-healing ulcers are becoming increasingly common1,2. Treatment currently requires the transplantation of pre-existing epithelial components, such as skin grafts, or therapy using cultured cells2. Here we develop alternative supplies of epidermal coverage for the treatment of these kinds of wounds. We generated expandable epithelial tissues using in vivo reprogramming of wound-resident mesenchymal cells. Transduction of four transcription factors that specify the skin-cell lineage enabled efficient and rapid de novo epithelialization from the surface of cutaneous ulcers in mice. Our findings may provide a new therapeutic avenue for treating skin wounds and could be extended to other disease situations in which tissue homeostasis and repair are impaired.


Assuntos
Reprogramação Celular , Células Epiteliais/citologia , Úlcera Cutânea/patologia , Pele/citologia , Ferimentos e Lesões/patologia , Animais , Linhagem da Célula , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Medicina Regenerativa , Pele/patologia , Úlcera Cutânea/terapia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Cicatrização , Ferimentos e Lesões/terapia
11.
Sci Rep ; 8(1): 6375, 2018 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-29686294

RESUMO

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.


Assuntos
Injúria Renal Aguda/terapia , Biomarcadores/metabolismo , Separação Celular/métodos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Transplante de Células-Tronco/métodos , Injúria Renal Aguda/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos
12.
Cell ; 171(7): 1495-1507.e15, 2017 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-29224783

RESUMO

Current genome-editing systems generally rely on inducing DNA double-strand breaks (DSBs). This may limit their utility in clinical therapies, as unwanted mutations caused by DSBs can have deleterious effects. CRISPR/Cas9 system has recently been repurposed to enable target gene activation, allowing regulation of endogenous gene expression without creating DSBs. However, in vivo implementation of this gain-of-function system has proven difficult. Here, we report a robust system for in vivo activation of endogenous target genes through trans-epigenetic remodeling. The system relies on recruitment of Cas9 and transcriptional activation complexes to target loci by modified single guide RNAs. As proof-of-concept, we used this technology to treat mouse models of diabetes, muscular dystrophy, and acute kidney disease. Results demonstrate that CRISPR/Cas9-mediated target gene activation can be achieved in vivo, leading to measurable phenotypes and amelioration of disease symptoms. This establishes new avenues for developing targeted epigenetic therapies against human diseases. VIDEO ABSTRACT.


Assuntos
Sistemas CRISPR-Cas , Epigênese Genética , Marcação de Genes/métodos , Terapia Genética/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Utrofina/genética , Animais , Sequência de Bases , Modelos Animais de Doenças , Distrofina/genética , Interleucina-10/genética , Proteínas Klotho , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Ativação Transcricional
13.
Cell ; 167(7): 1719-1733.e12, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27984723

RESUMO

Aging is the major risk factor for many human diseases. In vitro studies have demonstrated that cellular reprogramming to pluripotency reverses cellular age, but alteration of the aging process through reprogramming has not been directly demonstrated in vivo. Here, we report that partial reprogramming by short-term cyclic expression of Oct4, Sox2, Klf4, and c-Myc (OSKM) ameliorates cellular and physiological hallmarks of aging and prolongs lifespan in a mouse model of premature aging. Similarly, expression of OSKM in vivo improves recovery from metabolic disease and muscle injury in older wild-type mice. The amelioration of age-associated phenotypes by epigenetic remodeling during cellular reprogramming highlights the role of epigenetic dysregulation as a driver of mammalian aging. Establishing in vivo platforms to modulate age-associated epigenetic marks may provide further insights into the biology of aging.


Assuntos
Envelhecimento/genética , Reprogramação Celular , Epigênese Genética , Doenças Metabólicas/genética , Fatores de Transcrição/metabolismo , Senilidade Prematura/genética , Senilidade Prematura/metabolismo , Animais , Diabetes Mellitus Tipo 2/induzido quimicamente , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Lamina Tipo A/genética , Doenças Metabólicas/metabolismo , Doenças Metabólicas/prevenção & controle , Camundongos , Modelos Animais , Pâncreas/metabolismo , Sarcopenia/metabolismo
14.
Nature ; 540(7631): 144-149, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27851729

RESUMO

Targeted genome editing via engineered nucleases is an exciting area of biomedical research and holds potential for clinical applications. Despite rapid advances in the field, in vivo targeted transgene integration is still infeasible because current tools are inefficient, especially for non-dividing cells, which compose most adult tissues. This poses a barrier for uncovering fundamental biological principles and developing treatments for a broad range of genetic disorders. Based on clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9) technology, here we devise a homology-independent targeted integration (HITI) strategy, which allows for robust DNA knock-in in both dividing and non-dividing cells in vitro and, more importantly, in vivo (for example, in neurons of postnatal mammals). As a proof of concept of its therapeutic potential, we demonstrate the efficacy of HITI in improving visual function using a rat model of the retinal degeneration condition retinitis pigmentosa. The HITI method presented here establishes new avenues for basic research and targeted gene therapies.


Assuntos
Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Marcação de Genes/métodos , Genoma/genética , Retinose Pigmentar/genética , Retinose Pigmentar/terapia , Animais , Divisão Celular , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Terapia Genética/métodos , Neurônios/citologia , Neurônios/metabolismo , Ratos , Homologia de Sequência
15.
Cell Stem Cell ; 19(4): 516-529, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27570066

RESUMO

Transit-amplifying nephron progenitor cells (NPCs) generate all of the nephrons of the mammalian kidney during development. Their limited numbers, poor in vitro expansion, and difficult accessibility in humans have slowed basic and translational research into renal development and diseases. Here, we show that with appropriate 3D culture conditions, it is possible to support long-term expansion of primary mouse and human fetal NPCs as well as NPCs derived from human induced pluripotent stem cells (iPSCs). Expanded NPCs maintain genomic stability, molecular homogeneity, and nephrogenic potential in vitro, ex vivo, and in vivo. Cultured NPCs are amenable to gene targeting and can form nephron organoids that engraft in vivo, functionally couple to the host's circulatory system, and produce urine-like metabolites via filtration. Together, these findings provide a technological platform for studying human nephrogenesis, modeling and diagnosing renal diseases, and drug discovery.


Assuntos
Técnicas de Cultura de Células/métodos , Néfrons/citologia , Organogênese , Células-Tronco/citologia , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Edição de Genes , Humanos , Testes de Função Renal , Camundongos , Organoides/citologia , Comunicação Parácrina , Células-Tronco/metabolismo , Fatores de Tempo
16.
Sci Rep ; 6: 30013, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27418197

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Aneurisma Intracraniano/patologia , Metaloproteinase 1 da Matriz/sangue , Rim Policístico Autossômico Dominante/patologia , Hemorragia Subaracnóidea/patologia , Idoso , Animais , Biomarcadores/sangue , Diferenciação Celular , Células Cultivadas , Metilação de DNA/genética , Feminino , Humanos , Aneurisma Intracraniano/sangue , Masculino , Metaloproteinase 1 da Matriz/biossíntese , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Rim Policístico Autossômico Dominante/mortalidade , Fatores de Risco , Canais de Cátion TRPP/genética
17.
Stem Cells Transl Med ; 4(9): 980-92, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26198166

RESUMO

UNLABELLED: Acute kidney injury (AKI) is defined as a rapid loss of renal function resulting from various etiologies, with a mortality rate exceeding 60% among intensive care patients. Because conventional treatments have failed to alleviate this condition, the development of regenerative therapies using human induced pluripotent stem cells (hiPSCs) presents a promising new therapeutic option for AKI. We describe our methodology for generating renal progenitors from hiPSCs that show potential in ameliorating AKI. We established a multistep differentiation protocol for inducing hiPSCs into OSR1+SIX2+ renal progenitors capable of reconstituting three-dimensional proximal renal tubule-like structures in vitro and in vivo. Moreover, we found that renal subcapsular transplantation of hiPSC-derived renal progenitors ameliorated the AKI in mice induced by ischemia/reperfusion injury, significantly suppressing the elevation of blood urea nitrogen and serum creatinine levels and attenuating histopathological changes, such as tubular necrosis, tubule dilatation with casts, and interstitial fibrosis. To our knowledge, few reports demonstrating the therapeutic efficacy of cell therapy with renal lineage cells generated from hiPSCs have been published. Our results suggest that regenerative medicine strategies for kidney diseases could be developed using hiPSC-derived renal cells. SIGNIFICANCE: This report is the first to demonstrate that the transplantation of renal progenitor cells differentiated from human induced pluripotent stem (iPS) cells has therapeutic effectiveness in mouse models of acute kidney injury induced by ischemia/reperfusion injury. In addition, this report clearly demonstrates that the therapeutic benefits come from trophic effects by the renal progenitor cells, and it identifies the renoprotective factors secreted by the progenitors. The results of this study indicate the feasibility of developing regenerative medicine strategy using iPS cells against renal diseases.


Assuntos
Injúria Renal Aguda/terapia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Epiteliais/transplante , Células-Tronco Pluripotentes Induzidas/citologia , Necrose/prevenção & controle , Traumatismo por Reperfusão/terapia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Biomarcadores/metabolismo , Nitrogênio da Ureia Sanguínea , Diferenciação Celular , Células Cultivadas , Creatinina/sangue , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fibrose , Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Túbulos Renais/lesões , Túbulos Renais/metabolismo , Masculino , Camundongos , Camundongos SCID , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transplante Heterólogo , Urotélio/citologia , Urotélio/metabolismo
18.
PLoS One ; 9(1): e84881, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24454758

RESUMO

The first step in developing regenerative medicine approaches to treat renal diseases using pluripotent stem cells must be the generation of intermediate mesoderm (IM), an embryonic germ layer that gives rise to kidneys. In order to achieve this goal, establishing an efficient, stable and low-cost method for differentiating IM cells using small molecules is required. In this study, we identified two retinoids, AM580 and TTNPB, as potent IM inducers by high-throughput chemical screening, and established rapid (five days) and efficient (80% induction rate) IM differentiation from human iPSCs using only two small molecules: a Wnt pathway activator, CHIR99021, combined with either AM580 or TTNPB. The resulting human IM cells showed the ability to differentiate into multiple cell types that constitute adult kidneys, and to form renal tubule-like structures. These small molecule differentiation methods can bypass the mesendoderm step, directly inducing IM cells by activating Wnt, retinoic acid (RA), and bone morphogenetic protein (BMP) pathways. Such methods are powerful tools for studying kidney development and may potentially provide cell sources to generate renal lineage cells for regenerative therapy.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Rim/citologia , Mesoderma/citologia , Células-Tronco Pluripotentes/citologia , Animais , Linhagem Celular , Células-Tronco Embrionárias/metabolismo , Citometria de Fluxo , Ensaios de Triagem em Larga Escala , Humanos , Rim/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais
19.
PLoS One ; 8(6): e66759, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23826128

RESUMO

BACKGROUND: Gas6 is a growth factor that causes proliferation of mesangial cells in the development of glomerulonephritis. Gas6 can bind to three kinds of receptors; Axl, Dtk, and Mer. However, their expression and functions are not entirely clear in the different glomerular cell types. Meanwhile, representative cell cycle regulatory protein p27 has been reported to be expressed in podocytes in normal glomeruli with decreased expression in proliferating glomeruli, which inversely correlated with mesangial proliferation in human IgA nephropathy (IgAN). METHODS: The aim of this study is to clarify Gas6 involvement in the progression of IgAN. Expression of Gas6/Axl/Dtk was examined in 31 biopsy proven IgAN cases. We compared the expression levels with histological severity or clinical data. Moreover, we investigated the expression of Gas6 and its receptors in cultured podocytes. RESULTS: In 28 of 31 cases, Gas6 was upregulated mainly in podocytes. In the other 3 cases, Gas6 expression was induced in endothelial and mesangial cells, which was similar to animal nephritis models. Among 28 podocyte type cases, the expression level of Gas6 correlated with the mesangial hypercellularity score of IgAN Oxford classification and urine protein excretion. It also inversely correlated with p27 expression in glomeruli. As for the receptors, Axl was mainly expressed in endothelial and mesangial cells, while Dtk was expressed in podocytes. In vitro, Dtk was expressed in cultured murine podocytes, and the expression of p27 was decreased by Gas6 stimulation. CONCLUSIONS: Gas6 was uniquely upregulated in either endothelial/mesangial cells or podocytes in IgAN. The expression pattern can be used as a marker to classify IgAN. Gas6 has a possibility to be involved in not only mesangial proliferation via Axl, but also podocyte injury via Dtk in IgAN.


Assuntos
Glomerulonefrite por IGA/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células Cultivadas , Células Endoteliais/metabolismo , Glomerulonefrite por IGA/patologia , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Células Mesangiais/metabolismo , Podócitos/metabolismo
20.
J Biol Chem ; 287(24): 20430-42, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22474292

RESUMO

Activation of mesangial cells (MCs), which is characterized by induction of smooth muscle α-actin (SMA) expression, contributes to a key event in various renal diseases; however, the mechanisms controlling MC differentiation are still largely undefined. Activated Smad1 induced SMA in a dose-dependent manner in MCs. As a direct regulating molecule for SMA, we identified and characterized scleraxis (Scx) as a new phenotype modulator in advanced glycation end product (AGE)-exposed MCs. Scx physically associated with E12 and bound the E-box in the promoter of SMA and negatively regulated the AGE-induced SMA expression. Scx induced expression and secretion of bone morphogenetic protein 4 (BMP4), thereby controlling the Smad1 activation in AGE-treated MCs. In diabetic mice, Scx was concomitantly expressed with SMA in the glomeruli. Inhibitor of differentiation 1 (Id1) was further induced by extended treatment with AGE, thereby dislodging Scx from the SMA promoter. These data suggest that Scx and Id1 are involved in the BMP4-Smad1-SMA signal transduction pathway besides the TGFß1-Smad1-SMA signaling pathway and modulate phenotypic changes in MCs in diabetic nephropathy.


Assuntos
Actinas/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteína Morfogenética Óssea 4/biossíntese , Nefropatias Diabéticas/metabolismo , Mesângio Glomerular/metabolismo , Transdução de Sinais , Proteína Smad1/metabolismo , Actinas/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteína Morfogenética Óssea 4/genética , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Regulação da Expressão Gênica/genética , Mesângio Glomerular/patologia , Produtos Finais de Glicação Avançada/genética , Produtos Finais de Glicação Avançada/metabolismo , Proteína 1 Inibidora de Diferenciação/genética , Proteína 1 Inibidora de Diferenciação/metabolismo , Masculino , Camundongos , Elementos de Resposta/genética , Proteína Smad1/genética , Fator 3 de Transcrição/genética , Fator 3 de Transcrição/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA