Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther Methods Clin Dev ; 29: 329-346, 2023 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-37214315

RESUMO

Upscaling of kidney epithelial cells is crucial for renal regenerative medicine. Nonetheless, the adult kidney lacks a distinct stem cell hierarchy, limiting the ability to long-term propagate clonal populations of primary cells that retain renal identity. Toward this goal, we tested the paradigm of shifting the balance between differentiation and stemness in the kidney by introducing a single pluripotency factor, OCT4. Here we show that ectopic expression of OCT4 in human adult kidney epithelial cells (hKEpC) induces the cells to dedifferentiate, stably proliferate, and clonally emerge over many generations. Control hKEpC dedifferentiate, assume fibroblastic morphology, and completely lose clonogenic capacity. Analysis of gene expression and histone methylation patterns revealed that OCT4 represses the HNF1B gene module, which is critical for kidney epithelial differentiation, and concomitantly activates stemness-related pathways. OCT4-hKEpC can be long-term expanded in the dedifferentiated state that is primed for renal differentiation. Thus, when expanded OCT4-hKEpC are grown as kidney spheroids (OCT4-kSPH), they reactivate the HNF1B gene signature, redifferentiate, and efficiently generate renal structures in vivo. Hence, changes occurring in the cellular state of hKEpC following OCT4 induction, long-term propagation, and 3D aggregation afford rapid scale-up technology of primary renal tissue-forming cells.

2.
J Am Soc Nephrol ; 32(9): 2242-2254, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34112705

RESUMO

BACKGROUND: Although coronavirus disease 2019 (COVID-19) causes significan t morbidity, mainly from pulmonary involvement, extrapulmonary symptoms are also major componen ts of the disease. Kidney disease, usually presenting as AKI, is particularly severe among patients with COVID-19. It is unknown, however, whether such injury results from direct kidney infection with COVID-19's causative virus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), or from indirect mechanisms. METHODS: Using ex vivo cell models, we sought to analyze SARS-CoV-2 interactions with kidney tubular cells and assess direct tubular injury. These models comprised primary human kidney epithelial cells (derived from nephrectomies) and grown as either proliferating monolayers or quiescent three-dimensional kidney spheroids. RESULTS: We demonstrated that viral entry molecules and high baseline levels of type 1 IFN-related molecules were present in monolayers and kidney spheroids. Although both models support viral infection and replication, they did not exhibit a cytopathic effect and cell death, outcomes that were strongly present in SARS-CoV-2-infected controls (African green monkey kidney clone E6 [Vero E6] cultures). A comparison of monolayer and spheroid cultures demonstrated higher infectivity and replication of SARS-CoV-2 in actively proliferating monolayers, although the spheroid cultures exhibited high er levels of ACE2. Monolayers exhibited elevation of some tubular injury molecules-including molecules related to fibrosis (COL1A1 and STAT6) and dedifferentiation (SNAI2)-and a loss of cell identity, evident by reduction in megalin (LRP2). The three-dimensional spheroids were less prone to such injury. CONCLUSIONS: SARS-CoV-2 can infect kidney cells without a cytopathic effect. AKI-induced cellular proliferation may potentially intensify infectivity and tubular damage by SARS-CoV-2, suggesting that early intervention in AKI is warranted to help minimize kidney infection.


Assuntos
Injúria Renal Aguda/etiologia , Injúria Renal Aguda/virologia , COVID-19/complicações , SARS-CoV-2/patogenicidade , Esferoides Celulares/virologia , Animais , Células Cultivadas , Chlorocebus aethiops , Estudos de Coortes , Efeito Citopatogênico Viral , Células Epiteliais/patologia , Células Epiteliais/virologia , Interações entre Hospedeiro e Microrganismos , Humanos , Interferon Tipo I/metabolismo , Rim/imunologia , Rim/patologia , Rim/virologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Modelos Biológicos , Pandemias , Receptores Virais/metabolismo , Estudos Retrospectivos , SARS-CoV-2/fisiologia , Esferoides Celulares/patologia , Células Vero , Replicação Viral
4.
Sci Rep ; 10(1): 22097, 2020 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-33328501

RESUMO

In-vivo single cell clonal analysis in the adult mouse kidney has previously shown lineage-restricted clonal proliferation within varying nephron segments as a mechanism responsible for cell replacement and local regeneration. To analyze ex-vivo clonal growth, we now preformed limiting dilution to generate genuine clonal cultures from one single human renal epithelial cell, which can give rise to up to 3.4 * 106 cells, and analyzed their characteristics using transcriptomics. A comparison between clonal cultures revealed restriction to either proximal or distal kidney sub-lineages with distinct cellular and molecular characteristics; rapidly amplifying de-differentiated clones and a stably proliferating cuboidal epithelial-appearing clones, respectively. Furthermore, each showed distinct molecular features including cell-cycle, epithelial-mesenchymal transition, oxidative phosphorylation, BMP signaling pathway and cell surface markers. In addition, analysis of clonal versus bulk cultures show early clones to be more quiescent, with elevated expression of renal developmental genes and overall reduction in renal identity markers, but with an overlapping expression of nephron segment identifiers and multiple identity. Thus, ex-vivo clonal growth mimics the in-vivo situation displaying lineage-restricted precursor characteristics of mature renal cells. These data suggest that for reconstruction of varying renal lineages with human adult kidney based organoid technology and kidney regeneration ex-vivo, use of multiple heterogeneous precursors is warranted.


Assuntos
Evolução Clonal/genética , Rim/crescimento & desenvolvimento , Mesoderma/crescimento & desenvolvimento , Regeneração/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Biologia Computacional , Células Epiteliais/citologia , Transição Epitelial-Mesenquimal/genética , Humanos , Rim/citologia , Mesoderma/metabolismo , Néfrons/crescimento & desenvolvimento , Néfrons/metabolismo , Cultura Primária de Células , Análise de Célula Única , Células-Tronco/citologia
5.
J Am Soc Nephrol ; 31(12): 2757-2772, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32753400

RESUMO

BACKGROUND: Cell-based therapies aimed at replenishing renal parenchyma have been proposed as an approach for treating CKD. However, pathogenic mechanisms involved in CKD such as renal hypoxia result in loss of kidney function and limit engraftment and therapeutic effects of renal epithelial progenitors. Jointly administering vessel-forming cells (human mesenchymal stromal cells [MSCs] and endothelial colony-forming cells [ECFCs]) may potentially result in in vivo formation of vascular networks. METHODS: We administered renal tubule-forming cells derived from human adult and fetal kidneys (previously shown to exert a functional effect in CKD mice) into mice, alongside MSCs and ECFCs. We then assessed whether this would result in generation of "renovascular units" comprising both vessels and tubules with potential interaction. RESULTS: Directly injecting vessel-forming cells and renal tubule-forming cells into the subcutaneous and subrenal capsular space resulted in self-organization of donor-derived vascular networks that connected to host vasculature, alongside renal tubules comprising tubular epithelia of different nephron segments. Vessels derived from MSCs and ECFCs augmented in vivo tubulogenesis by the renal tubule-forming cells. In vitro coculture experiments showed that MSCs and ECFCs induced self-renewal and genes associated with mesenchymal-epithelial transition in renal tubule-forming cells, indicating paracrine effects. Notably, after renal injury, renal tubule-forming cells and vessel-forming cells infused into the renal artery did not penetrate the renal vascular network to generate vessels; only administering them into the kidney parenchyma resulted in similar generation of human renovascular units in vivo. CONCLUSIONS: Combined cell therapy of vessel-forming cells and renal tubule-forming cells aimed at alleviating renal hypoxia and enhancing tubulogenesis holds promise as the basis for new renal regenerative therapies.


Assuntos
Células Endoteliais/citologia , Glomérulos Renais/citologia , Túbulos Renais/citologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Proliferação de Células , Terapia Baseada em Transplante de Células e Tecidos , Técnicas de Cocultura , Humanos , Camundongos , Neovascularização Fisiológica
6.
Cell Rep ; 30(3): 852-869.e4, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31968258

RESUMO

End-stage renal disease is a worldwide epidemic requiring renal replacement therapy. Harvesting tissue from failing kidneys and autotransplantation of tissue progenitors could theoretically delay the need for dialysis. Here we use healthy and end-stage human adult kidneys to robustly expand proliferative kidney epithelial cells and establish 3D kidney epithelial cultures termed "nephrospheres." Formation of nephrospheres reestablishes renal identity and function in primary cultures. Transplantation into NOD/SCID mice shows that nephrospheres restore self-organogenetic properties lost in monolayer cultures, allowing long-term engraftment as tubular structures, potentially adding nephron segments and demonstrating self-organization as critical to survival. Furthermore, long-term tubular engraftment of nephrospheres is functionally beneficial in murine models of chronic kidney disease. Remarkably, nephrospheres inhibit pro-fibrotic collagen production in cultured fibroblasts via paracrine modulation, while transplanted nephrospheres induce transcriptional signatures of proliferation and release from quiescence, suggesting re-activation of endogenous repair. These data support the use of human nephrospheres for renal cell therapy.


Assuntos
Rim/lesões , Rim/patologia , Esferoides Celulares/patologia , Cicatrização , Animais , Diferenciação Celular , Proliferação de Células , Doença Crônica , Modelos Animais de Doenças , Células Epiteliais/patologia , Fibrose , Humanos , Rim/fisiopatologia , Camundongos Endogâmicos NOD , Camundongos SCID , Insuficiência Renal Crônica/patologia , Esferoides Celulares/transplante
7.
Otol Neurotol ; 40(10): e1030-e1036, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31436634

RESUMO

HYPOTHESIS: The scarred rim of chronic tympanic membrane (TM) perforation contains keratinocytes with potential for regeneration while maintaining their morphological and genetic characteristics. BACKGROUND: The squamous epithelium of the TM has a good regeneration capacity. Successful isolation and expansion of human TM keratinocytes (hTMKR) was reported from a full, en-bloc, healthy TM. METHODS: Trimmed margins of the TM perforation (harvested during tympanoplasty) underwent enzymatic digestion (collagenase or trypsin) and were seeded either with serum-containing medium (SCM) or keratinocyte serum-free medium (KSFM) and progenitor cell growth medium (PR) (KSFM:PR, 1:1). Gene expression analysis by real-time qRT-PCR was used to compare between human TM cells derived from scarred perforation margins (hTMKR), normal human skin keratinocytes (NhSKR), and human fibroblasts. RESULTS: Twelve patients were included in the study. In 9 of 12 cases (75%) single-cell isolation with fibroblastic or epithelial cell morphology (or both) was achieved. Cells seeded with KSFM:PR yielded epithelial morphology (hTMKR) while SCM culturing resulted in a fibroblastic morphology (hTMFib). Gene expression analysis revealed significant higher expression of VCAN (p = 0.002) and FOXC2 (p = 0.015) at the mRNA levels (normal hTMKR markers) in hTMKR compared to NhSKR. In addition, a comparison of gene expression between hTMKR and hTMFib revealed significantly higher levels of both VCAN (p = 0.045) and SLC6A14 (p = 0.036) among hTMKR. CONCLUSION: For the first time, we developed a protocol to isolate hTMKR from scarred TM perforation margins. Furthermore, we succeeded in achieving tissue expansion that preserved the characteristic of healthy TM cells. This study bridges "regenerative medicine" approach with clinical and surgical objectives.


Assuntos
Técnicas de Cultura de Células/métodos , Cicatriz , Células Epiteliais/citologia , Queratinócitos/citologia , Células-Tronco/citologia , Técnicas de Cultura de Tecidos/métodos , Perfuração da Membrana Timpânica/complicações , Membrana Timpânica , Timpanoplastia/métodos , Adolescente , Adulto , Idoso , Sistemas de Transporte de Aminoácidos , Criança , Cicatriz/patologia , Cicatriz/cirurgia , Estudos de Viabilidade , Feminino , Fibroblastos , Humanos , Masculino , Pessoa de Meia-Idade , Medicina Regenerativa/métodos , Coleta de Tecidos e Órgãos/métodos , Membrana Timpânica/citologia , Membrana Timpânica/patologia , Adulto Jovem
8.
Kidney Int Rep ; 3(1): 155-159, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29340326

RESUMO

INTRODUCTION: Renal angiomyolipoma (AML) is the most common benign renal tumor. Despite a generally benign histology, AML can result in significant morbidity, from intra-abdominal hemorrhage and reduction in kidney function. While classically associated with the autosomal dominant disorder tuberous sclerosis complex (TSC) or with pulmonary lymphangioleiomyomatosis, most AMLs are sporadic. Mammalian target of rapamycin complex 1 (mTORC1) inhibitors (e.g., sirolimus) have been found to be effective in treating TSC- or lymphangioleiomyomatosis-associated AML, but to date it is unknown whether this strategy is effective for sporadic AML. METHODS: We stained tumor specimens of sporadic AML patients for pS6 to assess for mTORC1 activation. RESULTS: We detected strong activation of the mTORC1 pathway, similar to TSC-associated AML. Consequently, we showed that in vitro treatment with sirolimus results in significant growth inhibition of the human sporadic AML cell line SV7Tert, similar to the effect seen when the same treatment is applied to the human TSC-associated AML cell line UMBSV-tel. To further investigate the potential of mTORC1 inhibition for treating sporadic AML and assess whether the in vitro results are clinically relevant, we identified a patient with sporadic, bilateral AMLs, showing continued tumor growth following a partial nephrectomy. Using immunostaining, we detected strong mTORC1 activation in the patient's AML tissue. Accordingly, upon treatment with sirolimus, we noted significant reduction in the patient's tumor volume and resolution of hydronephrosis, without any significant side effects. CONCLUSION: We propose mTORC1 inhibition as an effective treatment option for patients with sporadic AML, which represents the vast majority of patients with this tumor.

9.
Pediatr Res ; 83(1-2): 267-274, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28985199

RESUMO

The mammalian kidney is a highly complex organ, composed of various cell types within a unique structural framework. Nonetheless, in recent years, giant leaps in our understanding of nephrogenesis and the origin of new cells in the adult kidney have resulted in novel routes to regenerate damaged nephrons. While several strategies can be envisioned to achieve this aim, one common theme is the reliance on renal lineage cells, as extrarenal cells, such as bone marrow-derived cells, have been shown to be devoid of renal differentiation capacity. Herein, we will present the main motivation for the pursuit for cell-based therapies, which is the ever growing problem of chronic kidney disease (CKD), and discuss different strategies toward replenishing the damaged renal parenchyma. These include transplantation of fetal kidney grafts or fetal kidney stem cells, directed differentiation of pluripotent stem cells into kidney epithelia, establishment of renal progenitors from the adult kidney, and genetic reprogramming of mature kidney cells into a progenitor state. Taken together with novel techniques recapitulating the three-dimensional developmental environment, these advances are expected to take the field into a new era, bringing us closer than ever to the day when kidney stem cell-based therapy becomes a viable therapeutic option.


Assuntos
Falência Renal Crônica/terapia , Rim/citologia , Regeneração , Animais , Diferenciação Celular , Linhagem da Célula , Células Epiteliais/citologia , Homeostase , Humanos , Transplante de Rim , Camundongos , Néfrons/metabolismo , Organogênese , Células-Tronco Pluripotentes/citologia , Qualidade de Vida , Medicina Regenerativa , Transplante de Células-Tronco , Células-Tronco/citologia
11.
Stem Cell Reports ; 9(1): 279-291, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28552604

RESUMO

During nephrogenesis, stem/progenitor cells differentiate and give rise to early nephron structures that segment to proximal and distal nephron cell types. Previously, we prospectively isolated progenitors from human fetal kidney (hFK) utilizing a combination of surface markers. However, upon culture nephron progenitors differentiated and could not be robustly maintained in vitro. Here, by culturing hFK in a modified medium used for in vitro growth of mouse nephron progenitors, and by dissection of NCAM+/CD133- progenitor cells according to EpCAM expression (NCAM+/CD133-/EpCAM-, NCAM+/CD133-/EpCAMdim, NCAM+/CD133-/EpCAMbright), we show at single-cell resolution a preservation of uninduced and induced cap mesenchyme as well as a transitioning mesenchymal-epithelial state. Concomitantly, differentiating and differentiated epithelial lineages are also maintained. In vitro expansion of discrete stages of early human nephrogenesis in nephron stem cell cultures may be used for drug screening on a full repertoire of developing kidney cells and for prospective isolation of mesenchymal or epithelial renal lineages for regenerative medicine.


Assuntos
Células-Tronco Embrionárias Humanas/citologia , Rim/citologia , Néfrons/citologia , Néfrons/crescimento & desenvolvimento , Antígeno AC133/análise , Técnicas de Cultura de Células , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Molécula de Adesão da Célula Epitelial/análise , Humanos , Organogênese , Análise de Célula Única
12.
EMBO Mol Med ; 9(4): 508-530, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28275008

RESUMO

Angiomyolipoma (AML), the most common benign renal tumor, can result in severe morbidity from hemorrhage and renal failure. While mTORC1 activation is involved in its growth, mTORC1 inhibitors fail to eradicate AML, highlighting the need for new therapies. Moreover, the identity of the AML cell of origin is obscure. AML research, however, is hampered by the lack of in vivo models. Here, we establish a human AML-xenograft (Xn) model in mice, recapitulating AML at the histological and molecular levels. Microarray analysis demonstrated tumor growth in vivo to involve robust PPARγ-pathway activation. Similarly, immunostaining revealed strong PPARγ expression in human AML specimens. Accordingly, we demonstrate that while PPARγ agonism accelerates AML growth, PPARγ antagonism is inhibitory, strongly suppressing AML proliferation and tumor-initiating capacity, via a TGFB-mediated inhibition of PDGFB and CTGF. Finally, we show striking similarity between AML cell lines and mesenchymal stem cells (MSCs) in terms of antigen and gene expression and differentiation potential. Altogether, we establish the first in vivo human AML model, which provides evidence that AML may originate in a PPARγ-activated renal MSC lineage that is skewed toward adipocytes and smooth muscle and away from osteoblasts, and uncover PPARγ as a regulator of AML growth, which could serve as an attractive therapeutic target.


Assuntos
Angiomiolipoma/patologia , PPAR gama/metabolismo , Animais , Linhagem Celular Tumoral , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Perfilação da Expressão Gênica , Humanos , Células-Tronco Mesenquimais , Camundongos , Proteínas Proto-Oncogênicas c-sis/metabolismo , Terapêutica , Fator de Crescimento Transformador beta/metabolismo
13.
Sci Rep ; 6: 23562, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-27020553

RESUMO

When assembling a nephron during development a multipotent stem cell pool becomes restricted as differentiation ensues. A faulty differentiation arrest in this process leads to transformation and initiation of a Wilms' tumor. Mapping these transitions with respective surface markers affords accessibility to specific cell subpopulations. NCAM1 and CD133 have been previously suggested to mark human renal progenitor populations. Herein, using cell sorting, RNA sequencing, in vitro studies with serum-free media and in vivo xenotransplantation we demonstrate a sequential map that links human kidney development and tumorigenesis; In nephrogenesis, NCAM1(+)CD133(-) marks SIX2(+) multipotent renal stem cells transiting to NCAM1(+)CD133(+) differentiating segment-specific SIX2(-) epithelial progenitors and NCAM1(-)CD133(+) differentiated nephron cells. In tumorigenesis, NCAM1(+)CD133(-) marks SIX2(+) blastema that includes the ALDH1(+) WT cancer stem/initiating cells, while NCAM1(+)CD133(+) and NCAM1(-)CD133(+) specifying early and late epithelial differentiation, are severely restricted in tumor initiation capacity and tumor self-renewal. Thus, negative selection for CD133 is required for defining NCAM1(+) nephron stem cells in normal and malignant nephrogenesis.


Assuntos
Biomarcadores/metabolismo , Carcinogênese/genética , Rim/metabolismo , Células-Tronco Neoplásicas/metabolismo , Néfrons/metabolismo , Células-Tronco/metabolismo , Antígeno AC133/genética , Antígeno AC133/metabolismo , Animais , Antígeno CD56/genética , Antígeno CD56/metabolismo , Carcinogênese/metabolismo , Células Cultivadas , Criança , Pré-Escolar , Feminino , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Lactente , Rim/embriologia , Masculino , Camundongos Endogâmicos NOD , Células-Tronco Neoplásicas/patologia , Néfrons/citologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Organogênese/genética , Estudos Prospectivos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo , Células Tumorais Cultivadas
14.
Stem Cell Reports ; 3(1): 24-33, 2014 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-25068119

RESUMO

An open question remains in cancer stem cell (CSC) biology whether CSCs are by definition at the top of the differentiation hierarchy of the tumor. Wilms' tumor (WT), composed of blastema and differentiated renal elements resembling the nephrogenic zone of the developing kidney, is a valuable model for studying this question because early kidney differentiation is well characterized. WT neural cell adhesion molecule 1-positive (NCAM1(+)) aldehyde dehydrogenase 1-positive (ALDH1(+)) CSCs have been recently isolated and shown to harbor early renal progenitor traits. Herein, by generating pure blastema WT xenografts, composed solely of cells expressing the renal developmental markers SIX2 and NCAM1, we surprisingly show that sorted ALDH1(+) WT CSCs do not correspond to earliest renal stem cells. Rather, gene expression and proteomic comparative analyses disclose a cell type skewed more toward epithelial differentiation than the bulk of the blastema. Thus, WT CSCs are likely to dedifferentiate to propagate WT blastema.


Assuntos
Células-Tronco Neoplásicas/patologia , Tumor de Wilms/metabolismo , Tumor de Wilms/patologia , Família Aldeído Desidrogenase 1 , Animais , Antígeno CD56/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Feminino , Humanos , Isoenzimas/metabolismo , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Camundongos , Camundongos SCID , Modelos Biológicos , Células-Tronco Neoplásicas/metabolismo , Retinal Desidrogenase/metabolismo
15.
Am J Pathol ; 183(5): 1621-1633, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24055371

RESUMO

The nephron is composed of a monolayer of epithelial cells that make up its various compartments. In development, these cells begin as mesenchyme. NCAM1, abundant in the mesenchyme and early nephron lineage, ceases to express in mature kidney epithelia. We show that, once placed in culture and released from quiescence, adult human kidney epithelial cells (hKEpCs), uniformly positive for CD24/CD133, re-express NCAM1 in a specific cell subset that attains a stem/progenitor state. Immunosorted NCAM1(+) cells overexpressed early nephron progenitor markers (PAX2, SALL1, SIX2, WT1) and acquired a mesenchymal fate, indicated by high vimentim and reduced E-cadherin levels. Gene expression and microarray analysis disclosed both a proximal tubular origin of these cells and molecules regulating epithelial-mesenchymal transition. NCAM1(+) cells generated clonal progeny when cultured in the presence of fetal kidney conditioned medium, differentiated along mesenchymal lineages but retained the unique propensity to generate epithelial kidney spheres and produce epithelial renal tissue on single-cell grafting in chick CAM and mouse. Depletion of NCAM1(+) cells from hKEpCs abrogated stemness traits in vitro. Eliminating these cells during the regenerative response that follows glycerol-induced acute tubular necrosis worsened peak renal injury in vivo. Thus, higher clone-forming and developmental capacities characterize a distinct subset of adult kidney-derived cells. The ability to influence an endogenous regenerative response via NCAM1 targeting may lead to novel therapeutics for renal diseases.


Assuntos
Antígeno CD56/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Rim/patologia , Células-Tronco/metabolismo , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Adulto , Animais , Anticorpos/metabolismo , Biomarcadores/metabolismo , Nitrogênio da Ureia Sanguínea , Diferenciação Celular/genética , Proliferação de Células , Galinhas , Células Clonais , Regulação para Baixo/genética , Ontologia Genética , Células HEK293 , Humanos , Mesoderma/patologia , Camundongos , Anotação de Sequência Molecular , Néfrons/metabolismo , Néfrons/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Transcriptoma/genética , Regulação para Cima/genética
16.
EMBO Mol Med ; 5(10): 1556-68, 2013 10.
Artigo em Inglês | MEDLINE | ID: mdl-23996934

RESUMO

Identification of tissue-specific renal stem/progenitor cells with nephrogenic potential is a critical step in developing cell-based therapies for renal disease. In the human kidney, stem/progenitor cells are induced into the nephrogenic pathway to form nephrons until the 34 week of gestation, and no equivalent cell types can be traced in the adult kidney. Human nephron progenitor cells (hNPCs) have yet to be isolated. Here we show that growth of human foetal kidneys in serum-free defined conditions and prospective isolation of NCAM1(+) cells selects for nephron lineage that includes the SIX2-positive cap mesenchyme cells identifying a mitotically active population with in vitro clonogenic and stem/progenitor properties. After transplantation in the chick embryo, these cells-but not differentiated counterparts-efficiently formed various nephron tubule types. hNPCs engrafted and integrated in diseased murine kidneys and treatment of renal failure in the 5/6 nephrectomy kidney injury model had beneficial effects on renal function halting disease progression. These findings constitute the first definition of an intrinsic nephron precursor population, with major potential for cell-based therapeutic strategies and modelling of kidney disease.


Assuntos
Néfrons/citologia , Insuficiência Renal Crônica/cirurgia , Transplante de Células-Tronco , Células-Tronco/citologia , Animais , Antígeno CD56/metabolismo , Células Cultivadas , Embrião de Galinha , Galinhas , Membrana Corioalantoide/metabolismo , Membrana Corioalantoide/patologia , Embrião de Mamíferos/citologia , Feminino , Proteínas de Homeodomínio/metabolismo , Humanos , Túbulos Renais/patologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco/metabolismo
17.
Cell Reprogram ; 15(4): 281-92, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23841748

RESUMO

Recent studies have suggested that epigenetic modulation with chromatin-modifying agents can induce stemness and dedifferentiation and increase developmental plasticity. For instance, valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, has been shown to promote self-renewal/expansion of hematopoietic stem cells and facilitate the generation of induced pluripotent stem cells (iPSCs). Previously, we observed that downregulation of embryonic renal stem/progenitor genes in the adult kidney was associated, at least in part, with epigenetic silencing. Therefore, we hypothesized that VPA may alter the expression of these genes and reprogram mature human adult kidney epithelial cells (hKEpCs) to a stem/progenitor-like state. Here, using quantitative RT-PCR and flow cytometry [fluorescence-activated cell sorting (FACS)] analysis, we show in VPA-treated primary cultures of human adult and fetal kidney significant reinduction of the renal stem/progenitor markers SIX2, OSR1, SALL1, NCAM, and PSA-NCAM. Robust SIX2 mRNA re-expression was confirmed at the protein level by western blot and was associated with epigenetic changes of the histones at multiple sites of the SIX2 promoter leading to gene activation, significantly increased acetylation of histones H4, and methylation of lysine 4 on H3. Furthermore, we could demonstrate synergistic effects of VPA and Wnt antagonists on SIX2 and also OSR1 reinduction. Nevertheless, VPA resulted in upregulation of E-CADHERIN and reduction in VIMENTIN, preventing the skewing of hKEpCs towards a more replicative mesenchymal state required for clonogenic expansion and acquisition of stem cell characters, altogether inducing cell senescence at early passages. These results demonstrating that chromatin-modifying agents prevent dedifferentiation of hKEpCs have important clinical implications as they may limit ex-vivo self-renewal/expansion and possibly the in vivo renal regenerative capacity initiated by dedifferentiation.


Assuntos
Desdiferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Rim/embriologia , Ácido Valproico/farmacologia , Adulto , Animais , Desdiferenciação Celular/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Embrião de Galinha , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Células-Tronco Embrionárias/fisiologia , Células Epiteliais/fisiologia , Feto/citologia , Humanos , Rim/citologia
18.
J Am Soc Nephrol ; 24(4): 550-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23520208

RESUMO

Abnormal differentiation of the renal stem/progenitor pool into kidney tissue can lead to renal hypodysplasia (RHD), but the underlying causes of RHD are not well understood. In this multicenter study, we identified 20 Israeli pedigrees with isolated familial, nonsyndromic RHD and screened for mutations in candidate genes involved in kidney development, including PAX2, HNF1B, EYA1, SIX1, SIX2, SALL1, GDNF, WNT4, and WT1. In addition to previously reported RHD-causing genes, we found that two affected brothers were heterozygous for a missense variant in the WNT4 gene. Functional analysis of this variant revealed both antagonistic and agonistic canonical WNT stimuli, dependent on cell type. In HEK293 cells, WNT4 inhibited WNT3A induced canonical activation, and the WNT4 variant significantly enhanced this inhibition of the canonical WNT pathway. In contrast, in primary cultures of human fetal kidney cells, which maintain WNT activation and more closely represent WNT signaling in renal progenitors during nephrogenesis, this mutation caused significant loss of function, resulting in diminished canonical WNT/ß-catenin signaling. In conclusion, heterozygous WNT4 variants are likely to play a causative role in renal hypodysplasia.


Assuntos
Diferenciação Celular/genética , Nefropatias/genética , Via de Sinalização Wnt/genética , Proteína Wnt4/genética , Adolescente , Criança , Pré-Escolar , Feminino , Células HEK293 , Humanos , Lactente , Israel , Masculino , Mutação , Fator de Transcrição PAX2/genética , Análise de Sequência de DNA , Adulto Jovem
19.
Tissue Eng Part A ; 17(17-18): 2305-19, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21542667

RESUMO

Cell-based approaches utilizing autologous human renal cells require their isolation, expansion in vitro, and reintroduction back into the host for renal tissue regeneration. Nevertheless, human kidney epithelial cells (hKEpCs) lose their phenotype, dedifferentiate, and assume the appearance of fibroblasts after relatively few passages in culture. We hypothesized that growth conditions may influence hKEpC phenotype and function. hKEpCs retrieved from human nephrectomy tissue samples showed the ability to reproducibly form kidney spheres when grown in suspension culture developed in nonadherent conditions. Genetic labeling and time-lapse microscopy indicated, at least in part, the aggregation of hKEpCs into 3D spheroids rather than formation of pure clonally expanded spheres. Characterization of hKEpC spheroids by real-time polymerase chain reaction and FACS analysis showed upregulation of some renal developmental and "stemness" markers compared with monolayer and mostly an EpCAM(+)CD24(+)CD133(+)CD44(+) spheroid cell phenotype. Oligonucleotide microarrays, which were used to identify global transcriptional changes accompanying spheroid formation, showed predominantly upregulation of cell matrix/cell contact molecules and cellular biogenesis processes and downregulation of cell cycle, growth, and locomotion. Accordingly, hKEpC spheroids slowly proliferated as indicated by low Ki-67 staining, but when grafted in low cell numbers onto the chorioallantoic membrane (CAM) of the chick embryo, they exclusively reconstituted various renal tubular epithelia. Moreover, efficient generation of kidney spheroids was observed after long-term monolayer culture resulting in reestablishment of tubulogenic capacity upon CAM grafting. Thus, generation of a tubular organoid in hKEpC spheroids may provide a functional benefit for kidney-derived cells in vivo.


Assuntos
Rim/citologia , Organoides/citologia , Esferoides Celulares/citologia , Técnicas de Cultura de Células , Células Cultivadas , Citometria de Fluxo , Humanos , Análise em Microsséries , Esferoides Celulares/metabolismo
20.
PLoS One ; 4(8): e6709, 2009 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-19696931

RESUMO

In the human fetal kidney (HFK) self-renewing stem cells residing in the metanephric mesenchyme (MM)/blastema are induced to form all cell types of the nephron till 34(th) week of gestation. Definition of useful markers is crucial for the identification of HFK stem cells. Because wilms' tumor, a pediatric renal cancer, initiates from retention of renal stem cells, we hypothesized that surface antigens previously up-regulated in microarrays of both HFK and blastema-enriched stem-like wilms' tumor xenografts (NCAM, ACVRIIB, DLK1/PREF, GPR39, FZD7, FZD2, NTRK2) are likely to be relevant markers. Comprehensive profiling of these putative and of additional stem cell markers (CD34, CD133, c-Kit, CD90, CD105, CD24) in mid-gestation HFK was performed using immunostaining and FACS in conjunction with EpCAM, an epithelial surface marker that is absent from the MM and increases along nephron differentiation and hence can be separated into negative, dim or bright fractions. No marker was specifically localized to the MM. Nevertheless, FZD7 and NTRK2 were preferentially localized to the MM and emerging tubules (<10% of HFK cells) and were mostly present within the EpCAM(neg) and EpCAM(dim) fractions, indicating putative stem/progenitor markers. In contrast, single markers such as CD24 and CD133 as well as double-positive CD24(+)CD133(+) cells comprise >50% of HFK cells and predominantly co-express EpCAM(bright), indicating they are mostly markers of differentiation. Furthermore, localization of NCAM exclusively in the MM and in its nephron progenitor derivatives but also in stroma and the expression pattern of significantly elevated renal stem/progenitor genes Six2, Wt1, Cited1, and Sall1 in NCAM(+)EpCAM(-) and to a lesser extent in NCAM(+)EpCAM(+) fractions confirmed regional identity of cells and assisted us in pinpointing the presence of subpopulations that are putative MM-derived progenitor cells (NCAM(+)EpCAM(+)FZD7(+)), MM stem cells (NCAM(+)EpCAM(-)FZD7(+)) or both (NCAM(+)FZD7(+)). These results and concepts provide a framework for developing cell selection strategies for human renal cell-based therapies.


Assuntos
Biomarcadores/metabolismo , Rim/embriologia , Células-Tronco/citologia , Citometria de Fluxo , Humanos , Rim/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...