Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Sci Transl Med ; 12(536)2020 03 25.
Article in English | MEDLINE | ID: mdl-32213630

ABSTRACT

Acute tissue injury causes DNA damage and repair processes involving increased cell mitosis and polyploidization, leading to cell function alterations that may potentially drive cancer development. Here, we show that acute kidney injury (AKI) increased the risk for papillary renal cell carcinoma (pRCC) development and tumor relapse in humans as confirmed by data collected from several single-center and multicentric studies. Lineage tracing of tubular epithelial cells (TECs) after AKI induction and long-term follow-up in mice showed time-dependent onset of clonal papillary tumors in an adenoma-carcinoma sequence. Among AKI-related pathways, NOTCH1 overexpression in human pRCC associated with worse outcome and was specific for type 2 pRCC. Mice overexpressing NOTCH1 in TECs developed papillary adenomas and type 2 pRCCs, and AKI accelerated this process. Lineage tracing in mice identified single renal progenitors as the cell of origin of papillary tumors. Single-cell RNA sequencing showed that human renal progenitor transcriptome showed similarities to PT1, the putative cell of origin of human pRCC. Furthermore, NOTCH1 overexpression in cultured human renal progenitor cells induced tumor-like 3D growth. Thus, AKI can drive tumorigenesis from local tissue progenitor cells. In particular, we find that AKI promotes the development of pRCC from single progenitors through a classical adenoma-carcinoma sequence.


Subject(s)
Acute Kidney Injury , Adenoma , Carcinoma, Renal Cell , Kidney Neoplasms , Adenoma/genetics , Animals , Biomarkers, Tumor , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Mice , Neoplasm Recurrence, Local , Stem Cells
2.
Nat Commun ; 9(1): 1344, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29632300

ABSTRACT

Acute kidney injury (AKI) is considered largely reversible based on the capacity of surviving tubular cells to dedifferentiate and replace lost cells via cell division. Here we show by tracking individual tubular cells in conditional Pax8/Confetti mice that kidney function is  recovered after AKI despite substantial tubular cell loss. Cell cycle and ploidy analysis upon AKI in conditional Pax8/FUCCI2aR mice and human biopsies identify endocycle-mediated hypertrophy of tubular cells. By contrast, a small subset of Pax2+ tubular progenitors enriches via higher stress resistance and clonal expansion and regenerates necrotic tubule segments, a process that can be enhanced by suitable drugs. Thus,  renal functional recovery upon AKI involves remnant tubular cell hypertrophy via endocycle and limited progenitor-driven regeneration that can be pharmacologically enhanced.


Subject(s)
Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Acute Kidney Injury/genetics , Adult Stem Cells/pathology , Animals , Cell Cycle , Cell Dedifferentiation , Cell Enlargement , Cell Lineage , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Histone Deacetylase Inhibitors/pharmacology , Humans , Kidney Tubules/drug effects , Kidney Tubules/pathology , Kidney Tubules/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , PAX2 Transcription Factor/metabolism , PAX8 Transcription Factor/metabolism , Ploidies , Regeneration/drug effects , Single-Cell Analysis
3.
Stem Cells Int ; 2016: 4798639, 2016.
Article in English | MEDLINE | ID: mdl-27721835

ABSTRACT

Mesenchymal stem cells form a population of self-renewing, multipotent cells that can be isolated from several tissues. Multiple preclinical studies have demonstrated that the administration of exogenous MSC could prevent renal injury and could promote renal recovery through a series of complex mechanisms, in particular via immunomodulation of the immune system and release of paracrine factors and microvesicles. Due to their therapeutic potentials, MSC are being evaluated as a possible player in treatment of human kidney disease, and an increasing number of clinical trials to assess the safety, feasibility, and efficacy of MSC-based therapy in various kidney diseases have been proposed. In the present review, we will summarize the current knowledge on MSC infusion to treat acute kidney injury, chronic kidney disease, diabetic nephropathy, focal segmental glomerulosclerosis, systemic lupus erythematosus, and kidney transplantation. The data obtained from these clinical trials will provide further insight into safety, feasibility, and efficacy of MSC-based therapy in renal pathologies and allow the design of consensus protocol for clinical purpose.

4.
Stem Cells Int ; 2016: 8342625, 2016.
Article in English | MEDLINE | ID: mdl-27293448

ABSTRACT

Renal cell carcinoma (RCC) is a major neoplasm with high incidence in western countries. Tumors are heterogeneous and are composed of differentiated cancer cells, stromal cells, and cancer stem cells (CSCs). CSCs possess two main properties: self-renewal and proliferation. Additionally, they can generate new tumors once transplanted into immunodeficient mice. Several approaches have been described to identify them, through the expression of cell markers, functional assays, or a combination of both. As CSCs are involved in the resistance mechanisms to radio- and chemotherapies, several new strategies have been proposed to directly target CSCs in RCC. One approach drives CSCs to differentiate into cancer cells sensitive to conventional treatments, while the other proposes to eradicate them selectively. A series of innovative therapies aiming at eliminating CSCs have been designed to treat other types of cancer and have not been experimented with on RCC yet, but they reveal themselves to be promising. In conclusion, CSCs are an important player in carcinogenesis and represent a valid target for therapy in RCC patients.

5.
Stem Cell Reports ; 5(2): 248-63, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26235895

ABSTRACT

Podocyte loss is a general mechanism of glomerular dysfunction that initiates and drives the progression of chronic kidney disease, which affects 10% of the world population. Here, we evaluate whether the regenerative response to podocyte injury influences chronic kidney disease outcome. In models of focal segmental glomerulosclerosis performed in inducible transgenic mice where podocytes are tagged, remission or progression of disease was determined by the amount of regenerated podocytes. When the same model was established in inducible transgenic mice where renal progenitors are tagged, the disease remitted if renal progenitors successfully differentiated into podocytes, while it persisted if differentiation was ineffective, resulting in glomerulosclerosis. Treatment with BIO, a GSK3s inhibitor, significantly increased disease remission by enhancing renal progenitor sensitivity to the differentiation effect of endogenous retinoic acid. These results establish renal progenitors as critical determinants of glomerular disease outcome and a pharmacological enhancement of their differentiation as a possible therapeutic strategy.


Subject(s)
Cell Differentiation , Podocytes/cytology , Regeneration , Renal Insufficiency, Chronic/pathology , Stem Cells/cytology , Animals , Cells, Cultured , Glycogen Synthase Kinase 3/antagonists & inhibitors , Indoles/pharmacology , Indoles/therapeutic use , Mice , Mice, Inbred C57BL , Oximes/pharmacology , Oximes/therapeutic use , Podocytes/drug effects , Podocytes/metabolism , Renal Insufficiency, Chronic/drug therapy , Stem Cells/drug effects , Stem Cells/metabolism
6.
J Am Soc Nephrol ; 26(8): 1961-74, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25568173

ABSTRACT

The critical role of genetic and epigenetic factors in the pathogenesis of kidney disorders is gradually becoming clear, and the need for disease models that recapitulate human kidney disorders in a personalized manner is paramount. In this study, we describe a method to select and amplify renal progenitor cultures from the urine of patients with kidney disorders. Urine-derived human renal progenitors exhibited phenotype and functional properties identical to those purified from kidney tissue, including the capacity to differentiate into tubular cells and podocytes, as demonstrated by confocal microscopy, Western blot analysis of podocyte-specific proteins, and scanning electron microscopy. Lineage tracing studies performed with conditional transgenic mice, in which podocytes are irreversibly tagged upon tamoxifen treatment (NPHS2.iCreER;mT/mG), that were subjected to doxorubicin nephropathy demonstrated that renal progenitors are the only urinary cell population that can be amplified in long-term culture. To validate the use of these cells for personalized modeling of kidney disorders, renal progenitors were obtained from (1) the urine of children with nephrotic syndrome and carrying potentially pathogenic mutations in genes encoding for podocyte proteins and (2) the urine of children without genetic alterations, as validated by next-generation sequencing. Renal progenitors obtained from patients carrying pathogenic mutations generated podocytes that exhibited an abnormal cytoskeleton structure and functional abnormalities compared with those obtained from patients with proteinuria but without genetic mutations. The results of this study demonstrate that urine-derived patient-specific renal progenitor cultures may be an innovative research tool for modeling of genetic kidney disorders.


Subject(s)
Cell Culture Techniques , Kidney Diseases/congenital , Kidney/cytology , Stem Cells/cytology , Urine/cytology , Adolescent , Animals , Case-Control Studies , Child , Child, Preschool , Female , Humans , Infant , Male , Mice, Inbred BALB C , Mice, SCID , Mice, Transgenic
7.
J Am Soc Nephrol ; 24(11): 1756-68, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23949798

ABSTRACT

In CKD, the risk of kidney failure and death depends on the severity of proteinuria, which correlates with the extent of podocyte loss and glomerular scarring. We investigated whether proteinuria contributes directly to progressive glomerulosclerosis through the suppression of podocyte regeneration and found that individual components of proteinuria exert distinct effects on renal progenitor survival and differentiation toward a podocyte lineage. In particular, albumin prevented podocyte differentiation from human renal progenitors in vitro by sequestering retinoic acid, thus impairing retinoic acid response element (RARE)-mediated transcription of podocyte-specific genes. In mice with Adriamycin nephropathy, a model of human FSGS, blocking endogenous retinoic acid synthesis increased proteinuria and exacerbated glomerulosclerosis. This effect was related to a reduction in podocyte number, as validated through genetic podocyte labeling in NPHS2.Cre;mT/mG transgenic mice. In RARE-lacZ transgenic mice, albuminuria reduced retinoic acid bioavailability and impaired RARE activation in renal progenitors, inhibiting their differentiation into podocytes. Treatment with retinoic acid restored RARE activity and induced the expression of podocyte markers in renal progenitors, decreasing proteinuria and increasing podocyte number, as demonstrated in serial biopsy specimens. These results suggest that albumin loss through the damaged filtration barrier impairs podocyte regeneration by sequestering retinoic acid and promotes the generation of FSGS lesions. Our findings may explain why reducing proteinuria delays CKD progression and provide a biologic rationale for the clinical use of pharmacologic modulators to induce regression of glomerular diseases.


Subject(s)
Albuminuria/complications , Podocytes/physiology , Regeneration , Tretinoin/metabolism , Albuminuria/pathology , Animals , Cells, Cultured , Female , Glomerulosclerosis, Focal Segmental/etiology , Humans , Mice , Mice, SCID , Response Elements/physiology , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL