Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sara Pri-Chen is active.

Publication


Featured researches published by Sara Pri-Chen.


Embo Molecular Medicine | 2013

Identification of human nephron progenitors capable of generation of kidney structures and functional repair of chronic renal disease.

Orit Harari-Steinberg; Sally Metsuyanim; Dorit Omer; Yehudit Gnatek; Rotem Gershon; Sara Pri-Chen; Derya D. Ozdemir; Yaniv Lerenthal; Tzahi Noiman; Herzel Ben-Hur; Zvi Vaknin; David Schneider; Bruce J. Aronow; Ronald S. Goldstein; Peter Hohenstein; Benjamin Dekel

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.


American Journal of Pathology | 2013

Reactivation of NCAM1 Defines a Subpopulation of Human Adult Kidney Epithelial Cells with Clonogenic and Stem/Progenitor Properties

Ella Buzhor; Dorit Omer; Orit Harari-Steinberg; Zohar Dotan; Einav Vax; Sara Pri-Chen; Sally Metsuyanim; Oren Pleniceanu; Ronald S. Goldstein; Benjamin Dekel

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.


Scientific Reports | 2016

Dissecting Stages of Human Kidney Development and Tumorigenesis with Surface Markers Affords Simple Prospective Purification of Nephron Stem Cells

Naomi Pode-Shakked; Oren Pleniceanu; Rotem Gershon; Rachel Shukrun; Itamar Kanter; Efrat Bucris; Ben Pode-Shakked; Gal Tam; Hadar Tam; Revital Caspi; Sara Pri-Chen; Einav Vax; Guy Katz; Dorit Omer; Orit Harari-Steinberg; Tomer Kalisky; Benjamin Dekel

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.


Stem cell reports | 2014

Wilms’ Tumor Blastemal Stem Cells Dedifferentiate to Propagate the Tumor Bulk

Rachel Shukrun; Naomi Pode-Shakked; Oren Pleniceanu; Dorit Omer; Einav Vax; Eyal Peer; Sara Pri-Chen; Jasmine Jacob; Qianghua Hu; Orit Harari-Steinberg; Vicki Huff; Benjamin Dekel

Summary 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.


Current Eye Research | 2012

Avastin Treatment Reduces Retinal Neovascularization in a Mouse Model of Retinopathy of Prematurity

Ronen Rabinowitz; Ayelet Priel; Mordechai Rosner; Sara Pri-Chen; Abraham Spierer

Purpose: This study was designed to evaluate the effect of one intraperitoneal (IP) injection of bevacizumab (Avastin) on the severity of oxygen-induced retinopathy (OIR) in a mouse model. Materials and methods: Twenty-eight eyes of 14 mice with OIR were studied. There were nine mice in the bevacizumab-treated group (study group) and five mice in the saline-treated group (controls). The mouse OIR model consisted of a 5-day exposure to 75% oxygen. On postnatal day 12 (P12), Avastin 2.5 mg/kg was administered IP to the study group and 2.5 mg/kg normal saline was administered IP to the controls. All 14 mice underwent fluorescein angiography of the retinal vasculature on P17 and the following parameters were scored (Modified Retinopathy Scoring System, MRSS): blood vessel growth, formation of blood vessel tufts, extraretinal neovascularization, degree of central constriction, and tortuosity of vessels. In addition, the neovascular vessels were quantified on the hematoxylin and eosin (H&S)-stained paraffin sections of the eyes in a masked fashion. Results: The MRSS score in the Avastin-treated mice was significantly lower than that of the saline-treated mice (3.06 ± 1.63 versus 7.1 ± 2.01, respectively, p = 0.0021). The neovascularization count was also significantly lower in the study group (3.44 ± 1.81 versus 9.34 ± 3.23 for the controls, p = 0.0013). Conclusions: IP Avastin treatment reduced the extent of oxygen-induced retinopathy in a mouse model of retinopathy of prematurity.


Embo Molecular Medicine | 2017

PPARG is central to the initiation and propagation of human angiomyolipoma, suggesting its potential as a therapeutic target

Oren Pleniceanu; Racheli Shukrun; Dorit Omer; Einav Vax; Itamar Kanter; Klaudyna Dziedzic; Naomi Pode-Shakked; Michal Mark‐Daniei; Sara Pri-Chen; Yehudit Gnatek; Hadas Alfandary; Nira Varda-Bloom; Dekel D. Bar-Lev; Naomi Bollag; Rachel Shtainfeld; Leah Armon; Achia Urbach; Tomer Kalisky; Arnon Nagler; Orit Harari-Steinberg; Jack L. Arbiser; Benjamin Dekel

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.


Oncotarget | 2017

Pancreatic cancer ascites xenograft–an expeditious model mirroring advanced therapeutic resistant disease

Talia Golan; Chani Stossel; Michael Schvimer; Dikla Atias; Sharon Halperin; Ella Buzhor; Maria Raitses-Gurevich; Keren Cohen; Sara Pri-Chen; Julie L. Wilson; Robert E. Denroche; Ilinca Lungu; John M.S. Bartlett; Faridah Mbabaali; Yosef Yarden; Nishanth Belugali Nataraj; Steven Gallinger; Raanan Berger

Pancreatic ductal adenocarcinoma has limited treatment options. There is an urgent need for developing appropriate pre-clinical models recapitulating metastatic disease, the most common clinical scenario at presentation. Ascites accumulation occurs in up to 20–30% of patients with pancreatic cancer; this milieu represents a highly cellular research resource of metastatic peritoneal spread. In this study, we utilized pancreatic ascites/pleural effusion cancer cells to establish patient derived xenografts. Ascites/pleural effusion-patient derived xenografts were established from twelve independent cases. Xenografts were serially passed in nude mice and tissue bio-specimen banking has been established. Histopathology of emergent tumors demonstrates poorly to moderately differentiated, glandular and mucin producing tumors, mirroring morphology of primary pancreatic cancer tumors. Whole genome sequencing of six patient derived xenografts samples demonstrates common mutations and structural variations similar to those reported in primary pancreatic cancer. Xenograft tumors were dissociated to single-cells and in-vitro drug sensitivity screen assays demonstrated chemo-resistance, correlating with patient clinical scenarios, thus serving as a platform for clinically relevant translational research. Therefore, establishment of this novel ascites/pleural effusion patient derived xenograft model, with extensive histopathology and genomic characterization, opens an opportunity for the study of advanced aggressive pancreatic cancer. Characterization of metastatic disease and mechanisms of resistance to therapeutics may lead to the development of novel drug combinations.


Stem cell reports | 2018

In Vivo Expansion of Cancer Stemness Affords Novel Cancer Stem Cell Targets: Malignant Rhabdoid Tumor as an Example

Hana Golan; Rachel Shukrun; Revital Caspi; Einav Vax; Naomi Pode-Shakked; Sanja Goldberg; Oren Pleniceanu; Dekel D. Bar-Lev; Michal Mark-Danieli; Sara Pri-Chen; Jasmine Jacob-Hirsch; Itamar Kanter; Ariel Trink; Ginette Schiby; Ron Bilik; Tomer Kalisky; Orit Harari-Steinberg; Amos Toren; Benjamin Dekel

Summary Cancer stem cell (CSC) identification relies on transplantation assays of cell subpopulations sorted from fresh tumor samples. Here, we attempt to bypass limitations of abundant tumor source and predetermined immune selection by in vivo propagating patient-derived xenografts (PDX) from human malignant rhabdoid tumor (MRT), a rare and lethal pediatric neoplasm, to an advanced state in which most cells behave as CSCs. Stemness is then probed by comparative transcriptomics of serial PDXs generating a gene signature of epithelial to mesenchymal transition, invasion/motility, metastasis, and self-renewal, pinpointing putative MRT CSC markers. The relevance of these putative CSC molecules is analyzed by sorting tumorigenic fractions from early-passaged PDX according to one such molecule, deciphering expression in archived primary tumors, and testing the effects of CSC molecule inhibition on MRT growth. Using this platform, we identify ALDH1 and lysyl oxidase (LOX) as relevant targets and provide a larger framework for target and drug discovery in rare pediatric cancers.


Embo Molecular Medicine | 2017

Corrigendum to: Peroxisome proliferator-activated receptor gamma (PPARγ) is central to the initiation and propagation of human angiomyolipoma, suggesting its potential as a therapeutic target (EMBO Mol Med, (2017), 9, (508-530), 10.15252/emmm.201506111)

Oren Pleniceanu; Racheli Shukrun; Dorit Omer; Einav Vax; Itamar Kanter; Klaudyna Dziedzic; Naomi Pode-Shakked; Michal Mark‐Daniei; Sara Pri-Chen; Yehudit Gnatek; Hadas Alfandary; Nira Varda-Bloom; Dekel D. Bar-Lev; Naomi Bollag; Rachel Shtainfeld; Leah Armon; Achia Urbach; Tomer Kalisky; Arnon Nagler; Orit Harari-Steinberg; Jack L. Arbiser; Benjamin Dekel

The authors of the above research article have informed the journal that although the paper uses the official nomenclature PPARG to signify the peroxisome proliferator-activated receptor gamma gene, the most prevalent term used in the literature is PPARc. Therefore, the title, abstract, and keywords have been changed and now apply the latter term to improve searchability. The authors apologize for any inconvenience caused.


Embo Molecular Medicine | 2013

The isolation and characterization of renal cancer initiating cells from human Wilms' tumour xenografts unveils new therapeutic targets

Naomi Pode-Shakked; Rachel Shukrun; Michal Mark-Danieli; Peter Tsvetkov; Sarit Bahar; Sara Pri-Chen; Ronald S. Goldstein; Eithan Rom-Gross; Yoram Mor; Edward Fridman; Karen Meir; Amos J. Simon; Marcus Magister; Naftali Kaminski; Victor S. Goldmacher; Orit Harari-Steinberg; Benjamin Dekel

Collaboration


Dive into the Sara Pri-Chen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge