Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 10(1): 3474, 2020 02 26.
Article in English | MEDLINE | ID: mdl-32103065

ABSTRACT

Reliable approaches to identify stem cell mechanisms that mediate aggressive cancer could have great therapeutic value, based on the growing evidence of embryonic signatures in metastatic cancers. However, how to best identify and target stem-like mechanisms aberrantly acquired by cancer cells has been challenging. We harnessed the power of reprogramming to examine GRP78, a chaperone protein generally restricted to the endoplasmic reticulum in normal tissues, but which is expressed on the cell surface of human embryonic stem cells and many cancer types. We have discovered that (1) cell surface GRP78 (sGRP78) is expressed on iPSCs and is important in reprogramming, (2) sGRP78 promotes cellular functions in both pluripotent and breast cancer cells (3) overexpression of GRP78 in breast cancer cells leads to an induction of a CD24-/CD44+ tumor initiating cell (TIC) population (4) sGRP78+ breast cancer cells are enriched for stemness genes and appear to be a subset of TICs (5) sGRP78+ breast cancer cells show an enhanced ability to seed metastatic organ sites in vivo. These collective findings show that GRP78 has important functions in regulating both pluripotency and oncogenesis, and suggest that sGRP78 marks a stem-like population in breast cancer cells that has increased metastatic potential in vivo.


Subject(s)
Cell Differentiation , Cell Self Renewal , Heat-Shock Proteins/metabolism , Neoplastic Stem Cells/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Cellular Reprogramming , Endoplasmic Reticulum Chaperone BiP , Female , HEK293 Cells , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Mice, Knockout , Neoplastic Stem Cells/cytology , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation, Heterologous
3.
Cell Stem Cell ; 20(4): 505-517.e6, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28388429

ABSTRACT

Induced pluripotent stem cells (iPSCs) show variable methylation patterns between lines, some of which reflect aberrant differences relative to embryonic stem cells (ESCs). To examine whether this aberrant methylation results from genetic variation or non-genetic mechanisms, we generated human iPSCs from monozygotic twins to investigate how genetic background, clone, and passage number contribute. We found that aberrantly methylated CpGs are enriched in regulatory regions associated with MYC protein motifs and affect gene expression. We classified differentially methylated CpGs as being associated with genetic and/or non-genetic factors (clone and passage), and we found that aberrant methylation preferentially occurs at CpGs associated with clone-specific effects. We further found that clone-specific effects play a strong role in recurrent aberrant methylation at specific CpG sites across different studies. Our results argue that a non-genetic biological mechanism underlies aberrant methylation in iPSCs and that it is likely based on a probabilistic process involving MYC that takes place during or shortly after reprogramming.


Subject(s)
DNA Methylation/genetics , Induced Pluripotent Stem Cells/metabolism , Nucleotide Motifs/genetics , Proto-Oncogene Proteins c-myc/metabolism , Clone Cells , CpG Islands/genetics , Fibroblasts/metabolism , Gene Expression Regulation , Genetic Variation , Genome-Wide Association Study , Humans , Sequence Analysis, RNA , Transcription Factors/metabolism , Twins, Monozygotic/genetics
4.
Stem Cell Reports ; 8(4): 1086-1100, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28410642

ABSTRACT

Large-scale collections of induced pluripotent stem cells (iPSCs) could serve as powerful model systems for examining how genetic variation affects biology and disease. Here we describe the iPSCORE resource: a collection of systematically derived and characterized iPSC lines from 222 ethnically diverse individuals that allows for both familial and association-based genetic studies. iPSCORE lines are pluripotent with high genomic integrity (no or low numbers of somatic copy-number variants) as determined using high-throughput RNA-sequencing and genotyping arrays, respectively. Using iPSCs from a family of individuals, we show that iPSC-derived cardiomyocytes demonstrate gene expression patterns that cluster by genetic background, and can be used to examine variants associated with physiological and disease phenotypes. The iPSCORE collection contains representative individuals for risk and non-risk alleles for 95% of SNPs associated with human phenotypes through genome-wide association studies. Our study demonstrates the utility of iPSCORE for examining how genetic variants influence molecular and physiological traits in iPSCs and derived cell lines.


Subject(s)
Arrhythmias, Cardiac/genetics , Databases, Factual , Genetic Association Studies , Genetic Variation , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Arrhythmias, Cardiac/ethnology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cell Differentiation , Cell Line , Cellular Reprogramming/genetics , Genotype , High-Throughput Nucleotide Sequencing , Humans , Induced Pluripotent Stem Cells/cytology , Multigene Family , Myocytes, Cardiac/cytology , Oligonucleotide Array Sequence Analysis , Phenotype , Polymorphism, Single Nucleotide , Racial Groups
5.
Nat Commun ; 7: 10743, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-26899176

ABSTRACT

Glioma tumour-initiating cells (GTICs) can originate upon the transformation of neural progenitor cells (NPCs). Studies on GTICs have focused on primary tumours from which GTICs could be isolated and the use of human embryonic material. Recently, the somatic genomic landscape of human gliomas has been reported. RTK (receptor tyrosine kinase) and p53 signalling were found dysregulated in ∼90% and 86% of all primary tumours analysed, respectively. Here we report on the use of human-induced pluripotent stem cells (hiPSCs) for modelling gliomagenesis. Dysregulation of RTK and p53 signalling in hiPSC-derived NPCs (iNPCs) recapitulates GTIC properties in vitro. In vivo transplantation of transformed iNPCs leads to highly aggressive tumours containing undifferentiated stem cells and their differentiated derivatives. Metabolic modulation compromises GTIC viability. Last, screening of 101 anti-cancer compounds identifies three molecules specifically targeting transformed iNPCs and primary GTICs. Together, our results highlight the potential of hiPSCs for studying human tumourigenesis.


Subject(s)
Cell Transformation, Neoplastic , Glioma/etiology , Induced Pluripotent Stem Cells , Neoplastic Stem Cells/physiology , Neural Stem Cells/physiology , Cell Line, Tumor , Female , Humans , Male , SOXB1 Transcription Factors/metabolism , Tumor Stem Cell Assay
6.
Cell Stem Cell ; 13(3): 341-50, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23871606

ABSTRACT

Since the initial discovery that OCT4, SOX2, KLF4, and c-MYC overexpression sufficed for the induction of pluripotency in somatic cells, methodologies replacing the original factors have enhanced our understanding of the reprogramming process. However, unlike in mouse, OCT4 has not been replaced successfully during reprogramming of human cells. Here we report on a strategy to accomplish this replacement. Through a combination of transcriptome and bioinformatic analysis we have identified factors previously characterized as being lineage specifiers that are able to replace OCT4 and SOX2 in the reprogramming of human fibroblasts. Our results show that it is possible to replace OCT4 and SOX2 simultaneously with alternative lineage specifiers in the reprogramming of human cells. At a broader level, they also support a model in which counteracting lineage specification networks underlies the induction of pluripotency.


Subject(s)
Cell Dedifferentiation , Fibroblasts/physiology , GATA3 Transcription Factor/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/physiology , Cell Dedifferentiation/genetics , Cell Lineage , Cells, Cultured , Computational Biology/methods , GATA3 Transcription Factor/genetics , Gene Expression Profiling , Gene Expression Regulation/genetics , Guided Tissue Regeneration , Humans , Kruppel-Like Factor 4 , Octamer Transcription Factor-3/genetics , RNA, Small Interfering/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transgenes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...