Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nature ; 607(7917): 163-168, 2022 07.
Article in English | MEDLINE | ID: mdl-35768509

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) shows pronounced epithelial and mesenchymal cancer cell populations1-4. Cellular heterogeneity in PDAC is an important feature in disease subtype specification3-5, but how distinct PDAC subpopulations interact, and the molecular mechanisms that underlie PDAC cell fate decisions, are incompletely understood. Here we identify the BMP inhibitor GREM16,7 as a key regulator of cellular heterogeneity in pancreatic cancer in human and mouse. Grem1 inactivation in established PDAC in mice resulted in a direct conversion of epithelial into mesenchymal PDAC cells within days, suggesting that persistent GREM1 activity is required to maintain the epithelial PDAC subpopulations. By contrast, Grem1 overexpression caused an almost complete 'epithelialization' of highly mesenchymal PDAC, indicating that high GREM1 activity is sufficient to revert the mesenchymal fate of PDAC cells. Mechanistically, Grem1 was highly expressed in mesenchymal PDAC cells and inhibited the expression of the epithelial-mesenchymal transition transcription factors Snai1 (also known as Snail) and Snai2 (also known as Slug) in the epithelial cell compartment, therefore restricting epithelial-mesenchymal plasticity. Thus, constant suppression of BMP activity is essential to maintain epithelial PDAC cells, indicating that the maintenance of the cellular heterogeneity of pancreatic cancer requires continuous paracrine signalling elicited by a single soluble factor.


Subject(s)
Epithelial-Mesenchymal Transition , Intercellular Signaling Peptides and Proteins , Pancreatic Neoplasms , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mesoderm/pathology , Mice , Pancreatic Neoplasms/pathology , Snail Family Transcription Factors
2.
Nat Commun ; 13(1): 2070, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440539

ABSTRACT

Deubiquitylating enzymes (DUBs) play an essential role in targeted protein degradation and represent an emerging therapeutic paradigm in cancer. However, their therapeutic potential in pancreatic ductal adenocarcinoma (PDAC) has not been explored. Here, we develop a DUB discovery pipeline, combining activity-based proteomics with a loss-of-function genetic screen in patient-derived PDAC organoids and murine genetic models. This approach identifies USP25 as a master regulator of PDAC growth and maintenance. Genetic and pharmacological USP25 inhibition results in potent growth impairment in PDAC organoids, while normal pancreatic organoids are insensitive, and causes dramatic regression of patient-derived xenografts. Mechanistically, USP25 deubiquitinates and stabilizes the HIF-1α transcription factor. PDAC is characterized by a severely hypoxic microenvironment, and USP25 depletion abrogates HIF-1α transcriptional activity and impairs glycolysis, inducing PDAC cell death in the tumor hypoxic core. Thus, the USP25/HIF-1α axis is an essential mechanism of metabolic reprogramming and survival in PDAC, which can be therapeutically exploited.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Glycolysis/genetics , Humans , Mice , Pancreatic Neoplasms/metabolism , Tumor Microenvironment/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Pancreatic Neoplasms
4.
J Mol Cell Biol ; 11(7): 553-563, 2019 07 19.
Article in English | MEDLINE | ID: mdl-30925590

ABSTRACT

Normal tissues are organized in a hierarchical model, whereas at the apex of these hierarchies reside stem cells (SCs) capable of self-renewal and of producing differentiated cellular progenies, leading to normal development and homeostasis. Alike, tumors are organized in a hierarchical manner, with cancer SCs residing at the apex, contributing to the development and nourishment of tumors. p53, the well-known 'guardian of the genome', possesses various roles in embryonic development as well as in adult SC life and serves as the 'guardian of tissue hierarchy'. Moreover, p53 serves as a barrier for dedifferentiation and reprogramming by constraining the cells to a somatic state and preventing their conversion to SCs. On the contrary, the mutant forms of p53 that lost their tumor suppressor activity and gain oncogenic functions serve as 'inducers of tissue anarchy' and promote cancer development. In this review, we discuss these two sides of the p53 token that sentence a tissue either to an ordered hierarchy and life or to anarchy and death. A better understanding of these processes may open new horizons for the development of new cancer therapies.


Subject(s)
Adult Stem Cells/metabolism , Cell Dedifferentiation/genetics , Cellular Reprogramming/genetics , Homeostasis , Mutation , Neoplasms , Tumor Suppressor Protein p53 , Adult Stem Cells/pathology , Animals , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
Cell Death Differ ; 26(9): 1566-1581, 2019 09.
Article in English | MEDLINE | ID: mdl-30413783

ABSTRACT

It is well accepted that malignant transformation is associated with unique metabolism. Malignant transformation involves a variety of cellular pathways that are associated with initiation and progression of the malignant process that remain to be deciphered still. Here we used a mouse model of mutant p53 that presents a stepwise progressive transformation of adult Mesenchymal Stem Cells (MSCs). While the established parental p53Mut-MSCs induce tumors, the parental p53WT-MSCs that were established in parallel, did not. Furthermore, tumor lines derived from the parental p53Mut-MSCs (p53Mut-MSC-TLs), exhibited yet a more aggressive transformed phenotype, suggesting exacerbation in tumorigenesis. Metabolic tracing of these various cell types, indicated that while malignant transformation is echoed by a direct augmentation in glycolysis, the more aggressive p53Mut-MSC-TLs demonstrate increased mitochondrial oxidation that correlates with morphological changes in mitochondria mass and function. Finally, we show that these changes are p53Mut-dependent. Computational transcriptional analysis identified a mitochondrial gene signature specifically downregulated upon knock/out of p53Mut in MSC-TLs. Our results suggest that stem cells exhibiting different state of malignancy are also associated with a different quantitative and qualitative metabolic profile in a p53Mut-dependent manner. This may provide important insights for cancer prognosis and the use of specific metabolic inhibitors in a personalized designed cancer therapy.


Subject(s)
Mesenchymal Stem Cells/metabolism , Mitochondria/metabolism , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Glycolysis/genetics , Humans , Mesenchymal Stem Cells/pathology , Metabolome/genetics , Mice , Mitochondria/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism
6.
Cancer Res ; 78(20): 5833-5847, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30154152

ABSTRACT

Mutations in the tumor suppressor p53 are the most frequent alterations in human cancer. These mutations include p53-inactivating mutations as well as oncogenic gain-of-function (GOF) mutations that endow p53 with capabilities to promote tumor progression. A primary challenge in cancer therapy is targeting stemness features and cancer stem cells (CSC) that account for tumor initiation, metastasis, and cancer relapse. Here we show that in vitro cultivation of tumors derived from mutant p53 murine bone marrow mesenchymal stem cells (MSC) gives rise to aggressive tumor lines (TL). These MSC-TLs exhibited CSC features as displayed by their augmented oncogenicity and high expression of CSC markers. Comparative analyses between MSC-TL with their parental mutant p53 MSC allowed for identification of the molecular events underlying their tumorigenic properties, including an embryonic stem cell (ESC) gene signature specifically expressed in MSC-TLs. Knockout of mutant p53 led to a reduction in tumor development and tumorigenic cell frequency, which was accompanied by reduced expression of CSC markers and the ESC MSC-TL signature. In human cancer, MSC-TL ESC signature-derived genes correlated with poor patient survival and were highly expressed in human tumors harboring p53 hotspot mutations. These data indicate that the ESC gene signature-derived genes may serve as new stemness-based prognostic biomarkers as well as novel cancer therapeutic targets.Significance: Mesenchymal cancer stem cell-like cell lines express a mutant p53-dependent embryonic stem cell gene signature, which can serve as a potential prognostic biomarker and therapeutic target in cancer. Cancer Res; 78(20); 5833-47. ©2018 AACR.


Subject(s)
Carcinogenesis/genetics , Embryonic Stem Cells/cytology , Gene Expression Regulation, Neoplastic , Tumor Suppressor Protein p53/genetics , Animals , Biomarkers, Tumor , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation , Humans , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Mutation , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Prognosis
7.
Oncogene ; 37(12): 1669-1684, 2018 03.
Article in English | MEDLINE | ID: mdl-29343849

ABSTRACT

Emerging notion in carcinogenesis ascribes tumor initiation and aggressiveness to cancer stem cells (CSCs). Specifically, colorectal cancer (CRC) development was shown to be compatible with CSCs hypothesis. Mutations in p53 are highly frequent in CRC, and are known to facilitate tumor development and aggressiveness. Yet, the link between mutant p53 and colorectal CSCs is not well-established. In the present study, we set to examine whether oncogenic mutant p53 proteins may augment colorectal CSCs phenotype. By genetic manipulation of mutant p53 in several cellular systems, we demonstrated that mutant p53 enhances colorectal tumorigenesis. Moreover, mutant p53-expressing cell lines harbor larger sub-populations of cells highly expressing the known colorectal CSCs markers: CD44, Lgr5, and ALDH. This elevated expression is mediated by mutant p53 binding to CD44, Lgr5, and ALDH1A1 promoter sequences. Furthermore, ALDH1 was found to be involved in mutant p53-dependent chemotherapy resistance. Finally, analysis of ALDH1 and CD44 in human CRC biopsies indicated a positive correlation between their expression and the presence of oncogenic p53 missense mutations. These findings suggest novel insights pertaining the mechanism by which mutant p53 enhances CRC development, which involves the expansion of CSCs sub-populations within CRC tumors, and underscore the importance of targeting these sub-populations for CRC therapy.


Subject(s)
Colorectal Neoplasms/genetics , Gain of Function Mutation , Neoplastic Stem Cells/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Biomarkers, Tumor/genetics , Colorectal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Mice, Transgenic , Mutant Proteins/physiology , Mutation, Missense , Tumor Cells, Cultured
8.
Sci Rep ; 7(1): 788, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28400563

ABSTRACT

The Golgi apparatus is a dynamic organelle, which regulates the vesicular trafficking. While cellular trafficking requires active changes of the Golgi membranes, these are not accompanied by changes in the general Golgi's structure. However, cellular processes such as mitosis, apoptosis and migration require fragmentation of the Golgi complex. Currently, these changes are most commonly studied by basic immunofluorescence and quantified by manual and subjective classification of the Golgi structure in 100-500 stained cells. Several other high-throughput methods exist as well, but those are either complicated or do not provide enough morphological information. Therefore, a simple and informative high content methodology should be beneficial for the study of Golgi architecture. Here we describe the use of high-throughput imaging flow cytometry for quantification of Golgi fragmentation, which provides a simple way to analyze the changes in an automated, quantitative and non-biased manner. Furthermore, it provides a rapid and accurate way to analyze more than 50,000 cells per sample. Our results demonstrate that this method is robust and statistically powerful, thus, providing a much-needed analytical tool for future studies on Golgi dynamics, and can be adapted to other experimental systems.


Subject(s)
Flow Cytometry , Golgi Apparatus/metabolism , High-Throughput Screening Assays , Animals , Biomarkers , COS Cells , Chlorocebus aethiops , Golgi Apparatus/drug effects , HeLa Cells , Humans , Mice , Mitosis , Neoplasms/metabolism , Neoplasms/pathology
9.
Semin Cancer Biol ; 32: 10-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24406212

ABSTRACT

During recent years, it is becoming more and more evident that there is a tight connection between abnormal differentiation processes and cancer. While cancer and stem cells are very different, especially in terms of maintaining genomic integrity, these cell types also share many similar properties. In this review, we aim to provide an over-view of the roles of the key tumor suppressor, p53, in regulating normal differentiation and function of both stem cells and adult cells. When these functions are disrupted, undifferentiated cells may become transformed. Understanding the function of p53 in stem cells and its role in maintaining the balance between differentiation and malignant transformation can help shed light on cancer initiation and propagation, and hopefully also on cancer prevention and therapy.


Subject(s)
Cell Differentiation/genetics , Neoplasms/genetics , Neoplastic Stem Cells , Tumor Suppressor Protein p53/genetics , Animals , Humans , Mice , Tumor Suppressor Protein p53/physiology
10.
Proc Natl Acad Sci U S A ; 111(19): 7006-11, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24778235

ABSTRACT

p53 is a well-known tumor suppressor that is mutated in over 50% of human cancers. These mutations were shown to exhibit gain of oncogenic function compared with the deletion of the gene. Additionally, p53 has fundamental roles in differentiation and development; nevertheless, mutant p53 mice are viable and develop malignant tumors only on adulthood. We set out to reveal the mechanisms by which embryos are protected from mutant p53-induced transformation using ES cells (ESCs) that express a conformational mutant of p53. We found that, despite harboring mutant p53, the ESCs remain pluripotent and benign and have relatively normal karyotype compared with ESCs knocked out for p53. Additionally, using high-content RNA sequencing, we show that p53 is transcriptionally active in response to DNA damage in mutant ESCs and elevates p53 target genes, such as p21 and btg2. We also show that the conformation of mutant p53 protein in ESCs is stabilized to a WT conformation. Through MS-based interactome analyses, we identified a network of proteins, including the CCT complex, USP7, Aurora kinase, Nedd4, and Trim24, that bind mutant p53 and may shift its conformation to a WT form. We propose this conformational shift as a novel mechanism of maintenance of genomic integrity, despite p53 mutation. Harnessing the ability of these protein interactors to transform the oncogenic mutant p53 to the tumor suppressor WT form can be the basis for future development of p53-targeted cancer therapy.


Subject(s)
Cell Transformation, Neoplastic/genetics , Embryonic Stem Cells/cytology , Li-Fraumeni Syndrome/genetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Adenocarcinoma , Animals , Breast Neoplasms , Cell Line , Cell Line, Tumor , Cell Proliferation , Embryonic Development/genetics , Embryonic Stem Cells/physiology , Homeodomain Proteins/genetics , Humans , Li-Fraumeni Syndrome/metabolism , Loss of Heterozygosity/physiology , Mice , Mice, Knockout , Nanog Homeobox Protein , Protein Conformation , Proteomics , Tumor Suppressor Protein p53/metabolism
11.
Carcinogenesis ; 35(6): 1196-208, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24658181

ABSTRACT

It is well accepted that expression of mutant p53 involves the gain of oncogenic-specific activities accentuating the malignant phenotype. Depending on the specific cancer type, mutant p53 can contribute to either the early or the late events of the multiphase process underlying the transformation of a normal cell into a cancerous one. This multifactorial system is evident in ~50% of human cancers. Mutant p53 was shown to interfere with a variety of cellular functions that lead to augmented cell survival, cellular plasticity, aberration of DNA repair machinery and other effects. All these effects culminate in the acquisition of drug resistance often seen in cancer cells. Interestingly, drug resistance has also been suggested to be associated with cancer stem cells (CSCs), which reside within growing tumors. The notion that p53 plays a regulatory role in the life of stem cells, coupled with the observations that p53 mutations may contribute to the evolvement of CSCs makes it challenging to speculate that drug resistance and cancer recurrence are mediated by CSCs expressing mutant p53.


Subject(s)
Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Mutation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Genomic Instability , Humans , Signal Transduction , Tumor Suppressor Protein p53/metabolism
12.
Exp Cell Res ; 318(7): 789-99, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22305966

ABSTRACT

The PI3K-AKT pathway is frequently activated in human cancers, including breast cancer, and its activation appears to be critical for tumor maintenance. Some malignant cells are dependent on activated AKT for their survival; tumors exhibiting elevated AKT activity show sensitivity to its inhibition, providing an Achilles heel for their treatment. Here we show that the PKCη isoform is a negative regulator of the AKT signaling pathway. The IGF-I induced phosphorylation on Ser473 of AKT was inhibited by the PKCη-induced expression in MCF-7 breast adenocarcinoma cancer cells. This was further confirmed in shRNA PKCη-knocked-down MCF-7 cells, demonstrating elevated phosphorylation on AKT Ser473. While PKCη exhibited negative regulation on AKT phosphorylation it did not alter the IGF-I induced ERK phosphorylation. However, it enhanced ERK phosphorylation when stimulated by PDGF. Moreover, its effects on IGF-I/AKT and PDGF/ERK pathways were in correlation with cell proliferation. We further show that both PKCη and IGF-I confer protection against UV-induced apoptosis and cell death having additive effects. Although the protective effect of IGF-I involved activation of AKT, it was not affected by PKCη expression, suggesting that PKCη acts through a different route to increase cell survival. Hence, our studies show that PKCη provides negative control on AKT pathway leading to reduced cell proliferation, and further suggest that its presence/absence in breast cancer cells will affect cell death, which could be of therapeutic value.


Subject(s)
Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Insulin-Like Growth Factor I/metabolism , Oncogene Protein v-akt/metabolism , Protein Kinase C/metabolism , Adenocarcinoma/drug therapy , Breast Neoplasms/drug therapy , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Knockdown Techniques , Humans , Isoenzymes/metabolism , MAP Kinase Signaling System/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Protein Kinase C/genetics , RNA, Small Interfering/metabolism , Serine/metabolism , Signal Transduction/drug effects , Ultraviolet Rays
13.
Int Immunol ; 23(7): 453-61, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21652516

ABSTRACT

Priming of naive CD8 T cells by dendritic cells (DCs) entails both effective antigen presentation on MHC class I products and co-stimulatory signaling. Their optimal coupling is a major goal in the development of CTL-inducing vaccines. We recently reported that a membranal derivative of the invariant MHC-I light chain, ß(2)-microglobulin (ß(2)m), markedly stabilizes MHC-I molecules and can serve as a universal platform for exceptional presentation of genetically linked peptides. To test whether it is possible to equip the resulting MHC-I complexes with an inherent ability to activate antigen-presenting cells, we engrafted the intracellular Toll/IL-1 receptor domain of mouse Toll-like receptor (TLR) 4 or TLR2 onto the peptide-ß(2)m scaffold. We evaluated the level of peptide presentation and status of cell activation conferred by such constructs in stably transfected mouse RAW264.7 macrophages and mRNA-transfected mouse DC2.4 DCs. We show that the encoded peptide-ß(2)m-TLR polypeptides are expressed at the cell surface, pair with endogenous heavy chains, stabilize MHC-I products, prompt efficient peptide-specific T-cell recognition and confer a constitutively activated phenotype on the transfected cells, as judged by the up-regulation of pro-inflammatory genes and surface co-stimulatory molecules. Our results provide evidence that the product of a single recombinant gene can couple MHC peptide presentation to TLR-mediated signaling and offer a safe, economical and highly versatile modality for a novel category of genetic CTL-inducing vaccines.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , H-2 Antigens/immunology , Peptides/immunology , Animals , Genetic Vectors/genetics , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptides/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Transfection , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...