Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Proc Natl Acad Sci U S A ; 115(47): E11128-E11137, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30385632

ABSTRACT

Osteosarcoma (OS), the most common primary bone tumor, is highly metastatic with high chemotherapeutic resistance and poor survival rates. Using induced pluripotent stem cells (iPSCs) generated from Li-Fraumeni syndrome (LFS) patients, we investigate an oncogenic role of secreted frizzled-related protein 2 (SFRP2) in p53 mutation-associated OS development. Interestingly, we find that high SFRP2 expression in OS patient samples correlates with poor survival. Systems-level analyses identified that expression of SFRP2 increases during LFS OS development and can induce angiogenesis. Ectopic SFRP2 overexpression in normal osteoblast precursors is sufficient to suppress normal osteoblast differentiation and to promote OS phenotypes through induction of oncogenic molecules such as FOXM1 and CYR61 in a ß-catenin-independent manner. Conversely, inhibition of SFRP2, FOXM1, or CYR61 represses the tumorigenic potential. In summary, these findings demonstrate the oncogenic role of SFRP2 in the development of p53 mutation-associated OS and that inhibition of SFRP2 is a potential therapeutic strategy.


Subject(s)
Bone Neoplasms/genetics , Carcinogenesis/genetics , Li-Fraumeni Syndrome/pathology , Membrane Proteins/genetics , Osteosarcoma/genetics , Tumor Suppressor Protein p53/genetics , Animals , Bone Neoplasms/pathology , Cell Line, Tumor , Cysteine-Rich Protein 61/antagonists & inhibitors , Cysteine-Rich Protein 61/genetics , Forkhead Box Protein M1/antagonists & inhibitors , Forkhead Box Protein M1/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Li-Fraumeni Syndrome/genetics , Male , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Osteoblasts/cytology , Osteosarcoma/pathology
2.
Microb Ecol ; 79(1): 1-11, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31111178

ABSTRACT

Temperature increase may influence competition among phytoplankton species, potentially intensifying cyanobacteria blooms that can be favored by direct and indirect effects of temperature. In this study, we aimed to clarify how cyanobacteria can be favored by the direct effects of increased temperature compared to diatoms and chlorophytes. Strains of the most representative species of a eutrophic coastal lagoon (Microcystis aeruginosa, Planktothrix agardhii, Desmodesmus communis, and Cyclotella meneghiniana) were used to test the hypothesis that cyanobacteria would be favored by the direct effect of temperature increase. First, we evaluated the effect of temperature increase on growth in monocultures (batch and chemostats) at 25 and 30 °C and after in mixed cultures (chemostats). In batch monocultures, the cyanobacteria showed higher growth rates in 30 °C than in 25 °C. However, in continuous culture experiments (chemostats), growth rates of M. aeruginosa and P. agardhii were not affected by temperature, but the strains showed higher biovolume in steady-state with the temperature increase. In continuous mixed cultures, M. aeruginosa was always dominant and C. meneghiniana was excluded, regardless of temperature tested. D. communis was able to coexist with lower biomass. This study shows that rising temperatures can be detrimental to diatoms, even for a tropical strain. Although some studies indicate that the dominance of cyanobacteria in warmer climates may be due to the indirect effect of warming that will promote physical conditions in the environment more favorable to cyanobacteria, the outcomes of mixed cultures demonstrate that the direct effect of temperature can also favor the dominance of cyanobacteria.


Subject(s)
Chlorophyta/growth & development , Diatoms/growth & development , Microcystis/growth & development , Phytoplankton/growth & development , Biomass , Chlorophyta/radiation effects , Climate , Diatoms/radiation effects , Light , Microcystis/radiation effects , Phytoplankton/radiation effects , Temperature
3.
J Vis Exp ; (153)2019 11 04.
Article in English | MEDLINE | ID: mdl-31736500

ABSTRACT

The cellular and molecular mechanisms underlying specification of human hematopoietic stem cells (HSCs) remain elusive. Strategies to recapitulate human HSC emergence in vitro are required to overcome limitations in studying this complex developmental process. Here, we describe a protocol to generate hematopoietic stem and progenitor-like cells from human dermal fibroblasts employing a direct cell reprogramming approach. These cells transit through a hemogenic intermediate cell-type, resembling the endothelial-to-hematopoietic transition (EHT) characteristic of HSC specification. Fibroblasts were reprogrammed to hemogenic cells via transduction with GATA2, GFI1B and FOS transcription factors. This combination of three factors induced morphological changes, expression of hemogenic and hematopoietic markers and dynamic EHT transcriptional programs. Reprogrammed cells generate hematopoietic progeny and repopulate immunodeficient mice for three months. This protocol can be adapted towards the mechanistic dissection of the human EHT process as exemplified here by defining GATA2 targets during the early phases of reprogramming. Thus, human hemogenic reprogramming provides a simple and tractable approach to identify novel markers and regulators of human HSC emergence. In the future, faithful induction of hemogenic fate in fibroblasts may lead to the generation of patient-specific HSCs for transplantation.


Subject(s)
Cellular Reprogramming , Fibroblasts/physiology , Hematopoietic Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Adhesion , Cell Differentiation , Humans , Mice , Transcription Factors/genetics
4.
FEBS Lett ; 593(23): 3266-3287, 2019 12.
Article in English | MEDLINE | ID: mdl-31557312

ABSTRACT

Transcription factor (TF)-based reprogramming of somatic tissues holds great promise for regenerative medicine. Previously, we demonstrated that the TFs GATA2, GFI1B, and FOS convert mouse and human fibroblasts to hemogenic endothelial-like precursors that generate hematopoietic stem progenitor (HSPC)-like cells over time. This conversion is lacking in robustness both in yield and biological function. Herein, we show that inclusion of GFI1 to the reprogramming cocktail significantly expands the HSPC-like population. AFT024 coculture imparts functional potential to these cells and allows quantification of stem cell frequency. Altogether, we demonstrate an improved human hemogenic induction protocol that could provide a valuable human in vitro model of hematopoiesis for disease modeling and a platform for cell-based therapeutics. DATABASE: Gene expression data are available in the Gene Expression Omnibus (GEO) database under the accession number GSE130361.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Hemangioblasts/cytology , Hematopoietic Stem Cells/cytology , Animals , Coculture Techniques/methods , Fibroblasts/cytology , Fibroblasts/metabolism , GATA2 Transcription Factor/genetics , Gene Expression Regulation, Developmental/genetics , Hemangioblasts/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-fos/genetics , Repressor Proteins/genetics , Transcription Factors/genetics
5.
Cell Rep ; 25(10): 2821-2835.e7, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30517869

ABSTRACT

During development, hematopoietic stem and progenitor cells (HSPCs) arise from specialized endothelial cells by a process termed endothelial-to-hematopoietic transition (EHT). The genetic program driving human HSPC emergence remains largely unknown. We previously reported that the generation of hemogenic precursor cells from mouse fibroblasts recapitulates developmental hematopoiesis. Here, we demonstrate that human fibroblasts can be reprogrammed into hemogenic cells by the same transcription factors. Induced cells display dynamic EHT transcriptional programs, generate hematopoietic progeny, possess HSPC cell surface phenotype, and repopulate immunodeficient mice for 3 months. Mechanistically, GATA2 and GFI1B interact and co-occupy a cohort of targets. This cooperative binding is reflected by engagement of open enhancers and promoters, initiating silencing of fibroblast genes and activating the hemogenic program. However, GATA2 displays dominant and independent targeting activity during the early phases of reprogramming. These findings shed light on the processes controlling human HSC specification and support generation of reprogrammed HSCs for clinical applications.


Subject(s)
Cellular Reprogramming , Hemangioblasts/cytology , Hemangioblasts/metabolism , Transcription Factors/metabolism , Adult , Base Sequence , Enhancer Elements, Genetic/genetics , Fibroblasts/metabolism , GATA2 Transcription Factor/metabolism , Gene Expression Regulation , HEK293 Cells , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Infant, Newborn , Phenotype , Promoter Regions, Genetic/genetics , Protein Binding
6.
Sci Immunol ; 3(30)2018 12 07.
Article in English | MEDLINE | ID: mdl-30530727

ABSTRACT

Ectopic expression of transcription factors has been used to reprogram differentiated somatic cells toward pluripotency or to directly reprogram them to other somatic cell lineages. This concept has been explored in the context of regenerative medicine. Here, we set out to generate dendritic cells (DCs) capable of presenting antigens from mouse and human fibroblasts. By screening combinations of 18 transcription factors that are expressed in DCs, we have identified PU.1, IRF8, and BATF3 transcription factors as being sufficient to reprogram both mouse and human fibroblasts to induced DCs (iDCs). iDCs acquire a conventional DC type 1-like transcriptional program, with features of interferon-induced maturation. iDCs secrete inflammatory cytokines and have the ability to engulf, process, and present antigens to T cells. Furthermore, we demonstrate that murine iDCs generated here were able to cross-present antigens to CD8+ T cells. Our reprogramming system should facilitate better understanding of DC specification programs and serve as a platform for the development of patient-specific DCs for immunotherapy.


Subject(s)
Antigen-Presenting Cells/immunology , Fibroblasts/immunology , Animals , Cells, Cultured , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL