Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 99, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397934

ABSTRACT

CD4 and CD8 mark helper and cytotoxic T cell lineages, respectively, and serve as coreceptors for MHC-restricted TCR recognition. How coreceptor expression is matched with TCR specificity is central to understanding CD4/CD8 lineage choice, but visualising coreceptor gene activity in individual selection intermediates has been technically challenging. It therefore remains unclear whether the sequence of coreceptor gene expression in selection intermediates follows a stereotypic pattern, or is responsive to signaling. Here we use single cell RNA sequencing (scRNA-seq) to classify mouse thymocyte selection intermediates by coreceptor gene expression. In the unperturbed thymus, Cd4+Cd8a- selection intermediates appear before Cd4-Cd8a+ selection intermediates, but the timing of these subsets is flexible according to the strength of TCR signals. Our data show that selection intermediates discriminate MHC class prior to the loss of coreceptor expression and suggest a model where signal strength informs the timing of coreceptor gene activity and ultimately CD4/CD8 lineage choice.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Lineage/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Core Binding Factor Alpha 3 Subunit/metabolism , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Histocompatibility Antigens/metabolism , Lymphocyte Activation/genetics , Mice, Inbred C57BL , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Transcription Factors/metabolism
2.
Genome Biol ; 21(1): 112, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32393363

ABSTRACT

Robust protocols and automation now enable large-scale single-cell RNA and ATAC sequencing experiments and their application on biobank and clinical cohorts. However, technical biases introduced during sample acquisition can hinder solid, reproducible results, and a systematic benchmarking is required before entering large-scale data production. Here, we report the existence and extent of gene expression and chromatin accessibility artifacts introduced during sampling and identify experimental and computational solutions for their prevention.


Subject(s)
Artifacts , Genomics , Single-Cell Analysis , Cryopreservation , Epigenome , Female , Humans , Leukocytes, Mononuclear , Male , Time Factors , Transcriptome
3.
Elife ; 82019 03 12.
Article in English | MEDLINE | ID: mdl-30860479

ABSTRACT

Forced transcription factor expression can transdifferentiate somatic cells into other specialised cell types or reprogram them into induced pluripotent stem cells (iPSCs) with variable efficiency. To better understand the heterogeneity of these processes, we used single-cell RNA sequencing to follow the transdifferentation of murine pre-B cells into macrophages as well as their reprogramming into iPSCs. Even in these highly efficient systems, there was substantial variation in the speed and path of fate conversion. We predicted and validated that these differences are inversely coupled and arise in the starting cell population, with Mychigh large pre-BII cells transdifferentiating slowly but reprogramming efficiently and Myclow small pre-BII cells transdifferentiating rapidly but failing to reprogram. Strikingly, differences in Myc activity predict the efficiency of reprogramming across a wide range of somatic cell types. These results illustrate how single cell expression and computational analyses can identify the origins of heterogeneity in cell fate conversion processes.


Subject(s)
Cell Lineage , Cell Transdifferentiation , Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Precursor Cells, B-Lymphoid/cytology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA-Seq , Signal Transduction , Single-Cell Analysis , Transcriptome
4.
Int J Mol Sci ; 20(3)2019 Feb 08.
Article in English | MEDLINE | ID: mdl-30743987

ABSTRACT

The possibility of humans to live outside of Earth on another planet has attracted the attention of numerous scientists around the world. One of the greatest difficulties is that humans cannot live in an extra-Earth environment without proper equipment. In addition, the consequences of chronic gravity alterations in human body are not known. Here, we used planarians as a model system to test how gravity fluctuations could affect complex organisms. Planarians are an ideal system, since they can regenerate any missing part and they are continuously renewing their tissues. We performed a transcriptomic analysis of animals submitted to simulated microgravity (Random Positioning Machine, RPM) (s-µg) and hypergravity (8 g), and we observed that the transcriptional levels of several genes are affected. Surprisingly, we found the major differences in the s-µg group. The results obtained in the transcriptomic analysis were validated, demonstrating that our transcriptomic data is reliable. We also found that, in a sensitive environment, as under Hippo signaling silencing, gravity fluctuations potentiate the increase in cell proliferation. Our data revealed that changes in gravity severely affect genetic transcription and that these alterations potentiate molecular disorders that could promote the development of multiple diseases such as cancer.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Planarians/physiology , Transcriptome , Weightlessness , Animals , Cell Proliferation , Computational Biology/methods , Gene Expression Regulation , Gene Silencing , In Situ Hybridization , RNA Interference , Reproducibility of Results , Weightlessness Simulation
5.
Genome Res ; 28(6): 878-890, 2018 06.
Article in English | MEDLINE | ID: mdl-29724792

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) has significantly deepened our insights into complex tissues, with the latest techniques capable of processing tens of thousands of cells simultaneously. Analyzing increasing numbers of cells, however, generates extremely large data sets, extending processing time and challenging computing resources. Current scRNA-seq analysis tools are not designed to interrogate large data sets and often lack sensitivity to identify marker genes. With bigSCale, we provide a scalable analytical framework to analyze millions of cells, which addresses the challenges associated with large data sets. To handle the noise and sparsity of scRNA-seq data, bigSCale uses large sample sizes to estimate an accurate numerical model of noise. The framework further includes modules for differential expression analysis, cell clustering, and marker identification. A directed convolution strategy allows processing of extremely large data sets, while preserving transcript information from individual cells. We evaluated the performance of bigSCale using both a biological model of aberrant gene expression in patient-derived neuronal progenitor cells and simulated data sets, which underlines the speed and accuracy in differential expression analysis. To test its applicability for large data sets, we applied bigSCale to assess 1.3 million cells from the mouse developing forebrain. Its directed down-sampling strategy accumulates information from single cells into index cell transcriptomes, thereby defining cellular clusters with improved resolution. Accordingly, index cell clusters identified rare populations, such as reelin (Reln)-positive Cajal-Retzius neurons, for which we report previously unrecognized heterogeneity associated with distinct differentiation stages, spatial organization, and cellular function. Together, bigSCale presents a solution to address future challenges of large single-cell data sets.


Subject(s)
RNA/genetics , Single-Cell Analysis/methods , Software , Transcriptome/genetics , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Differentiation/genetics , Cluster Analysis , Extracellular Matrix Proteins/genetics , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Mice , Nerve Tissue Proteins/genetics , Neurons/metabolism , Reelin Protein , Serine Endopeptidases/genetics
6.
PLoS Biol ; 16(1): e2002399, 2018 01.
Article in English | MEDLINE | ID: mdl-29357350

ABSTRACT

The Hippo pathway plays a key role in regulating cell turnover in adult tissues, and abnormalities in this pathway are consistently associated with human cancers. Hippo was initially implicated in the control of cell proliferation and death, and its inhibition is linked to the expansion of stem cells and progenitors, leading to larger organ size and tumor formation. To understand the mechanism by which Hippo directs cell renewal and promotes stemness, we studied its function in planarians. These stem cell-based organisms are ideal models for the analysis of the complex cellular events underlying tissue renewal in the whole organism. hippo RNA interference (RNAi) in planarians decreased apoptotic cell death, induced cell cycle arrest, and could promote the dedifferentiation of postmitotic cells. hippo RNAi resulted in extensive undifferentiated areas and overgrowths, with no effect on body size or cell number. We propose an essential role for hippo in controlling cell cycle, restricting cell plasticity, and thereby preventing tumoral transformation.


Subject(s)
Cell Plasticity/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Apoptosis , Cell Cycle/physiology , Cell Differentiation , Cell Division , Cell Proliferation , Hippo Signaling Pathway , Planarians/metabolism , Planarians/physiology , RNA Interference , Signal Transduction , Stem Cells/physiology
7.
Genome Biol ; 18(1): 45, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28249587

ABSTRACT

A variety of single-cell RNA preparation procedures have been described. So far, protocols require fresh material, which hinders complex study designs. We describe a sample preservation method that maintains transcripts in viable single cells, allowing one to disconnect time and place of sampling from subsequent processing steps. We sequence single-cell transcriptomes from >1000 fresh and cryopreserved cells using 3'-end and full-length RNA preparation methods. Our results confirm that the conservation process did not alter transcriptional profiles. This substantially broadens the scope of applications in single-cell transcriptomics and could lead to a paradigm shift in future study designs.


Subject(s)
Cryopreservation , Gene Expression Profiling , Single-Cell Analysis , Transcriptome , Animals , Cell Line , Cluster Analysis , Disease Models, Animal , Gene Expression Profiling/methods , Heterografts , Humans , Mice , Neoplasms/genetics , Neoplasms/pathology , RNA/genetics , RNA Stability , Reproducibility of Results , Sequence Analysis, DNA , Single-Cell Analysis/methods
8.
Cell Stem Cell ; 20(6): 801-816.e7, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28285904

ABSTRACT

Highly proliferative Lgr5+ stem cells maintain the intestinal epithelium and are thought to be largely homogeneous. Although quiescent intestinal stem cell (ISC) populations have been described, the identity and features of such a population remain controversial. Here we report unanticipated heterogeneity within the Lgr5+ ISC pool. We found that expression of the RNA-binding protein Mex3a labels a slowly cycling subpopulation of Lgr5+ ISCs that contribute to all intestinal lineages with distinct kinetics. Single-cell transcriptome profiling revealed that Lgr5+ cells adopt two discrete states, one of which is defined by a Mex3a expression program and relatively low levels of proliferation genes. During homeostasis, Mex3a+ cells continually shift into the rapidly dividing, self-renewing ISC pool. Chemotherapy and radiation preferentially target rapidly dividing Lgr5+ cells but spare the Mex3a-high/Lgr5+ population, helping to promote regeneration of the intestinal epithelium following toxic insults. Thus, Mex3a defines a reserve-like ISC population within the Lgr5+ compartment.


Subject(s)
Cell Proliferation/physiology , Intestinal Mucosa/metabolism , RNA-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Animals , Intestinal Mucosa/cytology , Mice , Mice, Transgenic , RNA-Binding Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Stem Cells/cytology
9.
BMC Genomics ; 16: 361, 2015 May 08.
Article in English | MEDLINE | ID: mdl-25952370

ABSTRACT

BACKGROUND: The freshwater planarian Schmidtea mediterranea is recognised as a valuable model for research into adult stem cells and regeneration. With the advent of the high-throughput sequencing technologies, it has become feasible to undertake detailed transcriptional analysis of its unique stem cell population, the neoblasts. Nonetheless, a reliable reference for this type of studies is still lacking. RESULTS: Taking advantage of digital gene expression (DGE) sequencing technology we compare all the available transcriptomes for S. mediterranea and improve their annotation. These results are accessible via web for the community of researchers. Using the quantitative nature of DGE, we describe the transcriptional profile of neoblasts and present 42 new neoblast genes, including several cancer-related genes and transcription factors. Furthermore, we describe in detail the Smed-meis-like gene and the three Nuclear Factor Y subunits Smed-nf-YA, Smed-nf-YB-2 and Smed-nf-YC. CONCLUSIONS: DGE is a valuable tool for gene discovery, quantification and annotation. The application of DGE in S. mediterranea confirms the planarian stem cells or neoblasts as a complex population of pluripotent and multipotent cells regulated by a mixture of transcription factors and cancer-related genes.


Subject(s)
Genes, Helminth , Planarians/genetics , RNA, Helminth/analysis , Sequence Analysis, RNA/methods , Stem Cells/cytology , Animals , CCAAT-Binding Factor/genetics , Gene Expression Profiling , Gene Expression Regulation , Homeodomain Proteins/genetics , Models, Animal , Molecular Sequence Data , Planarians/cytology , Stem Cells/metabolism
10.
Development ; 141(9): 1835-47, 2014 May.
Article in English | MEDLINE | ID: mdl-24700819

ABSTRACT

During the regeneration of freshwater planarians, polarity and patterning programs play essential roles in determining whether a head or a tail regenerates at anterior or posterior-facing wounds. This decision is made very soon after amputation. The pivotal role of the Wnt/ß-catenin and Hh signaling pathways in re-establishing anterior-posterior (AP) polarity has been well documented. However, the mechanisms that control the growth and differentiation of the blastema in accordance with its AP identity are less well understood. Previous studies have described a role of Smed-egfr-3, a planarian epidermal growth factor receptor, in blastema growth and differentiation. Here, we identify Smed-egr-4, a zinc-finger transcription factor belonging to the early growth response gene family, as a putative downstream target of Smed-egfr-3. Smed-egr-4 is mainly expressed in the central nervous system and its silencing inhibits anterior regeneration without affecting the regeneration of posterior regions. Single and combinatorial RNA interference to target different elements of the Wnt/ß-catenin pathway, together with expression analysis of brain- and anterior-specific markers, revealed that Smed-egr-4: (1) is expressed in two phases - an early Smed-egfr-3-independent phase and a late Smed-egfr-3-dependent phase; (2) is necessary for the differentiation of the brain primordia in the early stages of regeneration; and (3) that it appears to antagonize the activity of the Wnt/ß-catenin pathway to allow head regeneration. These results suggest that a conserved EGFR/egr pathway plays an important role in cell differentiation during planarian regeneration and indicate an association between early brain differentiation and the proper progression of head regeneration.


Subject(s)
Brain/embryology , Early Growth Response Transcription Factors/metabolism , ErbB Receptors/metabolism , Planarians/embryology , Planarians/physiology , Regeneration/physiology , Signal Transduction , Animals , Biomarkers/metabolism , Body Patterning/genetics , Brain/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Head/embryology , Models, Biological , Organogenesis , Planarians/genetics , RNA Interference , Regeneration/genetics , Signal Transduction/genetics , Time Factors
11.
Int J Dev Biol ; 56(1-3): 117-25, 2012.
Article in English | MEDLINE | ID: mdl-22450999

ABSTRACT

The planarian adult stem cell (pASC) population has a specific molecular signature and can be easily visualized and isolated by flow cytometry. However, the lack of antibodies against specific surface markers for planarian cells prevents a deeper analysis of specific cell populations. Here, if we describe the results of the immunoscreening of pASC plasma membrane proteins (PMPs). A novel papain-based method for planarian cell dissociation enabling both high yield and improved cell viability was used to generate single cell preparations for PMP purification. PMPs were used for intraperitoneal immunization of mice and thus about 1000 hybridoma clones were generated and screened. Supernatants collected from the hybridoma clones were first screened by ELISA and then by live immuno-staining. About half of these supernatants stained all the planarian cells, whereas the other half specifically labeled a subfraction thereof. A detailed analysis of two hybridoma supernatants revealed that large subfractions of the X1, X2 and Xin populations differentially express specific membrane markers. Quantitative PCR data disclosed a correlation between the immunostaining results and the expression of markers of the early and late progeny, also for those pASCs in the S/G2/M phase of the cell cycle (X1 population). Thus, about two thirds of the cycling pASCs showed a specific membrane signature coupled with the expression of markers hitherto considered to be restricted to differentiating, post-mitotic progeny. In summary, a library of 66 monoclonal antibodies against planarian PMPs was generated. The analysis of two of the clones generated revealed that a subset of cells of the X1 population expresses early and late progeny markers, which might indicate that these cells are committed while still proliferating. The findings demonstrate the usefulness of our PMP antibody library for planarian research.


Subject(s)
Cell Division/physiology , G2 Phase/physiology , Membrane Proteins/immunology , Planarians/physiology , S Phase/physiology , Stem Cells/physiology , Animals , Antibodies, Monoclonal/immunology , Biomarkers/metabolism , Blotting, Western , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Hybridomas , Immunization , Immunoenzyme Techniques , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Mice , Planarians/cytology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Stem Cells/cytology
12.
Int J Dev Biol ; 56(1-3): 83-91, 2012.
Article in English | MEDLINE | ID: mdl-22252539

ABSTRACT

The development of a complex multicellular organism requires a careful coordination of growth, cell division, cell differentiation and cell death. All these processes must be under intricate and coordinated control, as they have to be integrated across all tissues. Freshwater planarians are especially plastic, in that they constantly replace somatic tissues from a pool of adult somatic stem cells and continuously undergo growth and degrowth as adult animals in response to nutrient availability. During these processes they appear to maintain perfect scale of tissues and organs. These life history traits make them an ideal model system to study growth and degrowth. We have studied the unique planarian process of degrowth. When food is not available, planarians are able to degrow to a minimum size, without any signs of adverse physiological outcomes. For example they maintain full regenerative capacity. Our current knowledge of how this is regulated at the molecular and cellular level is very limited. Planarian degrowth has been reported to result from a decrease in cell number rather than a decrease in cell size. Thus one obvious explanation for degrowth would be a decrease in stem cell proliferation. However evidence in the literature suggests this is not the case. We show that planarians maintain normal basal mitotic rates during degrowth but that the number of stem cell progeny decreases during starvation and degrowth. These observations are reversed upon feeding, indicating that they are dependent on nutritional status. An increase in cell death is also observed during degrowth, which is not rapidly reversed upon feeding. We conclude that degrowth is a result of cell death decreasing cell numbers and that the dynamics of neoblast self-renewal and differentiation adapt to nutrient conditions to allow maintenance of the neoblast population during the period of starvation.


Subject(s)
Cell Proliferation , Mitosis/physiology , Planarians/cytology , Planarians/physiology , Regeneration/physiology , Starvation , Stem Cells/physiology , Animals , Blotting, Western , Cell Death , Cell Differentiation , Immunoenzyme Techniques , In Situ Hybridization , Models, Biological , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Stem Cells/cytology
13.
BMC Genomics ; 12: 133, 2011 Feb 28.
Article in English | MEDLINE | ID: mdl-21356107

ABSTRACT

BACKGROUND: In recent years, planaria have emerged as an important model system for research into stem cells and regeneration. Attention is focused on their unique stem cells, the neoblasts, which can differentiate into any cell type present in the adult organism. Sequencing of the Schmidtea mediterranea genome and some expressed sequence tag projects have generated extensive data on the genetic profile of these cells. However, little information is available on their protein dynamics. RESULTS: We developed a proteomic strategy to identify neoblast-specific proteins. Here we describe the method and discuss the results in comparison to the genomic high-throughput analyses carried out in planaria and to proteomic studies using other stem cell systems. We also show functional data for some of the candidate genes selected in our proteomic approach. CONCLUSIONS: We have developed an accurate and reliable mass-spectra-based proteomics approach to complement previous genomic studies and to further achieve a more accurate understanding and description of the molecular and cellular processes related to the neoblasts.


Subject(s)
Planarians/cytology , Proteomics/methods , Stem Cells/metabolism , Animals , Cloning, Molecular , Computational Biology , Electrophoresis, Gel, Two-Dimensional , Genome, Helminth , In Situ Hybridization , Planarians/genetics , Planarians/metabolism , RNA Interference , Stem Cells/cytology
14.
BMC Genomics ; 11: 731, 2010 Dec 31.
Article in English | MEDLINE | ID: mdl-21194483

ABSTRACT

BACKGROUND: Freshwater planarians are an attractive model for regeneration and stem cell research and have become a promising tool in the field of regenerative medicine. With the availability of a sequenced planarian genome, the recent application of modern genetic and high-throughput tools has resulted in revitalized interest in these animals, long known for their amazing regenerative capabilities, which enable them to regrow even a new head after decapitation. However, a detailed description of the planarian transcriptome is essential for future investigation into regenerative processes using planarians as a model system. RESULTS: In order to complement and improve existing gene annotations, we used a 454 pyrosequencing approach to analyze the transcriptome of the planarian species Schmidtea mediterranea Altogether, 598,435 454-sequencing reads, with an average length of 327 bp, were assembled together with the ~10,000 sequences of the S. mediterranea UniGene set using different similarity cutoffs. The assembly was then mapped onto the current genome data. Remarkably, our Smed454 dataset contains more than 3 million novel transcribed nucleotides sequenced for the first time. A descriptive analysis of planarian splice sites was conducted on those Smed454 contigs that mapped univocally to the current genome assembly. Sequence analysis allowed us to identify genes encoding putative proteins with defined structural properties, such as transmembrane domains. Moreover, we annotated the Smed454 dataset using Gene Ontology, and identified putative homologues of several gene families that may play a key role during regeneration, such as neurotransmitter and hormone receptors, homeobox-containing genes, and genes related to eye function. CONCLUSIONS: We report the first planarian transcript dataset, Smed454, as an open resource tool that can be accessed via a web interface. Smed454 contains significant novel sequence information about most expressed genes of S. mediterranea. Analysis of the annotated data promises to contribute to identification of gene families poorly characterized at a functional level. The Smed454 transcriptome data will assist in the molecular characterization of S. mediterranea as a model organism, which will be useful to a broad scientific community.


Subject(s)
Databases, Genetic , Gene Expression Profiling , Planarians/genetics , Sequence Analysis, DNA/methods , Animals , Base Composition/genetics , Base Sequence , Cell Cycle/genetics , Cell Division/genetics , Consensus Sequence/genetics , DNA Damage/genetics , DNA Repair/genetics , Eye/metabolism , Genome/genetics , Homeodomain Proteins/genetics , Internet , Membrane Proteins/genetics , Molecular Sequence Annotation , Neurotransmitter Agents/genetics , Planarians/cytology , RNA Splice Sites/genetics , Receptors, Cell Surface/genetics , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
15.
Int J Dev Biol ; 53(8-10): 1317-27, 2009.
Article in English | MEDLINE | ID: mdl-19247944

ABSTRACT

Planarians can undergo dramatic changes in body size and regenerate their entire body plan from small pieces after cutting. This remarkable morphological plasticity has made them an excellent model in which to analyze phenomena such as morphogenesis, restoration of pattern and polarity, control of tissue proportions and tissue homeostasis. They have a unique population of pluripotent stem cells in the adult that can give rise to all differentiated cell types, including the germ cells. These cellular characteristics provide an excellent opportunity to study the mechanisms involved in the maintenance and differentiation of cell populations in intact and regenerating animals. Until recently, the planarian model system lacked opportunities for genetic analysis; however, this handicap was overcome in the last decade through the development of new molecular methods which have been successfully applied to planarians. These techniques have allowed analysis of the temporal and spatial expression of genes, as well as interference with gene function, generating the first phenotypes by loss or gain of function. Finally, the sequencing of the planarian genome has provided the essential tools for an in-depth analysis of the genomic regulation of this model system. In this review, we provide an overview of planarians as a model system for research into development and regeneration and describe new lines of investigation in this area.


Subject(s)
Morphogenesis/physiology , Planarians/physiology , Regeneration/physiology , Animals , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Microscopy, Electron , Models, Biological , Morphogenesis/genetics , Planarians/cytology , Planarians/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/ultrastructure , Regeneration/genetics , Research/trends , Research Design
SELECTION OF CITATIONS
SEARCH DETAIL