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










Publication year range
1.
Nat Commun ; 12(1): 7019, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857757

ABSTRACT

Yolk sac (YS) hematopoiesis is critical for the survival of the embryo and a major source of tissue-resident macrophages that persist into adulthood. Yet, the transcriptional and epigenetic regulation of YS hematopoiesis remains poorly characterized. Here we report that the epigenetic regulator Ezh2 is essential for YS hematopoiesis but dispensable for subsequent aorta-gonad-mesonephros (AGM) blood development. Loss of EZH2 activity in hemogenic endothelium (HE) leads to the generation of phenotypically intact but functionally deficient erythro-myeloid progenitors (EMPs), while the generation of primitive erythroid cells is not affected. EZH2 activity is critical for the generation of functional EMPs at the onset of the endothelial-to-hematopoietic transition but subsequently dispensable. We identify a lack of Wnt signaling downregulation as the primary reason for the production of non-functional EMPs. Together, our findings demonstrate a critical and stage-specific role of Ezh2 in modulating Wnt signaling during the generation of EMPs from YS HE.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Erythroid Cells/metabolism , Gene Expression Regulation, Developmental , Mouse Embryonic Stem Cells/metabolism , Myeloid Progenitor Cells/metabolism , Vesicular Transport Proteins/genetics , Yolk Sac/metabolism , Animals , Cell Differentiation , Embryo, Mammalian , Enhancer of Zeste Homolog 2 Protein/deficiency , Epigenesis, Genetic , Erythroid Cells/cytology , Female , Fetus , Genes, Reporter , Hematopoiesis/genetics , Liver/cytology , Liver/growth & development , Liver/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Myeloid Progenitor Cells/pathology , Primary Cell Culture , Vesicular Transport Proteins/metabolism , Wnt Signaling Pathway , Yolk Sac/cytology , Yolk Sac/growth & development , Red Fluorescent Protein
2.
J Mol Med (Berl) ; 98(9): 1301-1317, 2020 09.
Article in English | MEDLINE | ID: mdl-32778925

ABSTRACT

The IgMi mouse has normal B cell development; its B cells express an IgM B cell receptor but cannot class switch or secrete antibody. Thus, the IgMi mouse offers a model system by which to dissect out antibody-dependent and antibody-independent B cell function. Here, we provide the first detailed characterisation of the IgMi mouse post-Trichuris muris (T. muris) infection, describing expulsion phenotype, cytokine production, gut pathology and changes in T regulatory cells, T follicular helper cells and germinal centre B cells, in addition to RNA sequencing (RNA seq) analyses of wild-type littermates (WT) and mutant B cells prior to and post infection. IgMi mice were susceptible to a high-dose infection, with reduced Th2 cytokines and elevated B cell-derived IL-10 in mesenteric lymph nodes (MLN) compared to controls. A low-dose infection regime revealed IgMi mice to have significantly more apoptotic cells in the gut compared to WT mice, but no change in intestinal inflammation. IL-10 levels were again elevated. Collectively, this study showcases the potential of the IgMi mouse as a tool for understanding B cell biology and suggests that the B cell plays both antibody-dependent and antibody-independent roles post high- and low-dose T. muris infection. KEY MESSAGES: During a high-dose T. muris infection, B cells are important in maintaining the Th1/Th2 balance in the MLN through an antibody-independent mechanism. High levels of IL-10 in the MLN early post-infection, and the presence of IL-10-producing B cells, correlates with susceptibility to T. muris infection. B cells maintain gut homeostasis during chronic T. muris infection via an antibody-dependent mechanism.


Subject(s)
Antibodies/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Host-Parasite Interactions/immunology , Trichuriasis/immunology , Trichuriasis/parasitology , Trichuris/immunology , Animals , Apoptosis , Cytokines/biosynthesis , Disease Models, Animal , Disease Susceptibility/immunology , Female , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Knockout , Parasite Load
3.
Stem Cell Reports ; 11(5): 1061-1074, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30449319

ABSTRACT

The differentiation of human embryonic stem cells (hESCs) to hematopoietic lineages initiates with the specification of hemogenic endothelium, a transient specialized endothelial precursor of all blood cells. This in vitro system provides an invaluable model to dissect the emergence of hematopoiesis in humans. However, the study of hematopoiesis specification is hampered by a lack of consensus in the timing of hemogenic endothelium analysis and the full hematopoietic potential of this population. Here, our data reveal a sharp decline in the hemogenic potential of endothelium populations isolated over the course of hESC differentiation. Furthermore, by tracking the dynamic expression of CD31 and CD235a at the onset of hematopoiesis, we identified three populations of hematopoietic progenitors, representing primitive and definitive subsets that all emerge from the earliest specified hemogenic endothelium. Our data establish that hemogenic endothelium populations endowed with primitive and definitive hematopoietic potential are specified simultaneously from the mesoderm in differentiating hESCs.


Subject(s)
Hemangioblasts/metabolism , Hematopoiesis , Antigens, CD/metabolism , Cell Differentiation , Cell Lineage , Embryoid Bodies/cytology , Endothelial Cells/cytology , Humans , Stromal Cells/cytology , Transcription, Genetic
4.
Stem Cell Reports ; 10(4): 1369-1383, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29641990

ABSTRACT

The first hematopoietic stem and progenitor cells are generated during development from hemogenic endothelium (HE) through trans-differentiation. The molecular mechanisms underlying this endothelial-to-hematopoietic transition (EHT) remain poorly understood. Here, we explored the role of the epigenetic regulators HDAC1 and HDAC2 in the emergence of these first blood cells in vitro and in vivo. Loss of either of these epigenetic silencers through conditional genetic deletion reduced hematopoietic transition from HE, while combined deletion was incompatible with blood generation. We investigated the molecular basis of HDAC1 and HDAC2 requirement and identified TGF-ß signaling as one of the pathways controlled by HDAC1 and HDAC2. Accordingly, we experimentally demonstrated that activation of this pathway in HE cells reinforces hematopoietic development. Altogether, our results establish that HDAC1 and HDAC2 modulate TGF-ß signaling and suggest that stimulation of this pathway in HE cells would be beneficial for production of hematopoietic cells for regenerative therapies.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Hematopoiesis , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Benzamides/pharmacology , Cell Differentiation/drug effects , Dioxoles/pharmacology , Endothelial Cells/drug effects , Gene Deletion , Hemangioblasts/cytology , Hematopoiesis/drug effects , Histone Deacetylase 1/deficiency , Histone Deacetylase 2/deficiency , Histone Deacetylase Inhibitors/pharmacology , Mice , Signal Transduction/drug effects
5.
PLoS Genet ; 14(1): e1007127, 2018 01.
Article in English | MEDLINE | ID: mdl-29300724

ABSTRACT

In recent years, highly detailed characterization of adult bone marrow (BM) myeloid progenitors has been achieved and, as a result, the impact of somatic defects on different hematopoietic lineage fate decisions can be precisely determined. Fetal liver (FL) hematopoietic progenitor cells (HPCs) are poorly characterized in comparison, potentially hindering the study of the impact of genetic alterations on midgestation hematopoiesis. Numerous disorders, for example infant acute leukemias, have in utero origins and their study would therefore benefit from the ability to isolate highly purified progenitor subsets. We previously demonstrated that a Runx1 distal promoter (P1)-GFP::proximal promoter (P2)-hCD4 dual-reporter mouse (Mus musculus) model can be used to identify adult BM progenitor subsets with distinct lineage preferences. In this study, we undertook the characterization of the expression of Runx1-P1-GFP and P2-hCD4 in FL. Expression of P2-hCD4 in the FL immunophenotypic Megakaryocyte-Erythroid Progenitor (MEP) and Common Myeloid Progenitor (CMP) compartments corresponded to increased granulocytic/monocytic/megakaryocytic and decreased erythroid specification. Moreover, Runx1-P2-hCD4 expression correlated with several endogenous cell surface markers' expression, including CD31 and CD45, providing a new strategy for prospective identification of highly purified fetal myeloid progenitors in transgenic mouse models. We utilized this methodology to compare the impact of the deletion of either total RUNX1 or RUNX1C alone and to determine the fetal HPCs lineages most substantially affected. This new prospective identification of FL progenitors therefore raises the prospect of identifying the underlying gene networks responsible with greater precision than previously possible.


Subject(s)
Cell Lineage/genetics , Hematopoietic Stem Cells/cytology , Myeloid Progenitor Cells/cytology , Animals , Bone Marrow/embryology , Cell Differentiation , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Disease Models, Animal , Granulocytes/cytology , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Liver/cytology , Liver/embryology , Liver/metabolism , Megakaryocytes/cytology , Mice , Mice, Transgenic , Monocytes/cytology , Prospective Studies
6.
Blood ; 130(3): 271-284, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28490570

ABSTRACT

RUNX1 is crucial for the regulation of megakaryocyte specification, maturation, and thrombopoiesis. Runx1 possesses 2 promoters: the distal P1 and proximal P2 promoters. The major protein isoforms generated by P1 and P2 are RUNX1C and RUNX1B, respectively, which differ solely in their N-terminal amino acid sequences. RUNX1C is the most abundantly expressed isoform in adult hematopoiesis, present in all RUNX1-expressing populations, including the cKit+ hematopoietic stem and progenitor cells. RUNX1B expression is more restricted, being highly expressed in the megakaryocyte lineage but downregulated during erythropoiesis. We generated a Runx1 P1 knock-in of RUNX1B, termed P1-MRIPV This mouse line lacks RUNX1C expression but has normal total RUNX1 levels, solely comprising RUNX1B. Using this mouse line, we establish a specific requirement for the P1-RUNX1C isoform in megakaryopoiesis, which cannot be entirely compensated for by RUNX1B overexpression. P1 knock-in megakaryocyte progenitors have reduced proliferative capacity and undergo increased cell death, resulting in thrombocytopenia. P1 knock-in premegakaryocyte/erythroid progenitors demonstrate an erythroid-specification bias, evident from increased erythroid colony-forming ability and decreased megakaryocyte output. At a transcriptional level, multiple erythroid-specific genes are upregulated and megakaryocyte-specific transcripts are downregulated. In addition, proapoptotic pathways are activated in P1 knock-in premegakaryocyte/erythroid progenitors, presumably accounting for the increased cell death in the megakaryocyte progenitor compartment. Unlike in the conditional adult Runx1 null models, megakaryocytic maturation is not affected in the P1 knock-in mice, suggesting that RUNX1B can regulate endomitosis and thrombopoiesis. Therefore, despite the high degree of structural similarity, RUNX1B and RUNX1C isoforms have distinct and specific roles in adult megakaryopoiesis.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Megakaryocyte Progenitor Cells/metabolism , Megakaryocytes/metabolism , RNA, Messenger/genetics , Thrombocytopenia/genetics , Thrombopoiesis/genetics , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Death , Cell Lineage/genetics , Cell Proliferation , Core Binding Factor Alpha 2 Subunit/metabolism , Flow Cytometry , Gene Expression Regulation , Humans , Immunophenotyping , Megakaryocyte Progenitor Cells/pathology , Megakaryocytes/pathology , Mice , Mice, Transgenic , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Thrombocytopenia/metabolism , Thrombocytopenia/pathology
7.
Cancer Cell ; 31(5): 621-634.e6, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28416184

ABSTRACT

Aberrant WNT signaling drives colorectal cancer (CRC). Here, we identify TIAM1 as a critical antagonist of CRC progression through inhibiting TAZ and YAP, effectors of WNT signaling. We demonstrate that TIAM1 shuttles between the cytoplasm and nucleus antagonizing TAZ/YAP by distinct mechanisms in the two compartments. In the cytoplasm, TIAM1 localizes to the destruction complex and promotes TAZ degradation by enhancing its interaction with ßTrCP. Nuclear TIAM1 suppresses TAZ/YAP interaction with TEADs, inhibiting expression of TAZ/YAP target genes implicated in epithelial-mesenchymal transition, cell migration, and invasion, and consequently suppresses CRC cell migration and invasion. Importantly, high nuclear TIAM1 in clinical specimens associates with increased CRC patient survival. Together, our findings suggest that in CRC TIAM1 suppresses tumor progression by regulating YAP/TAZ activity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Colorectal Neoplasms/metabolism , Epithelial Cells/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Active Transport, Cell Nucleus , Adaptor Proteins, Signal Transducing/genetics , Animals , Caco-2 Cells , Cell Cycle Proteins , Cell Nucleus/metabolism , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cytoplasm/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Humans , Intestinal Mucosa/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness , Phenotype , Phosphoproteins/genetics , Proteolysis , RNA Interference , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , Trans-Activators , Transcription Factors , Transcription, Genetic , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transfection , Wnt Signaling Pathway , YAP-Signaling Proteins , Zebrafish/embryology , beta-Transducin Repeat-Containing Proteins/genetics , beta-Transducin Repeat-Containing Proteins/metabolism
8.
Development ; 143(23): 4341-4351, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27802172

ABSTRACT

Endothelial to hematopoietic transition (EHT) is a dynamic process involving the shutting down of endothelial gene expression and switching on of hematopoietic gene transcription. Although the factors regulating EHT in hemogenic endothelium (HE) of the dorsal aorta have been relatively well studied, the molecular regulation of yolk sac HE remains poorly understood. Here, we show that SOX7 inhibits the expression of RUNX1 target genes in HE, while having no effect on RUNX1 expression itself. We establish that SOX7 directly interacts with RUNX1 and inhibits its transcriptional activity. Through this interaction we demonstrate that SOX7 hinders RUNX1 DNA binding as well as the interaction between RUNX1 and its co-factor CBFß. Finally, we show by single-cell expression profiling and immunofluorescence that SOX7 is broadly expressed across the RUNX1+ yolk sac HE population compared with SOX17. Collectively, these data demonstrate for the first time how direct protein-protein interactions between endothelial and hematopoietic transcription factors regulate contrasting transcriptional programs during HE differentiation and EHT.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Endothelium/cytology , Hemangioblasts/cytology , SOXF Transcription Factors/metabolism , Yolk Sac/cytology , Animals , Cell Differentiation , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/antagonists & inhibitors , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , HMGB Proteins/metabolism , Hematopoietic Stem Cells/cytology , Male , Mice , Mice, Inbred C57BL , SOXF Transcription Factors/genetics , Transcription, Genetic/physiology
10.
Cell Rep ; 15(10): 2185-2199, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27239041

ABSTRACT

It is now well-established that hematopoietic stem cells (HSCs) and progenitor cells originate from a specialized subset of endothelium, termed hemogenic endothelium (HE), via an endothelial-to-hematopoietic transition. However, the molecular mechanisms determining which endothelial progenitors possess this hemogenic potential are currently unknown. Here, we investigated the changes in hemogenic potential in endothelial progenitors at the early stages of embryonic development. Using an ETV2::GFP reporter mouse to isolate emerging endothelial progenitors, we observed a dramatic decrease in hemogenic potential between embryonic day (E)7.5 and E8.5. At the molecular level, Runx1 is expressed at much lower levels in E8.5 intra-embryonic progenitors, while Bmi1 expression is increased. Remarkably, the ectopic expression of Runx1 in these progenitors fully restores their hemogenic potential, as does the suppression of BMI1 function. Altogether, our data demonstrate that hemogenic competency in recently specified endothelial progenitors is restrained through the active silencing of Runx1 expression.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Embryonic Development , Endothelial Progenitor Cells/metabolism , Gene Silencing , Hemangioblasts/cytology , Animals , Bone Morphogenetic Protein 4/metabolism , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Endothelial Progenitor Cells/cytology , Female , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/metabolism , Hemangioblasts/metabolism , Hematopoiesis/genetics , Immunophenotyping , Male , Mice, Inbred ICR , Oligonucleotide Array Sequence Analysis , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Single-Cell Analysis , Smad Proteins/metabolism , Transcription Factors/metabolism
11.
PLoS Genet ; 12(1): e1005814, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26808730

ABSTRACT

The Core Binding Factor (CBF) protein RUNX1 is a master regulator of definitive hematopoiesis, crucial for hematopoietic stem cell (HSC) emergence during ontogeny. RUNX1 also plays vital roles in adult mice, in regulating the correct specification of numerous blood lineages. Akin to the other mammalian Runx genes, Runx1 has two promoters P1 (distal) and P2 (proximal) which generate distinct protein isoforms. The activities and specific relevance of these two promoters in adult hematopoiesis remain to be fully elucidated. Utilizing a dual reporter mouse model we demonstrate that the distal P1 promoter is broadly active in adult hematopoietic stem and progenitor cell (HSPC) populations. By contrast the activity of the proximal P2 promoter is more restricted and its upregulation, in both the immature Lineage- Sca1high cKithigh (LSK) and bipotential Pre-Megakaryocytic/Erythroid Progenitor (PreMegE) populations, coincides with a loss of erythroid (Ery) specification. Accordingly the PreMegE population can be prospectively separated into "pro-erythroid" and "pro-megakaryocyte" populations based on Runx1 P2 activity. Comparative gene expression analyses between Runx1 P2+ and P2- populations indicated that levels of CD34 expression could substitute for P2 activity to distinguish these two cell populations in wild type (WT) bone marrow (BM). Prospective isolation of these two populations will enable the further investigation of molecular mechanisms involved in megakaryocytic/erythroid (Mk/Ery) cell fate decisions. Having characterized the extensive activity of P1, we utilized a P1-GFP homozygous mouse model to analyze the impact of the complete absence of Runx1 P1 expression in adult mice and observed strong defects in the T cell lineage. Finally, we investigated how the leukemic fusion protein AML1-ETO9a might influence Runx1 promoter usage. Short-term AML1-ETO9a induction in BM resulted in preferential P2 upregulation, suggesting its expression may be important to establish a pre-leukemic environment.


Subject(s)
Cell Lineage/genetics , Core Binding Factor Alpha 2 Subunit/biosynthesis , Hematopoiesis/genetics , Hematopoietic Stem Cells , Animals , Cell Differentiation/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental , Megakaryocytes/cytology , Mice , Promoter Regions, Genetic , T-Lymphocytes/metabolism
12.
Nature ; 518(7540): 495-501, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25719666

ABSTRACT

Pancreatic cancer remains one of the most lethal of malignancies and a major health burden. We performed whole-genome sequencing and copy number variation (CNV) analysis of 100 pancreatic ductal adenocarcinomas (PDACs). Chromosomal rearrangements leading to gene disruption were prevalent, affecting genes known to be important in pancreatic cancer (TP53, SMAD4, CDKN2A, ARID1A and ROBO2) and new candidate drivers of pancreatic carcinogenesis (KDM6A and PREX2). Patterns of structural variation (variation in chromosomal structure) classified PDACs into 4 subtypes with potential clinical utility: the subtypes were termed stable, locally rearranged, scattered and unstable. A significant proportion harboured focal amplifications, many of which contained druggable oncogenes (ERBB2, MET, FGFR1, CDK6, PIK3R3 and PIK3CA), but at low individual patient prevalence. Genomic instability co-segregated with inactivation of DNA maintenance genes (BRCA1, BRCA2 or PALB2) and a mutational signature of DNA damage repair deficiency. Of 8 patients who received platinum therapy, 4 of 5 individuals with these measures of defective DNA maintenance responded.


Subject(s)
DNA Mutational Analysis , Genome, Human/genetics , Genomics , Mutation/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , DNA Repair/genetics , Female , Genes, BRCA1 , Genes, BRCA2 , Genetic Markers/genetics , Genomic Instability/genetics , Genotype , Humans , Mice , Pancreatic Neoplasms/classification , Pancreatic Neoplasms/drug therapy , Platinum/pharmacology , Point Mutation/genetics , Poly(ADP-ribose) Polymerase Inhibitors , Xenograft Model Antitumor Assays
13.
Biotechniques ; 57(1): 31-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25005691

ABSTRACT

Somatic rearrangements, which are commonly found in human cancer genomes, contribute to the progression and maintenance of cancers. Conventionally, the verification of somatic rearrangements comprises many manual steps and Sanger sequencing. This is labor intensive when verifying a large number of rearrangements in a large cohort. To increase the verification throughput, we devised a high-throughput workflow that utilizes benchtop next-generation sequencing and in-house bioinformatics tools to link the laboratory processes. In the proposed workflow, primers are automatically designed. PCR and an optional gel electrophoresis step to confirm the somatic nature of the rearrangements are performed. PCR products of somatic events are pooled for Ion Torrent PGM and/or Illumina MiSeq sequencing, the resulting sequence reads are assembled into consensus contigs by a consensus assembler, and an automated BLAT is used to resolve the breakpoints to base level. We compared sequences and breakpoints of verified somatic rearrangements between the conventional and high-throughput workflow. The results showed that next-generation sequencing methods are comparable to conventional Sanger sequencing. The identified breakpoints obtained from next-generation sequencing methods were highly accurate and reproducible. Furthermore, the proposed workflow allows hundreds of events to be processed in a shorter time frame compared with the conventional workflow.


Subject(s)
Chromosome Breakpoints , Chromosomes, Human , High-Throughput Nucleotide Sequencing/methods , Base Sequence , Carcinoma, Pancreatic Ductal/genetics , Chromosome Aberrations , DNA Primers , Humans , Molecular Sequence Data , Neoplasms/genetics , Pancreatic Neoplasms/genetics , Polymerase Chain Reaction/methods , Workflow
SELECTION OF CITATIONS
SEARCH DETAIL
...