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 ; 545(7655): 439-445, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28514438

ABSTRACT

Developmental pathways that orchestrate the fleeting transition of endothelial cells into haematopoietic stem cells remain undefined. Here we demonstrate a tractable approach for fully reprogramming adult mouse endothelial cells to haematopoietic stem cells (rEC-HSCs) through transient expression of the transcription-factor-encoding genes Fosb, Gfi1, Runx1, and Spi1 (collectively denoted hereafter as FGRS) and vascular-niche-derived angiocrine factors. The induction phase (days 0-8) of conversion is initiated by expression of FGRS in mature endothelial cells, which results in endogenous Runx1 expression. During the specification phase (days 8-20), RUNX1+ FGRS-transduced endothelial cells commit to a haematopoietic fate, yielding rEC-HSCs that no longer require FGRS expression. The vascular niche drives a robust self-renewal and expansion phase of rEC-HSCs (days 20-28). rEC-HSCs have a transcriptome and long-term self-renewal capacity similar to those of adult haematopoietic stem cells, and can be used for clonal engraftment and serial primary and secondary multi-lineage reconstitution, including antigen-dependent adaptive immune function. Inhibition of TGFß and CXCR7 or activation of BMP and CXCR4 signalling enhanced generation of rEC-HSCs. Pluripotency-independent conversion of endothelial cells into autologous authentic engraftable haematopoietic stem cells could aid treatment of haematological disorders.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Endothelium/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Adaptive Immunity , Aging/genetics , Animals , Cell Line , Cell Lineage , Cell Self Renewal , Clone Cells/cytology , Clone Cells/transplantation , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
2.
Nat Commun ; 8: 13963, 2017 01 16.
Article in English | MEDLINE | ID: mdl-28091527

ABSTRACT

Transplanting vascular endothelial cells (ECs) to support metabolism and express regenerative paracrine factors is a strategy to treat vasculopathies and to promote tissue regeneration. However, transplantation strategies have been challenging to develop, because ECs are difficult to culture and little is known about how to direct them to stably integrate into vasculature. Here we show that only amniotic cells could convert to cells that maintain EC gene expression. Even so, these converted cells perform sub-optimally in transplantation studies. Constitutive Akt signalling increases expression of EC morphogenesis genes, including Sox17, shifts the genomic targeting of Fli1 to favour nearby Sox consensus sites and enhances the vascular function of converted cells. Enforced expression of Sox17 increases expression of morphogenesis genes and promotes integration of transplanted converted cells into injured vessels. Thus, Ets transcription factors specify non-vascular, amniotic cells to EC-like cells, whereas Sox17 expression is required to confer EC function.


Subject(s)
Endothelial Cells/transplantation , Endothelium, Vascular/metabolism , SOXF Transcription Factors/metabolism , Vascular Diseases/therapy , Amnion/cytology , Amnion/embryology , Amnion/metabolism , Animals , Endothelial Cells/metabolism , Endothelium, Vascular/physiopathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Regeneration , SOXF Transcription Factors/genetics , Vascular Diseases/genetics , Vascular Diseases/metabolism , Vascular Diseases/physiopathology
3.
Development ; 143(5): 774-9, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26811383

ABSTRACT

Congenital heart defects are the most common birth defects in humans, and those that affect the proper alignment of the outflow tracts and septation of the ventricles are a highly significant cause of morbidity and mortality in infants. A late differentiating population of cardiac progenitors, referred to as the anterior second heart field (AHF), gives rise to the outflow tract and the majority of the right ventricle and provides an embryological context for understanding cardiac outflow tract alignment and membranous ventricular septal defects. However, the transcriptional pathways controlling AHF development and their roles in congenital heart defects remain incompletely elucidated. Here, we inactivated the gene encoding the transcription factor MEF2C in the AHF in mice. Loss of Mef2c function in the AHF results in a spectrum of outflow tract alignment defects ranging from overriding aorta to double-outlet right ventricle and dextro-transposition of the great arteries. We identify Tdgf1, which encodes a Nodal co-receptor (also known as Cripto), as a direct transcriptional target of MEF2C in the outflow tract via an AHF-restricted Tdgf1 enhancer. Importantly, both the MEF2C and TDGF1 genes are associated with congenital heart defects in humans. Thus, these studies establish a direct transcriptional pathway between the core cardiac transcription factor MEF2C and the human congenital heart disease gene TDGF1. Moreover, we found a range of outflow tract alignment defects resulting from a single genetic lesion, supporting the idea that AHF-derived outflow tract alignment defects may constitute an embryological spectrum rather than distinct anomalies.


Subject(s)
Epidermal Growth Factor/physiology , Gene Expression Regulation, Developmental , Membrane Glycoproteins/physiology , Neoplasm Proteins/physiology , Animals , Animals, Newborn , Disease Models, Animal , Epidermal Growth Factor/genetics , Female , Gene Deletion , Heart/embryology , Heart Defects, Congenital/genetics , Heart Septal Defects, Ventricular/genetics , Heart Ventricles , Humans , In Situ Hybridization , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/physiology , Male , Membrane Glycoproteins/genetics , Mice , Morphogenesis/genetics , Neoplasm Proteins/genetics , Organogenesis , Sequence Analysis, RNA , Tissue Distribution , Transcription, Genetic , Transposition of Great Vessels/genetics
4.
Nat Protoc ; 10(12): 1975-85, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26540589

ABSTRACT

Endothelial cells (ECs) have essential roles in organ development and regeneration, and therefore they could be used for regenerative therapies. However, generation of abundant functional endothelium from pluripotent stem cells has been difficult because ECs generated by many existing strategies have limited proliferative potential and display vascular instability. The latter difficulty is of particular importance because cells that lose their identity over time could be unsuitable for therapeutic use. Here, we describe a 3-week platform for directly converting human mid-gestation lineage-committed amniotic fluid-derived cells (ACs) into a stable and expandable population of vascular ECs (rAC-VECs) without using pluripotency factors. By transient expression of the ETS transcription factor ETV2 for 2 weeks and constitutive expression the ETS transcription factors FLI1 and ERG1, concomitant with TGF-ß inhibition for 3 weeks, epithelial and mesenchymal ACs are converted, with high efficiency, into functional rAC-VECs. These rAC-VECs maintain their vascular repertoire and morphology over numerous passages in vitro, and they form functional vessels when implanted in vivo. rAC-VECs can be detected in recipient mice months after implantation. Thus, rAC-VECs can be used to establish a cellular platform to uncover the molecular determinants of vascular development and heterogeneity and potentially represent ideal ECs for the treatment of regenerative disorders.


Subject(s)
Amniotic Fluid/cytology , Cell Transdifferentiation , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Animals , Cell Culture Techniques/methods , Cells, Cultured , Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Endothelial Cells/transplantation , Gene Expression Regulation, Developmental , Humans , Mice, SCID , Proto-Oncogene Protein c-fli-1/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/antagonists & inhibitors
5.
Stem Cell Reports ; 5(5): 881-894, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26441307

ABSTRACT

Hematopoietic stem cells (HSCs) inhabit distinct microenvironments within the adult bone marrow (BM), which govern the delicate balance between HSC quiescence, self-renewal, and differentiation. Previous reports have proposed that HSCs localize to the vascular niche, comprised of endothelium and tightly associated perivascular cells. Herein, we examine the capacity of BM endothelial cells (BMECs) to support ex vivo and in vivo hematopoiesis. We demonstrate that AKT1-activated BMECs (BMEC-Akt1) have a unique transcription factor/cytokine profile that supports functional HSCs in lieu of complex serum and cytokine supplementation. Additionally, transplantation of BMEC-Akt1 cells enhanced regenerative hematopoiesis following myeloablative irradiation. These data demonstrate that BMEC-Akt1 cultures can be used as a platform for the discovery of pro-HSC factors and justify the utility of BMECs as a cellular therapy. This technical advance may lead to the development of therapies designed to decrease pancytopenias associated with myeloablative regimens used to treat a wide array of disease states.


Subject(s)
Endothelial Progenitor Cells/cytology , Hematopoiesis , Stem Cell Niche , Animals , Cytokines/metabolism , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/transplantation , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Stromal Cells/transplantation
6.
Dev Cell ; 26(1): 45-58, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23830865

ABSTRACT

Vegf signaling specifies arterial fate during early vascular development by inducing the transcription of Delta-like 4 (Dll4), the earliest Notch ligand gene expressed in arterial precursor cells. Dll4 expression precedes that of Notch receptors in arteries, and factors that direct its arterial-specific expression are not known. To identify the transcriptional program that initiates arterial Dll4 expression, we characterized an arterial-specific and Vegf-responsive enhancer of Dll4. Our findings demonstrate that Notch signaling is not required for initiation of Dll4 expression in arteries and suggest that Notch instead functions as a maintenance factor. Importantly, we find that Vegf signaling activates MAP kinase (MAPK)-dependent E26 transformation-specific sequence (ETS) factors in the arterial endothelium to drive expression of Dll4 and Notch4. These findings identify a Vegf/MAPK-dependent transcriptional pathway that specifies arterial identity by activating Notch signaling components and illustrate how signaling cascades can modulate broadly expressed transcription factors to achieve tissue-specific transcriptional outputs.


Subject(s)
Aorta/physiology , Gene Expression Regulation, Developmental , Vascular Endothelial Growth Factor A/metabolism , Adaptor Proteins, Signal Transducing , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/metabolism , Aorta/metabolism , Binding Sites , Calcium-Binding Proteins , Endocardium/embryology , Endocardium/metabolism , Enhancer Elements, Genetic , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Organ Specificity , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor, Notch4 , Receptors, Notch/genetics , Receptors, Notch/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic , Transcriptional Regulator ERG , Vascular Endothelial Growth Factor A/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
7.
Dev Cell ; 26(2): 204-19, 2013 Jul 29.
Article in English | MEDLINE | ID: mdl-23871589

ABSTRACT

Microvascular endothelial cells (ECs) within different tissues are endowed with distinct but as yet unrecognized structural, phenotypic, and functional attributes. We devised EC purification, cultivation, profiling, and transplantation models that establish tissue-specific molecular libraries of ECs devoid of lymphatic ECs or parenchymal cells. These libraries identify attributes that confer ECs with their organotypic features. We show that clusters of transcription factors, angiocrine growth factors, adhesion molecules, and chemokines are expressed in unique combinations by ECs of each organ. Furthermore, ECs respond distinctly in tissue regeneration models, hepatectomy, and myeloablation. To test the data set, we developed a transplantation model that employs generic ECs differentiated from embryonic stem cells. Transplanted generic ECs engraft into regenerating tissues and acquire features of organotypic ECs. Collectively, we demonstrate the utility of informational databases of ECs toward uncovering the extravascular and intrinsic signals that define EC heterogeneity. These factors could be exploited therapeutically to engineer tissue-specific ECs for regeneration.


Subject(s)
Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Endothelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Regeneration , Transcription Factors/metabolism , Animals , Cell Adhesion Molecules/biosynthesis , Cell Differentiation , Cells, Cultured , Chemokines/biosynthesis , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Endothelial Cells/cytology , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Mice , Microvessels/metabolism , Transcription Factors/biosynthesis
8.
Cell ; 151(3): 559-75, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23084400

ABSTRACT

ETS transcription factors ETV2, FLI1, and ERG1 specify pluripotent stem cells into induced vascular endothelial cells (iVECs). However, iVECs are unstable and drift toward nonvascular cells. We show that human midgestation c-Kit(-) lineage-committed amniotic cells (ACs) can be reprogrammed into vascular endothelial cells (rAC-VECs) without transitioning through a pluripotent state. Transient ETV2 expression in ACs generates immature rAC-VECs, whereas coexpression with FLI1/ERG1 endows rAC-VECs with a vascular repertoire and morphology matching mature endothelial cells (ECs). Brief TGFß-inhibition functionalizes VEGFR2 signaling, augmenting specification of ACs into rAC-VECs. Genome-wide transcriptional analyses showed that rAC-VECs are similar to adult ECs in which vascular-specific genes are expressed and nonvascular genes are silenced. Functionally, rAC-VECs form stable vasculature in Matrigel plugs and regenerating livers. Therefore, short-term ETV2 expression and TGFß inhibition with constitutive ERG1/FLI1 coexpression reprogram mature ACs into durable rAC-VECs with clinical-scale expansion potential. Banking of HLA-typed rAC-VECs establishes a vascular inventory for treatment of diverse disorders.


Subject(s)
Amniotic Fluid/cytology , Cell Differentiation , Endothelial Cells/cytology , Proto-Oncogene Proteins c-ets/metabolism , Retroviridae Proteins, Oncogenic/metabolism , Transforming Growth Factor beta/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans
9.
Dev Biol ; 361(2): 439-49, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22056786

ABSTRACT

The developing heart contains an inner tube of specialized endothelium known as endocardium, which performs multiple essential functions. In spite of the essential role of the endocardium in heart development and function, the transcriptional pathways that regulate its development remain largely undefined. GATA4 is a zinc finger transcription factor that is expressed in multiple cardiovascular lineages and is required for endocardial cushion development and embryonic viability, but the transcriptional pathways upstream of Gata4 in the endocardium and its derivatives in the endocardial cushions are unknown. Here, we describe a distal enhancer from the mouse Gata4 gene that is briefly active in multiple cardiac lineages early in cardiac development but restricts to the endocardium where it remains active through cardiogenesis. The activity of this Gata4 cardiac enhancer in transgenic embryos and in cultured aortic endothelial cells is dependent on four ETS sites. To identify which ETS transcription factors might be involved in Gata4 regulation via the ETS sites in the enhancer, we determined the expression profile of 24 distinct ETS factors in embryonic mouse hearts. Among multiple ETS transcripts present, ETS1, FLI1, ETV1, ETV5, ERG, and ETV6 were the most abundant in the early embryonic heart. We found that ETS1, FLI1, and ERG were strongly expressed in the heart at embryonic day 8.5 and that ETS1 and ERG bound to the endogenous Gata4 enhancer in cultured endothelial cells. Thus, these studies define the ETS expression profile in the early embryonic heart and identify an ETS-dependent enhancer from the Gata4 locus.


Subject(s)
Enhancer Elements, Genetic , GATA4 Transcription Factor/genetics , Heart/embryology , Proto-Oncogene Proteins c-ets/metabolism , Animals , Base Pairing/genetics , Base Sequence , Binding Sites , Cattle , Conserved Sequence/genetics , Endocardium/cytology , Endocardium/embryology , Endocardium/metabolism , GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Genetic Loci/genetics , Mice , Molecular Sequence Data , Myocardium/cytology , Myocardium/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets/genetics , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors , Transcriptional Regulator ERG , Transgenes/genetics
10.
J Clin Invest ; 121(7): 2668-78, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21633168

ABSTRACT

Numerous studies have suggested a link between the angiogenic FGF and VEGF signaling pathways; however, the nature of this link has not been established. To evaluate this relationship, we investigated VEGF signaling in ECs with disrupted FGF signaling in vitro and in vivo. ECs lacking FGF signaling became unresponsive to VEGF, caused by downregulation of VEGF receptor 2 (VEGFR2) expression after reduced Vegfr2 enhancer activation. FGF mediated VEGFR2 expression via activation of Erk1/2. Transcriptional analysis revealed that Ets transcription factors controlled VEGFR2 expression in an FGF- and Erk1/2-dependent manner. Mice with defective FGF signaling exhibited loss of vascular integrity and reduced vascular morphogenesis. Thus, basal FGF stimulation of the endothelium is required for maintenance of VEGFR2 expression and the ability to respond to VEGF stimulation and accounts for the hierarchic control of vascular formation by FGFs and VEGF.


Subject(s)
Fibroblast Growth Factors/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factors/metabolism , Animals , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/physiology , Enzyme Activation , Fibroblast Growth Factors/genetics , Hindlimb/blood supply , Hindlimb/pathology , Humans , Ischemia/metabolism , Ischemia/pathology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neovascularization, Physiologic/physiology , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-2/genetics
11.
Dev Biol ; 346(2): 346-55, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20692247

ABSTRACT

The embryonic endoderm is a multipotent progenitor cell population that gives rise to the epithelia of the digestive and respiratory tracts, the liver and the pancreas. Among the transcription factors that have been shown to be important for endoderm development and gut morphogenesis is GATA4. Despite the important role of GATA4 in endoderm development, its transcriptional regulation is not well understood. In this study, we identified an intronic enhancer from the mouse Gata4 gene that directs expression to the definitive endoderm in the early embryo. The activity of this enhancer is initially broad in all endodermal progenitors, as demonstrated by fate mapping analysis using the Cre/loxP system, but becomes restricted to the dorsal foregut and midgut, and associated organs such as dorsal pancreas and stomach. The function of the intronic Gata4 enhancer is dependent upon a conserved Forkhead transcription factor-binding site, which is bound by recombinant FoxA2 in vitro. These studies identify Gata4 as a direct transcriptional target of FoxA2 in the hierarchy of the transcriptional regulatory network that controls the development of the definitive endoderm.


Subject(s)
Endoderm/embryology , Enhancer Elements, Genetic/genetics , GATA4 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 3-beta/metabolism , Introns/genetics , Animals , Base Sequence , Binding Sites , Embryo, Mammalian/metabolism , GATA4 Transcription Factor/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Mice , Mice, Transgenic , Molecular Sequence Data
12.
Dev Dyn ; 238(10): 2588-98, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19777593

ABSTRACT

Several transcription factors function in the specification and differentiation of the endoderm, including the zinc finger transcription factor GATA4. Despite its essential role in endoderm development, the transcriptional control of the Gata4 gene in the developing endoderm and its derivatives remains incompletely understood. Here, we identify a distal enhancer from the Gata4 gene, which directs expression exclusively to the visceral and definitive endoderm of transgenic mouse embryos. The activity of this enhancer is initially broad within the definitive endoderm but later restricts to developing endoderm-derived tissues, including pancreas, glandular stomach, and duodenum. The activity of this enhancer in vivo is dependent on evolutionarily-conserved HOX- and GATA-binding sites, which are bound by PDX-1 and GATA4, respectively. These studies establish Gata4 as a direct transcriptional target of homeodomain and GATA transcription factors in the endoderm and support a model in which GATA4 functions in the transcriptional network for pancreas formation.


Subject(s)
Endoderm/physiology , Enhancer Elements, Genetic , GATA Transcription Factors/metabolism , GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Transcription, Genetic , Animals , Base Sequence , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/metabolism , GATA Transcription Factors/genetics , GATA4 Transcription Factor/genetics , Homeodomain Proteins/genetics , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Opossums/anatomy & histology , Opossums/embryology , Opossums/genetics , Pancreas/cytology , Pancreas/embryology , Sequence Alignment
13.
Immunity ; 26(4): 503-17, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17398124

ABSTRACT

Natural killer (NK) cells are important effector cells in the control of infections. The cellular and molecular signals required for NK cell activation in vivo remain poorly defined. By using a mouse model for the inducible ablation of dendritic cells (DCs), we showed that the in vivo priming of NK cell responses to viral and bacterial pathogens required the presence of CD11c(high) DCs. After peripheral Toll-like receptor (TLR) stimulation, NK cells were recruited to local lymph nodes, and their interaction with DCs resulted in the emergence of effector NK cells in the periphery. NK cell priming was dependent on the recognition of type I IFN signals by DCs and the subsequent production and trans-presentation of IL-15 by DCs to resting NK cells. CD11c(high) DC-derived IL-15 was necessary and sufficient for the priming of NK cells. Our data define a unique in vivo role of DCs for the priming of NK cells, revealing a striking and previously unappreciated homology to T lymphocytes of the adaptive immune system.


Subject(s)
Antigen Presentation , Cross-Priming , Dendritic Cells/immunology , Interleukin-15/immunology , Killer Cells, Natural/immunology , Animals , CD11c Antigen/analysis , Dendritic Cells/chemistry , Interferon Type I , Listeriosis/immunology , Lymph Nodes/immunology , Macrophages/immunology , Mice , Mice, Inbred Strains , Receptors, Antigen, T-Cell , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...