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










Publication year range
1.
Front Immunol ; 12: 667213, 2021.
Article in English | MEDLINE | ID: mdl-34084168

ABSTRACT

An inflammatory response requires leukocytes to migrate from the circulation across the vascular lining into the tissue to clear the invading pathogen. Whereas a lot of attention is focused on how leukocytes make their way through the endothelial monolayer, it is less clear how leukocytes migrate underneath the endothelium before they enter the tissue. Upon finalization of the diapedesis step, leukocytes reside in the subendothelial space and encounter endothelial focal adhesions. Using TIRF microscopy, we show that neutrophils navigate around these focal adhesions. Neutrophils recognize focal adhesions as physical obstacles and deform to get around them. Increasing the number of focal adhesions by silencing the small GTPase RhoJ slows down basolateral crawling of neutrophils. However, apical crawling and diapedesis itself are not affected by RhoJ depletion. Increasing the number of focal adhesions drastically by expressing the Rac1 GEF Tiam1 make neutrophils to avoid migrating underneath these Tiam1-expressing endothelial cells. Together, our results show that focal adhesions mark the basolateral migration path of neutrophils.


Subject(s)
Endothelial Cells/physiology , Focal Adhesions/physiology , Neutrophils/physiology , Transendothelial and Transepithelial Migration/physiology , Cell Line , Humans , Leukocytes/physiology , Umbilical Cord/pathology
2.
JACC Basic Transl Sci ; 5(12): 1187-1206, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33426376

ABSTRACT

In situ tissue engineering that uses resorbable synthetic heart valve scaffolds is an affordable and practical approach for heart valve replacement; therefore, it is attractive for clinical use. This study showed no consistent collagen organization in the predefined direction of electrospun scaffolds made from a resorbable supramolecular elastomer with random or circumferentially aligned fibers, after 12 months of implantation in sheep. These unexpected findings and the observed intervalvular variability highlight the need for a mechanistic understanding of the long-term in situ remodeling processes in large animal models to improve predictability of outcome toward robust and safe clinical application.

3.
Front Cardiovasc Med ; 5: 105, 2018.
Article in English | MEDLINE | ID: mdl-30159315

ABSTRACT

The creation of living heart valve replacements via tissue engineering is actively being pursued by many research groups. Numerous strategies have been described, aimed either at culturing autologous living valves in a bioreactor (in vitro) or inducing endogenous regeneration by the host via resorbable scaffolds (in situ). Whereas a lot of effort is being invested in the optimization of heart valve scaffold parameters and culturing conditions, the pathophysiological in vivo remodeling processes to which tissue-engineered heart valves are subjected upon implantation have been largely under-investigated. This is partly due to the unavailability of suitable immunohistochemical tools specific to sheep, which serves as the gold standard animal model in translational research on heart valve replacements. Therefore, the goal of this study was to comprise and validate a comprehensive sheep-specific panel of antibodies for the immunohistochemical analysis of tissue-engineered heart valve explants. For the selection of our panel we took inspiration from previous histopathological studies describing the morphology, extracellular matrix composition and cellular composition of native human heart valves throughout development and adult stages. Moreover, we included a range of immunological markers, which are particularly relevant to assess the host inflammatory response evoked by the implanted heart valve. The markers specifically identifying extracellular matrix components and cell phenotypes were tested on formalin-fixed paraffin-embedded sections of native sheep aortic valves. Markers for inflammation and apoptosis were tested on ovine spleen and kidney tissues. Taken together, this panel of antibodies could serve as a tool to study the spatiotemporal expression of proteins in remodeling tissue-engineered heart valves after implantation in a sheep model, thereby contributing to our understanding of the in vivo processes which ultimately determine long-term success or failure of tissue-engineered heart valves.

4.
Nat Commun ; 7: 12210, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27417273

ABSTRACT

Vascular homoeostasis, development and disease critically depend on the regulation of endothelial cell-cell junctions. Here we uncover a new role for the F-BAR protein pacsin2 in the control of VE-cadherin-based endothelial adhesion. Pacsin2 concentrates at focal adherens junctions (FAJs) that are experiencing unbalanced actomyosin-based pulling. FAJs move in response to differences in local cytoskeletal geometry and pacsin2 is recruited consistently to the trailing end of fast-moving FAJs via a mechanism that requires an intact F-BAR domain. Photoconversion, photobleaching, immunofluorescence and super-resolution microscopy reveal polarized dynamics, and organization of junctional proteins between the front of FAJs and their trailing ends. Interestingly, pacsin2 recruitment inhibits internalization of the VE-cadherin complex from FAJ trailing ends and is important for endothelial monolayer integrity. Together, these findings reveal a novel junction protective mechanism during polarized trafficking of VE-cadherin, which supports barrier maintenance within dynamic endothelial tissue.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adherens Junctions/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Actomyosin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Antigens, CD/genetics , Cadherins/genetics , Focal Adhesions/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Microscopy, Fluorescence/methods
5.
PLoS One ; 11(2): e0149020, 2016.
Article in English | MEDLINE | ID: mdl-26867221

ABSTRACT

There is limited information about age-specific structural and functional properties of human heart valves, while this information is key to the development and evaluation of living valve replacements for pediatric and adolescent patients. Here, we present an extended data set of structure-function properties of cryopreserved human pulmonary and aortic heart valves, providing age-specific information for living valve replacements. Tissue composition, morphology, mechanical properties, and maturation of leaflets from 16 pairs of structurally unaffected aortic and pulmonary valves of human donors (fetal-53 years) were analyzed. Interestingly, no major differences were observed between the aortic and pulmonary valves. Valve annulus and leaflet dimensions increase throughout life. The typical three-layered leaflet structure is present before birth, but becomes more distinct with age. After birth, cell numbers decrease rapidly, while remaining cells obtain a quiescent phenotype and reside in the ventricularis and spongiosa. With age and maturation-but more pronounced in aortic valves-the matrix shows an increasing amount of collagen and collagen cross-links and a reduction in glycosaminoglycans. These matrix changes correlate with increasing leaflet stiffness with age. Our data provide a new and comprehensive overview of the changes of structure-function properties of fetal to adult human semilunar heart valves that can be used to evaluate and optimize future therapies, such as tissue engineering of heart valves. Changing hemodynamic conditions with age can explain initial changes in matrix composition and consequent mechanical properties, but cannot explain the ongoing changes in valve dimensions and matrix composition at older age.


Subject(s)
Cryopreservation , Heart Valves/anatomy & histology , Heart Valves/embryology , Adolescent , Adult , Age Factors , Aortic Valve/anatomy & histology , Aortic Valve/embryology , Aortic Valve/pathology , Child , Child, Preschool , Cryopreservation/methods , Fetus , Glycosaminoglycans/chemistry , Heart Valves/pathology , Hemodynamics , Humans , Infant , Infant, Newborn , Microscopy, Fluorescence , Middle Aged , Phenotype , Pulmonary Valve/anatomy & histology , Pulmonary Valve/embryology , Pulmonary Valve/pathology , Stress, Mechanical , Tensile Strength , Young Adult
6.
Tissue Eng Part A ; 22(1-2): 123-32, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26466917

ABSTRACT

Synthetic polymers are widely used to fabricate porous scaffolds for the regeneration of cardiovascular tissues. To ensure mechanical integrity, a balance between the rate of scaffold absorption and tissue formation is of high importance. A higher rate of tissue formation is expected in fast-degrading materials than in slow-degrading materials. This could be a result of synthetic cells, which aim to compensate for the fast loss of mechanical integrity of the scaffold by deposition of collagen fibers. Here, we studied the effect of fast-degrading polyglycolic acid scaffolds coated with poly-4-hydroxybutyrate (PGA-P4HB) and slow-degrading poly-ɛ-caprolactone (PCL) scaffolds on amount of tissue, composition, and mechanical characteristics in time, and compared these engineered values with values for native human heart valves. Electrospun PGA-P4HB and PCL scaffolds were either kept unseeded in culture or were seeded with human vascular-derived cells. Tissue formation, extracellular matrix (ECM) composition, remaining scaffold weight, tissue-to-scaffold weight ratio, and mechanical properties were analyzed every week up to 6 weeks. Mass of unseeded PCL scaffolds remained stable during culture, whereas PGA-P4HB scaffolds degraded rapidly. When seeded with cells, both scaffold types demonstrated increasing amounts of tissue with time, which was more pronounced for PGA-P4HB-based tissues during the first 2 weeks; however, PCL-based tissues resulted in the highest amount of tissue after 6 weeks. This study is the first to provide insight into the tissue-to-scaffold weight ratio, therewith allowing for a fair comparison between engineered tissues cultured on scaffolds as well as between native heart valve tissues. Although the absolute amount of ECM components differed between the engineered tissues, the ratio between ECM components was similar after 6 weeks. PCL-based tissues maintained their shape, whereas the PGA-P4HB-based tissues deformed during culture. After 6 weeks, PCL-based engineered tissues showed amounts of cells and ECM that were comparable to the number of human native heart valve leaflets, whereas values were lower in the PGA-P4HB-based tissues. Although increasing in time, the number of collagen crosslinks were below native values in all engineered tissues. In conclusion, this study indicates that slow-degrading scaffold materials are favored over fast-degrading materials to create organized ECM-rich tissues in vitro, which keep their three-dimensional structure before implantation.


Subject(s)
Heart Valves , Polyesters/chemistry , Polyglycolic Acid/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Cells, Cultured , Humans
7.
Arterioscler Thromb Vasc Biol ; 34(9): 2059-67, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25012130

ABSTRACT

OBJECTIVE: Vascular endothelial-cadherin- and integrin-based cell adhesions are crucial for endothelial barrier function. Formation and disassembly of these adhesions controls endothelial remodeling during vascular repair, angiogenesis, and inflammation. In vitro studies indicate that vascular cytokines control adhesion through regulation of the actin cytoskeleton, but it remains unknown whether such regulation occurs in human vessels. We aimed to investigate regulation of the actin cytoskeleton and cell adhesions within the endothelium of human arteries and veins. APPROACH AND RESULTS: We used an ex vivo protocol for immunofluorescence in human vessels, allowing detailed en face microscopy of endothelial monolayers. We compared arteries and veins of the umbilical cord and mesenteric, epigastric, and breast tissues and find that the presence of central F-actin fibers distinguishes the endothelial phenotype of adult arteries from veins. F-actin in endothelium of adult veins as well as in umbilical vasculature predominantly localizes cortically at the cell boundaries. By contrast, prominent endothelial F-actin fibers in adult arteries anchor mostly to focal adhesions containing integrin-binding proteins paxillin and focal adhesion kinase and follow the orientation of the extracellular matrix protein fibronectin. Other arterial F-actin fibers end in vascular endothelial-cadherin-based endothelial focal adherens junctions. In vitro adhesion experiments on compliant substrates demonstrate that formation of focal adhesions is strongly induced by extracellular matrix rigidity, irrespective of arterial or venous origin of endothelial cells. CONCLUSIONS: Our data show that F-actin-anchored focal adhesions distinguish endothelial phenotypes of human arteries from veins. We conclude that the biomechanical properties of the vascular extracellular matrix determine this endothelial characteristic.


Subject(s)
Actins/analysis , Arteries/cytology , Endothelium, Vascular/cytology , Focal Adhesions , Veins/cytology , Actin Cytoskeleton/ultrastructure , Adult , Antigens, CD/analysis , Breast/blood supply , Cadherins/analysis , Cells, Cultured , Endothelial Cells , Epigastric Arteries/cytology , Extracellular Matrix/physiology , Extracellular Matrix Proteins/physiology , Female , Humans , Infant, Newborn , Mesenteric Arteries/cytology , Mesenteric Veins/cytology , Microscopy, Confocal , Microscopy, Fluorescence , Phenotype , Umbilical Arteries/cytology , Umbilical Veins/cytology
8.
Cell Tissue Res ; 352(3): 727-37, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23430473

ABSTRACT

Mechanical conditioning is often used to enhance collagen synthesis, remodeling and maturation and, hence, the structural and mechanical properties of engineered cardiovascular tissues. Intermittent straining, i.e., alternating periods of cyclic and static strain, has previously been shown to result in more mature tissue compared with continuous cyclic straining. Nevertheless, the underlying mechanism is unknown. We have determined the short-term effects of continuous cyclic strain and of cyclic strain followed by static strain at the gene expression level to improve insight into the mechano-regulatory mechanism of intermittent conditioning on collagen synthesis, remodeling and maturation. Tissue-engineered constructs, consisting of human vascular-derived cells seeded onto rapidly degrading PGA/P4HB scaffolds, were conditioned with 4% strain at 1 Hz for 3 h in order to study the immediate effects of cyclic strain (n=18). Next, the constructs were either subjected to ongoing cyclic strain (4% at 1 Hz; n=9) or to static strain (n=9). Expression levels of genes involved in collagen synthesis, remodeling and maturation were studied at various time points up to 24 h within each straining protocol. The results indicate that a period of static strain following cyclic strain favors collagen synthesis and remodeling, whereas ongoing cyclic strain shifts this balance toward collagen remodeling and maturation. The data suggest that, with prolonged culture, the conditioning protocol should be changed from intermittent straining to continuous cyclic straining to improve collagen maturation after its synthesis and, hence, the tissue (mechanical) properties.


Subject(s)
Cardiovascular System/metabolism , Collagen/metabolism , Extracellular Matrix/metabolism , Stress, Mechanical , Tissue Engineering/methods , Transcriptome , Gene Expression Regulation , Humans , Reproducibility of Results , Tissue Scaffolds
9.
Regen Med ; 7(1): 59-70, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22168498

ABSTRACT

AIM: Clinical application of tissue engineered heart valves requires precise control of the tissue culture process to predict tissue composition and mechanical properties prior to implantation, and to understand the variation in tissue outcome. To this end we investigated cellular phenotype and tissue properties of ovine (n = 8) and human (n = 7) tissue engineered heart valve constructs to quantify variations in tissue outcome within species, study the differences between species and determine possible indicators of tissue outcome. MATERIALS & METHODS: Tissue constructs consisted of polyglycolic acid/poly-4-hydroxybutyrate scaffolds, seeded with myofibroblasts obtained from the jugular vein (sheep) or the saphenous vein (from humans undergoing cardiac surgery) and cultured under static conditions. Prior to seeding, protein expression of α-smooth muscle actin, vimentin, nonmuscle myosin heavy chain and heat shock protein 47 were determined to identify differences at an early stage of the tissue engineering process. After 4 weeks of culture, tissue composition and mechanical properties were quantified as indicators of tissue outcome. RESULTS: After 4 weeks of tissue culture, tissue properties of all ovine constructs were comparable, while there was a larger variation in the properties of the human constructs, especially the elastic modulus and collagen content. In addition, ovine constructs differed in composition from the human constructs. An increased number of α-smooth muscle actin-positive cells before seeding was correlated with the collagen content in the engineered heart valve constructs. Moreover, tissue stiffness increased with increasing collagen content. CONCLUSION: The results suggest that the culture process of ovine tissues can be controlled, whereas the mechanical properties, and hence functionality, of tissues originating from human material are more difficult to control. On-line evaluation of tissue properties during culture or more early cellular markers to predict the properties of autologous tissues cultured for individual patients are, therefore, of utmost importance for future clinical application of autologous heart valve tissue engineering. As an example, this study shows that α-smooth muscle actin might be an indicator of tissue mechanical properties.


Subject(s)
Heart Valve Prosthesis , Tissue Engineering/methods , Animals , Biomechanical Phenomena , Cell Count , Extracellular Matrix/metabolism , Female , Humans , Male , Middle Aged , Myofibroblasts/cytology , Myofibroblasts/metabolism , Phenotype , Sheep, Domestic
10.
Tissue Eng Part C Methods ; 17(5): 607-17, 2011 May.
Article in English | MEDLINE | ID: mdl-21284560

ABSTRACT

In autologous heart valve tissue engineering, there is an ongoing search for alternatives of fetal bovine serum (FBS). Human platelet-lysate (PL) might be a promising substitute. In the present article, we aimed to examine the tissue formation, functionality, and mechanical properties of engineered three-dimensional tissue constructs cultured in PL as a substitute for FBS. Our results show that tissue constructs that were cultured in PL and FBS produce similar amounts of collagen, glycosoaminoglycans, and collagen crosslinks, and that the cellular phenotype remains unchanged. Nevertheless, mechanical testing showed that the ultimate tensile strength in PL constructs was on average approximately three times lower as compared to FBS (0.25 vs. 0.74 MPa, respectively, p<0.01), and also the elastic modulus was almost three times lower (1.33 MPa of PL constructs vs. 3.94 MPa of FBS constructs, p<0.01). Additional tests indicated that this difference might be explained by different collagen fiber architecture possibly due to increased production of matrix-degrading proteases by cells cultured in PL. In summary, our results indicate that PL is not preferred for the culture of strong heart valve tissue constructs.


Subject(s)
Blood Platelets/cytology , Cell Extracts/pharmacology , Heart Valve Prosthesis , Serum/metabolism , Tissue Culture Techniques/methods , Tissue Engineering/methods , Animals , Biomarkers/metabolism , Biomechanical Phenomena/drug effects , Cattle , Collagen/metabolism , Culture Media/pharmacology , Elastic Modulus/drug effects , Fluorescent Antibody Technique , Humans , Myofibroblasts/cytology , Myofibroblasts/drug effects , Myofibroblasts/metabolism
11.
Biomaterials ; 30(3): 344-53, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18930540

ABSTRACT

Photopolymerizable hydrogels, formed by UV-exposure of photosensitive polymers in the presence of photoinitiators, are widely used materials in tissue engineering research employed for cellular entrapment and patterning. During photopolymerization, the entrapped cells are directly exposed to polymer and photoinitiator molecules. To develop strategies that prevent potential photoexposure-damage to osteoprogenitor cells, it is important to further characterize the effects of photopolymerization on the exposed cells. In this study we analyzed the viability, proliferation and osteogenic differentiation of multipotent stromal cell (MSC) monolayers after exposure to UV-light in the presence of Irgacure 2959, a frequently used photoinitiator in tissue engineering research. Cell cycle progression, apoptosis and osteogenic differentiation of encapsulated goat MSCs were studied in photopolymerized methacrylate-derivatized hyaluronic acid hydrogel and methacrylated hyperbranched polyglycerol gel. We demonstrate adverse effects of photopolymerization on viability, proliferation and reentry into the cell cycle of the exposed cells in monolayers, whereas the MSCs retain the ability to differentiate towards the osteogenic lineage. We further show that upon encapsulation in photopolymerizable hydrogels the viability of the embedded cells is unaffected by the photopolymerization conditions, while osteogenic differentiation depends on the type of hydrogel used.


Subject(s)
Hydrogels/metabolism , Polymers/metabolism , Stem Cells/metabolism , Stem Cells/radiation effects , Ultraviolet Rays , Alkaline Phosphatase/metabolism , Animals , Apoptosis/radiation effects , Caspase 3/metabolism , Cell Count , Cell Cycle/radiation effects , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Cellular Senescence/radiation effects , Colony-Forming Units Assay , DNA Damage , Goats , Hyaluronic Acid/chemistry , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/metabolism , Osteogenesis/radiation effects , Stem Cells/cytology , Stem Cells/enzymology , beta-Galactosidase/metabolism
12.
Biomacromolecules ; 9(3): 919-26, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18288801

ABSTRACT

In situ forming hydrogels based on thermosensitive polymers have attractive properties for tissue engineering. However, the physical interactions in these hydrogels are not strong enough to yield gels with sufficient stability for many of the proposed applications. In this study, additional covalent cross-links were introduced by photopolymerization to improve the mechanical properties and the stability of thermosensitive hydrogels. Methacrylate groups were coupled to the side chains of triblock copolymers (ABA) with thermosensitive poly( N-(2-hydroxypropyl) methacrylamide lactate) A blocks and a hydrophilic poly(ethylene glycol) B block. These polymers exhibit lower critical solution temperature (LCST) behavior in aqueous solution and the cloud point decreased with increasing amounts of methacrylate groups. These methacrylate groups were photopolymerized above the LCST to render covalent cross-links within the hydrophobic domains. The mechanical properties of photopolymerized hydrogels were substantially improved and their stability was prolonged significantly compared to nonphotopolymerized hydrogels. Whereas non-UV-cured gels disintegrated within 2 days at physiological pH and temperature, the photopolymerized gels degraded in 10 to 25 days depending on the degree of cross-linking. To assess biocompatibility, goat mesenchymal stem cells were seeded on the hydrogel surface or encapsulated within the gel and they remained viable as demonstrated by a LIVE/DEAD cell viability/cytotoxicity assay. Expression of alkaline phosphatase and production of collagen I demonstrated the functionality of the mesenchymal stem cells and their ability to differentiate upon encapsulation. Due to the improved mechanical properties, stability, and adequate cytocompatibility, the photopolymerized thermosensitive hydrogels can be regarded as highly potential materials for applications in tissue engineering.


Subject(s)
Biocompatible Materials/chemistry , Bone Substitutes , Hydrogels/chemistry , Tissue Engineering , Acrylamides/chemistry , Alkaline Phosphatase/metabolism , Animals , Biocompatible Materials/chemical synthesis , Biocompatible Materials/pharmacology , Cell Differentiation , Collagen Type I/metabolism , Goats , Hydrogels/chemical synthesis , Hydrogels/pharmacology , Lactates/chemistry , Materials Testing , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Photochemistry , Polyethylene Glycols/chemistry , Polymers/chemical synthesis , Polymers/chemistry , Polymers/pharmacology , Rheology , Temperature , Ultraviolet Rays
13.
PLoS Genet ; 3(12): e225, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18159945

ABSTRACT

Shp2 is a cytoplasmic protein-tyrosine phosphatase that is essential for normal development. Activating and inactivating mutations have been identified in humans to cause the related Noonan and LEOPARD syndromes, respectively. The cell biological cause of these syndromes remains to be determined. We have used the zebrafish to assess the role of Shp2 in early development. Here, we report that morpholino-mediated knockdown of Shp2 in zebrafish resulted in defects during gastrulation. Cell tracing experiments demonstrated that Shp2 knockdown induced defects in convergence and extension cell movements. In situ hybridization using a panel of markers indicated that cell fate was not affected by Shp2 knock down. The Shp2 knockdown-induced defects were rescued by active Fyn and Yes and by active RhoA. We generated mutants of Shp2 with mutations that were identified in human patients with Noonan or LEOPARD Syndrome and established that Noonan Shp2 was activated and LEOPARD Shp2 lacked catalytic protein-tyrosine phosphatase activity. Expression of Noonan or LEOPARD mutant Shp2 in zebrafish embryos induced convergence and extension cell movement defects without affecting cell fate. Moreover, these embryos displayed craniofacial and cardiac defects, reminiscent of human symptoms. Noonan and LEOPARD mutant Shp2s were not additive nor synergistic, consistent with the mutant Shp2s having activating and inactivating roles in the same signaling pathway. Our results demonstrate that Shp2 is required for normal convergence and extension cell movements during gastrulation and that Src family kinases and RhoA were downstream of Shp2. Expression of Noonan or LEOPARD Shp2 phenocopied the craniofacial and cardiac defects of human patients. The finding that defective Shp2 signaling induced cell movement defects as early as gastrulation may have implications for the monitoring and diagnosis of Noonan and LEOPARD syndrome.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Cell Differentiation , Cell Movement , Disease Models, Animal , Gastrulation , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gene Targeting , Humans , LEOPARD Syndrome/enzymology , LEOPARD Syndrome/genetics , Mutation , Noonan Syndrome/enzymology , Noonan Syndrome/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Proto-Oncogene Proteins c-fyn/physiology , Proto-Oncogene Proteins c-yes/physiology , Signal Transduction , Zebrafish/physiology , Zebrafish Proteins/physiology , rhoA GTP-Binding Protein/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...