Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Toxicol Pathol ; 50(1): 60-117, 2022 01.
Article in English | MEDLINE | ID: mdl-34872401

ABSTRACT

The use of the mouse as a model organism is common in translational research. This mouse-human similarity holds true for placental development as well. Proper formation of the placenta is vital for development and survival of the maturing embryo. Placentation involves sequential steps with both embryonic and maternal cell lineages playing important roles. The first step in placental development is formation of the blastocyst wall (approximate embryonic days [E] 3.0-3.5). After implantation (∼E4.5), extraembryonic endoderm progressively lines the inner surface of the blastocyst wall (∼E4.5-5.0), forming the yolk sac that provides histiotrophic support to the embryo; subsequently, formation of the umbilical vessels (∼E8.5) supports transition to the chorioallantoic placenta and hemotrophic nutrition. The fully mature ("definitive") placenta is established by ∼E12.5. Abnormal placental development often leads to embryonic mortality, with the timing of death depending on when placental insufficiency takes place and which cells are involved. This comprehensive macroscopic and microscopic atlas highlights the key features of normal and abnormal mouse placental development from E4.5 to E18.5. This in-depth overview of a transient (and thus seldom-analyzed) developmental tissue should serve as a useful reference to aid researchers in identifying and describing mouse placental changes in engineered, induced, and spontaneous disease models.


Subject(s)
Placenta , Placentation , Animals , Cell Lineage , Embryo Implantation , Embryo, Mammalian , Female , Mice , Pregnancy
2.
Toxicol Pathol ; 48(6): 747-755, 2020 08.
Article in English | MEDLINE | ID: mdl-32815483

ABSTRACT

The National Toxicology Program (NTP) now uses an extended longitudinal sectioning protocol for the uterus to better evaluate female rodent reproductive tract toxicity for all developmental and reproductive toxicology and 2-year toxicity and carcinogenicity bioassays. The previous protocol for toxicity/carcinogenicity studies involved 1 cross section midway through each uterine horn and collection of uterine cervix and vagina only if gross lesions were present. Here we compare the histological findings of the original cross sections with the additional longitudinal sections of residual uterine tissues of 7 chronic NTP rat bioassays. The goal of this study was to determine whether there might be any advantages to examining additional uterine tissue. The longitudinal protocol allowed for 10 to 20 times more uterine tissue for evaluation. Results indicate that the potential advantages of a more complete evaluation of female reproductive tract tissue include increased detection of reproductive targets, increased detection of neoplastic and nonneoplastic lesions, improved detection of tissue origin of neoplasms, less reliance on gross identification of lesions, improved accuracy in the application of severity grades, and increased detection of preneoplastic or subtle lesions.


Subject(s)
Neoplasms , Reproduction , Animals , Biological Assay , Carcinogenicity Tests , Female , Histological Techniques , Rats , Uterus
3.
J Clin Invest ; 127(7): 2569-2585, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28530642

ABSTRACT

Capillary malformation-arteriovenous malformation (CM-AVM) is a blood and lymphatic vessel (LV) disorder that is caused by inherited inactivating mutations of the RASA1 gene, which encodes p120 RasGAP (RASA1), a negative regulator of the Ras small GTP-binding protein. How RASA1 mutations lead to the LV leakage defects that occur in CM-AVM is not understood. Here, we report that disruption of the Rasa1 gene in adult mice resulted in loss of LV endothelial cells (LECs) specifically from the leaflets of intraluminal valves in collecting LVs. As a result, valves were unable to prevent fluid backflow and the vessels were ineffective pumps. Furthermore, disruption of Rasa1 in midgestation resulted in LEC apoptosis in developing LV valves and consequently failed LV valvulogenesis. Similar phenotypes were observed in induced RASA1-deficient adult mice and embryos expressing a catalytically inactive RASA1R780Q mutation. Thus, RASA1 catalytic activity is essential for the function and development of LV valves. These data provide a partial explanation for LV leakage defects and potentially other LV abnormalities observed in CM-AVM.


Subject(s)
Apoptosis , Endothelial Cells/metabolism , Lymphatic Vessels/metabolism , Mutation, Missense , p120 GTPase Activating Protein/metabolism , Animals , Mice , Mice, Mutant Strains , p120 GTPase Activating Protein/genetics
4.
J Immunol ; 195(1): 31-5, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26002977

ABSTRACT

Ras GTPase-activating proteins (RasGAPs) inhibit signal transduction initiated through the Ras small GTP-binding protein. However, which members of the RasGAP family act as negative regulators of T cell responses is not completely understood. In this study, we investigated potential roles for the RasGAPs RASA1 and neurofibromin 1 (NF1) in T cells through the generation and analysis of T cell-specific RASA1 and NF1 double-deficient mice. In contrast to mice lacking either RasGAP alone in T cells, double-deficient mice developed T cell acute lymphoblastic leukemia/lymphoma, which originated at an early point in T cell development and was dependent on activating mutations in the Notch1 gene. These findings highlight RASA1 and NF1 as cotumor suppressors in the T cell lineage.


Subject(s)
Neurofibromin 1/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptor, Notch1/genetics , p120 GTPase Activating Protein/genetics , Animals , Gene Deletion , Gene Expression Regulation , Mice , Mice, Knockout , Mutation , Neurofibromin 1/deficiency , Neurofibromin 1/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/immunology , Signal Transduction , Spleen/immunology , Spleen/pathology , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thymus Gland/immunology , Thymus Gland/pathology , Time Factors , p120 GTPase Activating Protein/deficiency , p120 GTPase Activating Protein/immunology
5.
Am J Pathol ; 184(12): 3163-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25283357

ABSTRACT

Capillary malformation-arteriovenous malformation (CM-AVM) is an autosomal dominant blood vascular (BV) disorder characterized by CM and fast flow BV lesions. Inactivating mutations of the RASA1 gene are the cause of CM-AVM in most cases. RASA1 is a GTPase-activating protein that acts as a negative regulator of the Ras small GTP-binding protein. In addition, RASA1 performs Ras-independent functions in intracellular signal transduction. Whether CM-AVM results from loss of an ability of RASA1 to regulate Ras or loss of a Ras-independent function of RASA1 is unknown. To address this, we generated Rasa1 knockin mice with an R780Q point mutation that abrogates RASA1 catalytic activity specifically. Homozygous Rasa1(R780Q/R780Q) mice showed the same severe BV abnormalities as Rasa1-null mice and died midgestation. This finding indicates that BV abnormalities in CM-AVM develop as a result of loss of an ability of RASA1 to control Ras activation and not loss of a Ras-independent function of this molecule. More important, findings indicate that inhibition of Ras signaling is likely to represent an effective means of therapy for this disease.


Subject(s)
Arteriovenous Malformations/genetics , Blood Vessels/abnormalities , Capillaries/abnormalities , Port-Wine Stain/genetics , p120 GTPase Activating Protein/genetics , Alleles , Animals , Catalysis , Crosses, Genetic , DNA Mutational Analysis , Gene Knock-In Techniques , Homozygote , Immunohistochemistry , Introns , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Point Mutation , Signal Transduction
6.
Mol Immunol ; 55(3-4): 292-302, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23522726

ABSTRACT

TCR-mediated activation of the Ras signaling pathway is critical for T cell development in the thymus and function in the periphery. However, which members of a family of Ras GTPase-activating proteins (RasGAPs) negatively regulate Ras activation in T cells is unknown. In this study we examined a potential function for the neurofibromin 1 (NF1) RasGAP in the T cell lineage with the use of T cell-specific NF1-deficient mice. Surprisingly, on an MHC class I-restricted TCR transgenic background, NF1 was found to promote thymocyte positive selection. By contrast, NF1 neither promoted nor inhibited the negative selection of thymocytes. In the periphery, NF1 was found to be necessary for the maintenance of normal numbers of naïve CD4⁺ and CD8⁺ T cells but was dispensable as a regulator of TCR-induced Ras activation, cytokine synthesis, proliferation and differentiation and death. These findings point to a novel unexpected role for NF1 in T cell development as well as a regulator of T cell homeostasis.


Subject(s)
Cell Differentiation/immunology , Neurofibromin 1/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , ras Proteins/metabolism , Animals , Clonal Selection, Antigen-Mediated/immunology , Female , Lymphocyte Activation/immunology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurofibromin 1/deficiency , Neurofibromin 1/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/cytology
7.
Sci Signal ; 6(264): re1, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23443682

ABSTRACT

Inactivation of the small guanosine triphosphate-binding protein Ras during receptor signal transduction is mediated by Ras guanosine triphosphatase (GTPase)-activating proteins (RasGAPs). Ten different RasGAPs have been identified and have overlapping patterns of tissue distribution. However, genetic analyses are revealing critical nonredundant functions for each RasGAP in tissue homeostasis and as regulators of disease processes in mouse and man. Here, we discuss advances in understanding the role of RasGAPs in the maintenance of tissue integrity.


Subject(s)
Homeostasis/physiology , Models, Biological , Signal Transduction/physiology , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/physiology , ras Proteins/physiology , Animals , Cell Membrane/metabolism , Humans , Mice , Neurofibromin 1/physiology , Protein Structure, Tertiary , p120 GTPase Activating Protein/physiology
8.
J Clin Invest ; 122(2): 733-47, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22232212

ABSTRACT

RASA1 (also known as p120 RasGAP) is a Ras GTPase-activating protein that functions as a regulator of blood vessel growth in adult mice and humans. In humans, RASA1 mutations cause capillary malformation-arteriovenous malformation (CM-AVM); whether it also functions as a regulator of the lymphatic vasculature is unknown. We investigated this issue using mice in which Rasa1 could be inducibly deleted by administration of tamoxifen. Systemic loss of RASA1 resulted in a lymphatic vessel disorder characterized by extensive lymphatic vessel hyperplasia and leakage and early lethality caused by chylothorax (lymphatic fluid accumulation in the pleural cavity). Lymphatic vessel hyperplasia was a consequence of increased proliferation of lymphatic endothelial cells (LECs) and was also observed in mice in which induced deletion of Rasa1 was restricted to LECs. RASA1-deficient LECs showed evidence of constitutive activation of Ras in situ. Furthermore, in isolated RASA1-deficient LECs, activation of the Ras signaling pathway was prolonged and cellular proliferation was enhanced after ligand binding to different growth factor receptors, including VEGFR-3. Blockade of VEGFR-3 was sufficient to inhibit the development of lymphatic vessel hyperplasia after loss of RASA1 in vivo. These findings reveal a role for RASA1 as a physiological negative regulator of LEC growth that maintains the lymphatic vasculature in a quiescent functional state through its ability to inhibit Ras signal transduction initiated through LEC-expressed growth factor receptors such as VEGFR-3.


Subject(s)
Endothelial Cells/physiology , Lymphatic Diseases/pathology , Lymphatic Vessels/pathology , Lymphatic Vessels/physiology , p120 GTPase Activating Protein/metabolism , Animals , Cell Proliferation , Endothelial Cells/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Hyperplasia , Lymphangiogenesis/physiology , Lymphatic Diseases/physiopathology , Lymphatic Vessels/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism , p120 GTPase Activating Protein/genetics , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...