Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Sci Rep ; 13(1): 9193, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37280306

ABSTRACT

The integrated stress response (ISR)-activated transcription factors ATF4 and CHOP/DDIT3 may regulate oligodendrocyte (OL) survival, tissue damage and functional impairment/recovery in white matter pathologies, including traumatic spinal cord injury (SCI). Accordingly, in OLs of OL-specific RiboTag mice, Atf4, Chop/Ddit3 and their downstream target gene transcripts were acutely upregulated at 2, but not 10, days post-contusive T9 SCI coinciding with maximal loss of spinal cord tissue. Unexpectedly, another, OL-specific upregulation of Atf4/Chop followed at 42 days post-injury. However, wild type versus OL-specific Atf4-/- or Chop-/- mice showed similar white matter sparing and OL loss at the injury epicenter, as well as unaffected hindlimb function recovery as determined by the Basso mouse scale. In contrast, the horizontal ladder test revealed persistent worsening or improvement of fine locomotor control in OL-Atf4-/- or OL-Chop-/- mice, respectively. Moreover, chronically, OL-Atf-/- mice showed decreased walking speed during plantar stepping despite greater compensatory forelimb usage. Therefore, ATF4 supports, while CHOP antagonizes, fine locomotor control during post-SCI recovery. No correlation between those effects and white matter sparing together with chronic activation of the OL ISR suggest that in OLs, ATF4 and CHOP regulate function of spinal cord circuitries that mediate fine locomotor control during post-SCI recovery.


Subject(s)
Contusions , Spinal Cord Injuries , Animals , Mice , Contusions/pathology , Oligodendroglia/pathology , Recovery of Function/physiology , Spinal Cord/pathology , Transcription Factor CHOP/genetics , Transcription Factors
2.
Asian J Androl ; 23(4): 415-420, 2021.
Article in English | MEDLINE | ID: mdl-33473011

ABSTRACT

To improve the diagnostic efficiency of prostate cancer (PCa) and reduce unnecessary biopsies, we defined and analyzed the diagnostic efficiency of peripheral zone prostate-specific antigen (PSA) density (PZ-PSAD). Patients who underwent systematic 12-core prostate biopsies in Shanghai General Hospital (Shanghai, China) between January 2012 and January 2018 were retrospectively identified (n = 529). Another group of patients with benign prostatic hyperplasia (n = 100) were randomly preselected to obtain the PSA density of the non-PCa cohort (N-PSAD). Prostate volumes and transition zone volumes were measured using multiparameter magnetic resonance imaging (mpMRI) and were combined with PSA and N-PSAD to obtain the PZ-PSAD from a specific algorithm. Receiver operating characteristic (ROC) curve analysis was used to assess the PCa detection efficiency in patients stratified by PSA level, and the area under the ROC curve (AUC) of PZ-PSAD was higher than that of PSA, PSA density (PSAD), and transition zone PSA density (TZ-PSAD). PZ-PSAD could amend the diagnosis for more than half of the patients with inaccurate transrectal ultrasonography (TRUS) and mpMRI results. When TRUS and mpMRI findings were ambiguous to predict PCa (PIRADS score ≤3), PZ-PSAD could increase the positive rate of biopsy from 21.7% to 54.7%, and help 63.8% (150/235) of patients avoid unnecessary prostate biopsy. In patients whose PSA was 4.0-10.0 ng ml-1, 10.1-20.0 ng ml-1, and >20.0 ng ml-1, the ideal PZ-PSAD cut-off value for predicting clinically significant PCa was 0.019 ng ml-2, 0.297 ng ml-2, and 1.180 ng ml-2, respectively (sensitivity >90%). Compared with PSA, PSAD, and TZ-PSAD, the efficiency of PZ-PSAD for predicting PCa is the highest, leading to fewer missed diagnoses and unnecessary biopsies.


Subject(s)
Predictive Value of Tests , Prostate-Specific Antigen/analysis , Prostatic Neoplasms/diagnosis , Aged , Aged, 80 and over , China , Cohort Studies , Humans , Male , Middle Aged , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Retrospective Studies , Sensitivity and Specificity , Statistics, Nonparametric
3.
Reprod Toxicol ; 77: 137-142, 2018 04.
Article in English | MEDLINE | ID: mdl-29526646

ABSTRACT

Numerous studies have been conducted to understand the molecular mechanisms controlling mammalian secondary palate development such as growth, reorientation and fusion. However, little is known about the signaling factors regulating palate initiation. Mouse fibroblast growth factor (FGF) receptor 2 gene (Fgfr2) is expressed on E11.5 in the palate outgrowth within the maxillary process, in a region that is responsible for palate cell specification and shelf initiation. Fgfr2 continues to express in palate on E12.5 and E13.5 in both epithelial and mesenchymal cells, and inactivation of Fgfr2 expression in mesenchymal cells using floxed Fgfr2 allele and Osr2-Cre leads to cleft palate at various stages including reorientation, horizontal growth and fusion. Notably, some mutant embryos displayed no sign of palate shelf formation suggesting that FGF receptor 2 mediated FGF signaling may play an important role in palate initiation.


Subject(s)
Palate/growth & development , Receptor, Fibroblast Growth Factor, Type 2/genetics , Animals , Cleft Palate/genetics , Female , Loss of Function Mutation , Male , Mesenchymal Stem Cells/metabolism , Mice , Palate/cytology , Palate/metabolism
4.
Breast Cancer Res Treat ; 158(2): 263-76, 2016 07.
Article in English | MEDLINE | ID: mdl-27357812

ABSTRACT

Gametogenetin-binding protein 2 (GGNBP2) is encoded in human chromosome 17q12-q23, a region known as a breast and ovarian cancer susceptibility locus. GGNBP2, also referred to ZFP403, has a single C2H2 zinc finger and a consensus LxxLL nuclear receptor-binding motif. Here, we demonstrate that GGNBP2 expression is reduced in primary human breast tumors and in breast cancer cell lines, including T47D, MCF-7, LCC9, LY2, and MDA-MB-231 compared with normal, immortalized estrogen receptor α (ERα) negative MCF-10A and MCF10F breast epithelial cells. Overexpression of GGNBP2 inhibits the proliferation of T47D and MCF-7 ERα positive breast cancer cells without affecting MCF-10A and MCF10F. Stable GGNBP2 overexpression in T47D cells inhibits 17ß-estradiol (E2)-stimulated proliferation as well as migration, invasion, anchorage-independent growth in vitro, and xenograft tumor growth in mice. We further demonstrate that GGNBP2 protein physically interacts with ERα, inhibits E2-induced activation of estrogen response element-driven reporter activity, and attenuates ER target gene expression in T47D cells. In summary, our in vitro and in vivo findings suggest that GGNBP2 is a novel breast cancer tumor suppressor functioning as a nuclear receptor corepressor to inhibit ERα activity and tumorigenesis.


Subject(s)
Breast Neoplasms/metabolism , Down-Regulation , Estrogen Receptor alpha/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Estradiol/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , Neoplasm Transplantation , Response Elements/drug effects
5.
Dev Growth Differ ; 58(6): 522-9, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27273137

ABSTRACT

During mouse gastrulation, the primitive streak is formed on the posterior side of the embryo. Cells migrate out of the primitive streak to form the future mesoderm and endoderm. Fate mapping studies revealed a group of cell migrate through the proximal end of the primitive streak and give rise to the extraembryonic mesoderm tissues such as the yolk sac blood islands and allantois. However, it is not clear whether the formation of a morphological primitive streak is required for the development of these extraembryonic mesodermal tissues. Loss of the Cripto gene in mice dramatically reduces, but does not completely abolish, Nodal activity leading to the absence of a morphological primitive streak. However, embryonic erythrocytes are still formed and assembled into the blood islands. In addition, Cripto mutant embryos form allantoic buds. However, Drap1 mutant embryos have excessive Nodal activity in the epiblast cells before gastrulation and form an expanded primitive streak, but no yolk sac blood islands or allantoic bud formation. Lefty2 embryos also have elevated levels of Nodal activity in the primitive streak during gastrulation, and undergo normal blood island and allantois formation. We therefore speculate that low level of Nodal activity disrupts the formation of morphological primitive streak on the posterior side, but still allows the formation of primitive streak cells on the proximal side, which give rise to the extraembryonic mesodermal tissues formation. Excessive Nodal activity in the epiblast at pre-gastrulation stage, but not in the primitive streak cells during gastrulation, disrupts extraembryonic mesoderm development.


Subject(s)
Mesoderm/cytology , Mesoderm/embryology , Animals , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Left-Right Determination Factors/genetics , Left-Right Determination Factors/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Mutant Strains , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nodal Protein/genetics , Nodal Protein/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
6.
Biol Reprod ; 94(2): 41, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26764350

ABSTRACT

The Ggnbp2 null mutant embryos died in utero between Embryonic Days 13.5 to 15.5 with dysmorphic placentae, characterized by excessive nonvascular cell nests consisting of proliferative trophoblastic tissue and abundant trophoblast stem cells (TSCs) in the labyrinth. Lethality of Ggnbp2 null embryos was caused by insufficient placental perfusion as a result of remarkable decreases in both fetal and maternal blood vessels in the labyrinth. These defects were accompanied by a significant elevation of c-Met expression and phosphorylation and its downstream effector Stat3 activation. Knockdown of Ggnbp2 in wild-type TSCs in vitro provoked the proliferation but delayed the differentiation with an upregulation of c-Met expression and an enhanced phosphorylation of c-Met and Stat3. In contrast, overexpression of Ggnbp2 in wild-type TSCs exhibited completely opposite effects compared to knockdown TSCs. These results suggest that loss of GGNBP2 in the placenta aberrantly overactivates c-Met-Stat3 signaling, alters TSC proliferation and differentiation, and ultimately compromises the structure of placental vascular labyrinth. Our studies for the first time demonstrate that GGNBP2 is an essential factor for pregnancy success acting through the maintenance of a balance of TSC proliferation and differentiation during placental development.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Placentation/genetics , Stem Cells/cytology , Trophoblasts/cytology , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Female , Mice , Mice, Transgenic , Phosphorylation , Pregnancy , Proto-Oncogene Proteins c-met/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Stem Cells/metabolism , Trophoblasts/metabolism
7.
Dev Biol ; 407(1): 57-67, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26278034

ABSTRACT

Organogenesis is orchestrated by cell and tissue interactions mediated by molecular signals. Identification of relevant signals, and the tissues that generate and receive them, are important goals of developmental research. Here, we demonstrate that Retinoic Acid (RA) is a critical signaling molecule important for morphogenesis of mammalian submandibular salivary glands (SMG). By examining late stage RA deficient embryos of Rdh10 mutant mice we show that SMG development requires RA in a dose-dependent manner. Additionally, we find that active RA signaling occurs in SMG tissues, arising earlier than any other known marker of SMG development and persisting throughout gland morphogenesis. At the initial bud stage of development, we find RA production occurs in SMG mesenchyme, while RA signaling occurs in epithelium. We also demonstrate active RA signaling occurs in glands cultured ex vivo, and treatment with an inhibitor of RA signaling blocks growth and branching. Together these data identify RA signaling as a direct regulator of SMG organogenesis.


Subject(s)
Embryonic Development/drug effects , Submandibular Gland/embryology , Tretinoin/pharmacology , Alcohol Oxidoreductases/physiology , Animals , Mesoderm/metabolism , Mice , Morphogenesis , Signal Transduction , Tretinoin/metabolism , Vitamin A/metabolism
8.
Microrna ; 4(1): 64-71, 2015.
Article in English | MEDLINE | ID: mdl-26159804

ABSTRACT

Clefting of the secondary palate is the most common birth defect in humans. Midline fusion of the bilateral palatal processes is thought to involve apoptosis, epithelial to mesenchymal transition, and cell migration of the medial edge epithelium (MEE), the specialized cells of the palate that mediate fusion of the palatal processes during fetal development. Data presented in this manuscript are the result of analyses designed to identify microRNAs that are expressed and regulated by TGFß3 in developing palatal MEE. The expression of 7 microRNAs was downregulated and 1 upregulated in isolated MEE from wildtype murine fetuses on gestational day (GD) 13.5 to GD14.5 (prior to and during epithelial fusion of the palatal processes, respectively). Among this group were miRNAs linked to apoptosis (miR-378) and epithelial to mesenchymal transformation (miR-200b, miR-205, and miR-93). Tgfß3(-/-) fetuses, which present with a complete and isolated cleft of the secondary palate, exhibited marked dysregulation of distinct miRNAs both in the palatal MEE and mesenchyme when compared to comparable wild-type tissue. These included, among others, miRNAs known to affect apoptosis (miR-206 and miR-186). Dysregulation of miRNAs in the mesenchyme underlying the palatal MEE of Tgfß3(-/-) fetuses is also discussed in relation to epithelial-mesenchymal transformation of the MEE. These results are the first systematic analysis of the expression of microRNAs in isolated fetal palatal epithelium and mesenchyme. Moreover, analysis of the Tgfß3 knockout mouse model has enabled identification of miRNAs with altered expression that may contribute to the cleft palate phenotype.


Subject(s)
Cleft Palate/embryology , Mice/embryology , MicroRNAs/genetics , Palate/embryology , Transforming Growth Factor beta3/genetics , Animals , Cleft Palate/genetics , Epithelium/embryology , Epithelium/metabolism , Fetus/embryology , Fetus/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Laser Capture Microdissection , Mice/genetics , Mice, Knockout , Palate/metabolism
9.
J Dev Biol ; 3(1): 2-10, 2015 Mar.
Article in English | MEDLINE | ID: mdl-27134830

ABSTRACT

The effect of strain background on gene function in growth and development has been well documented. However, it has not been extensively reported whether the strain background affects the gene expression pattern. Here, we found that the expression of homeobox gene Meox-2 and FGF receptor 1 gene Fgfr1 during mouse palate development is strain-dependent. On the C57B6 inbred background, Meox-2 is expressed in the palatal outgrowth on Embryonic Day 11.5 (E11.5); the expression shifts posteriorly and is restricted to the back of palate on E14.5. On the Swiss Webster outbred background, Meox-2 expression covers both anterior and posterior regions with the same intensity from E12.5 to E14.5. On the Black Swiss background, Meox-2 expression also covers the entire palate A-P axis, but is much weaker in the anterior region on E14.5. Fgfr1 also displays distinct expression patterns in the palatal outgrowth on E11.5 in these three strains. On the Black Swiss outbred background, the expression is restricted to the anterior palatal outgrowth. In marked contrast, the expression in the Swiss Webster outbred strain is located exclusively in the posterior palate outgrowth on E11.5, whereas in the C57B6 inbred strain, the expression is undetectable in the palatal outgrowth on E11.5.

10.
Reprod Toxicol ; 50: 129-33, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25450421

ABSTRACT

Cleft palate is a common birth defect affecting 1 in 700 births. Transforming growth factor-ßs (TGF-ßs) are important signaling molecules, and their functions in murine palate development have received great attention. TGF-ß3 is expressed exclusively in palatal epithelial cells and mediates epithelial fusion, whereas the importance of TGF-ß1 and 2 in palate have not yet been demonstrated in vivo, since inactivation of Tgf-ß1 or Tgf-ß2 genes in mice did not reveal significant palate defects. We hypothesized that TGF-ß1 and TGF-ß2 can compensate each other during palate formation. To test this, we generated Tgf-ß1 and Tgf-ß2 compound mutant mice and found that approximately 40% of [Tgf-ß1(+/-); Tgf-ß2(-/-)] compound mutant embryos display cleft palate on C57 background. In addition, 26% of Tgf-ß2(-/-) embryos on 129 background, but not in C57 or Black Swiss, displayed cleft palate. TGF-ß1 and 2 functions are required for murine palate development in strain-dependent manner.


Subject(s)
Palate/embryology , Transforming Growth Factor beta1/physiology , Transforming Growth Factor beta2/physiology , Animals , Mice , Mice, Inbred C57BL , Species Specificity
11.
PLoS One ; 9(12): e115140, 2014.
Article in English | MEDLINE | ID: mdl-25514676

ABSTRACT

Tyro3, Axl and Mertk (TAM) receptor tyrosine kinases play multiple functional roles by either providing intrinsic trophic support for cell growth or regulating the expression of target genes that are important in the homeostatic regulation of immune responses. TAM receptors have been shown to regulate adult hippocampal neurogenesis by negatively regulation of glial cell activation in central nervous system (CNS). In the present study, we further demonstrated that all three TAM receptors were expressed by cultured primary neural stem cells (NSCs) and played a direct growth trophic role in NSCs proliferation, neuronal differentiation and survival. The cultured primary NSCs lacking TAM receptors exhibited slower growth, reduced proliferation and increased apoptosis as shown by decreased BrdU incorporation and increased TUNEL labeling, than those from the WT NSCs. In addition, the neuronal differentiation and maturation of the mutant NSCs were impeded, as characterized by less neuronal differentiation (ß-tubulin III+) and neurite outgrowth than their WT counterparts. To elucidate the underlying mechanism that the TAM receptors play on the differentiating NSCs, we examined the expression profile of neurotrophins and their receptors by real-time qPCR on the total RNAs from hippocampus and primary NSCs; and found that the TKO NSC showed a significant reduction in the expression of both nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), but accompanied by compensational increases in the expression of the TrkA, TrkB, TrkC and p75 receptors. These results suggest that TAM receptors support NSCs survival, proliferation and differentiation by regulating expression of neurotrophins, especially the NGF.


Subject(s)
Neural Stem Cells/metabolism , Neurogenesis/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Apoptosis/genetics , Brain-Derived Neurotrophic Factor/biosynthesis , Cell Proliferation , Cell Survival , Cells, Cultured , Hippocampus/cytology , Hippocampus/metabolism , Mice , Mice, Knockout , Nerve Growth Factors/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptor, trkA/biosynthesis , Receptor, trkB/biosynthesis , Receptor, trkC/biosynthesis , Receptors, Nerve Growth Factor/biosynthesis , Recombinant Proteins , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
12.
Dev Dyn ; 243(12): 1536-43, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25104574

ABSTRACT

BACKGROUND: Transforming growth factor-ß3 (TGF-ß3) plays a central role in mediating secondary palate fusion along the facial midline. However, the mechanisms by which TGF-ß3 functions during secondary palate fusion are still poorly understood. RESULTS: We found that mouse cytokeratin 6α and 17 mRNAs were expressed exclusively in the palate medial edge epithelium on embryonic day 14.5, and this expression was completely abolished in Tgf-ß3 mutant embryos. In contrast, we found that Jagged2 was initially expressed throughout the palate epithelium, but was specifically down-regulated in the medial edge epithelium during palatal fusion. Jagged2 down-regulation was regulated by TGF-ß3, since Jagged2 was persistently expressed in palatal medial edge epithelium in Tgf-ß3 null mutant embryos. Moreover, addition of DAPT, a specific inhibitor of Notch signaling, partially rescued the fusion defects in Tgf-ß3 null mutant palatal shelves. CONCLUSIONS: Based on these results, together with the previous study indicating that the loss of Jagged2 function promotes embryonic oral epithelial fusion, we concluded that TGF-ß3 mediates palate fusion in part by down-regulating Jagged2 expression in palatal medial edge epithelium. In addition, cytokeratin 6α and 17 are two TGF-ß3 downstream target genes in palate medial edge epithelium differentiation.


Subject(s)
Embryo, Mammalian/embryology , Mouth Mucosa/embryology , Palate/embryology , Transforming Growth Factor beta3/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line , Embryo, Mammalian/cytology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Keratin-6/biosynthesis , Keratin-6/genetics , Keratins/biosynthesis , Keratins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Mutant Strains , Palate/cytology , Serrate-Jagged Proteins , Transforming Growth Factor beta3/genetics
13.
Int J Dev Biol ; 58(9): 713-7, 2014.
Article in English | MEDLINE | ID: mdl-25896208

ABSTRACT

Recent studies have shown that mouse palatal mesenchymal cells undergo regional specification along the anterior-posterior (A-P) axis defined by anterior Shox2 and Msx1 expression and posterior Meox2 expression. A-P regional specification of the medial edge epithelium, which is directly responsible for palate fusion, has long been proposed, but it has not yet been demonstrated due to the lack of regional specific markers. In this study, we have demonstrated that the palate medial edge epithelium is regionalized along the A-P axis, similar to that for the underlying mesenchyme. Mmp13, a medial edge epithelium specific marker, was uniformly expressed from anterior to posterior in wild-type mouse palatal shelves. Previous studies demonstrated that medial edge epithelium expression of Mmp13 was regulated by TGF-beta3. We have found that the changes in Mmp13 expression in TGF-beta3 knockouts varied along the A-P axis, and can be broken down into three distinct regions. These regions correlated with regional specification of the underlying medial edge mesenchymal cells and timing of palate fusion. Mouse palate medial edge epithelium along the A-P axis can be divided into different regions according to the differential response to the loss of TGF-beta3.


Subject(s)
Embryo, Mammalian/cytology , Epithelium/embryology , Mesoderm/embryology , Mouth Mucosa/embryology , Palate/embryology , Animals , Embryo, Mammalian/metabolism , Epithelium/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization , Matrix Metalloproteinase 13/genetics , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Palate/metabolism , Transforming Growth Factor beta3/physiology
14.
Int J Dev Biol ; 57(9-10): 793-7, 2013.
Article in English | MEDLINE | ID: mdl-24307303

ABSTRACT

Vertebrate cardiac progenitor cells are initially allocated in two distinct domains, the first and second heart fields. It has been demonstrated that first heart field cells give rise to the myocardial cells in the left ventricle and part of the atria, whereas second heart field cells move into the developing heart tube and contribute to the myocardium of the outflow tract and right ventricle and the majority of atria. In this study, we have examined the expression of the mouse Cripto gene and the lineage of Cripto-expressing cells, focusing on its relationship with cardiac myocyte differentiation. The mouse Cripto gene is initially expressed at late head fold (LHF) stages in the cardiac crescent region, known as the first heart field; later in the medial region of the early heart tube, and by embryonic day 8.5, it is localized to the outflow tract. Using a Cripto-LacZ allele, we found that Cripto- expressing progeny cells contribute to the myocardium of the entire outflow tract and right ventricle, as well as to a majority of cells within the left ventricle. In contrast, no Cripto- expressing progeny cells were found in the atria or atrio-ventricular canal. Therefore, Cripto is transiently expressed in early differentiating myocardial cells of the left ventricle, right ventricle and outflow tract between LHF stages and E8.5. Cripto expression is subsequently downregulated as cells undergo further differentiation.


Subject(s)
Epidermal Growth Factor/genetics , Heart Ventricles/embryology , Membrane Glycoproteins/genetics , Myocytes, Cardiac/metabolism , Neoplasm Proteins/genetics , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cell Differentiation , Epidermal Growth Factor/biosynthesis , Gene Expression Regulation, Developmental , Heart Atria/embryology , LIM-Homeodomain Proteins/metabolism , Membrane Glycoproteins/biosynthesis , Mice , Myocytes, Cardiac/cytology , Myosin Light Chains/metabolism , Neoplasm Proteins/biosynthesis , RNA, Messenger/biosynthesis , Transcription Factors/metabolism
15.
J Immunol ; 191(12): 6165-77, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24244024

ABSTRACT

TAM tyrosine kinases play multiple functional roles, including regulation of the target genes important in homeostatic regulation of cytokine receptors or TLR-mediated signal transduction pathways. In this study, we show that TAM receptors affect adult hippocampal neurogenesis and loss of TAM receptors impairs hippocampal neurogenesis, largely attributed to exaggerated inflammatory responses by microglia characterized by increased MAPK and NF-κB activation and elevated production of proinflammatory cytokines that are detrimental to neuron stem cell proliferation and neuronal differentiation. Injection of LPS causes even more severe inhibition of BrdU incorporation in the Tyro3(-/-)Axl(-/-)Mertk(-/-) triple-knockout (TKO) brains, consistent with the LPS-elicited enhanced expression of proinflammatory mediators, for example, IL-1ß, IL-6, TNF-α, and inducible NO synthase, and this effect is antagonized by coinjection of the anti-inflammatory drug indomethacin in wild-type but not TKO brains. Conditioned medium from TKO microglia cultures inhibits neuron stem cell proliferation and neuronal differentiation. IL-6 knockout in Axl(-/-)Mertk(-/-) double-knockout mice overcomes the inflammatory inhibition of neurogenesis, suggesting that IL-6 is a major downstream neurotoxic mediator under homeostatic regulation by TAM receptors in microglia. Additionally, autonomous trophic function of the TAM receptors on the proliferating neuronal progenitors may also promote progenitor differentiation into immature neurons.


Subject(s)
Dentate Gyrus/pathology , Microglia/physiology , Neurogenesis/physiology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Astrocytes/metabolism , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cytokines/biosynthesis , Cytokines/genetics , DNA Replication , Encephalitis/immunology , Encephalitis/pathology , Gene Expression Regulation , Indomethacin/pharmacology , Interleukin-6/antagonists & inhibitors , Interleukin-6/deficiency , Interleukin-6/genetics , Interleukin-6/physiology , Lipopolysaccharides/toxicity , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , NF-kappa B/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/genetics , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Toll-Like Receptors/immunology , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
16.
Dev Biol ; 381(1): 170-8, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23747598

ABSTRACT

During mouse gastrulation, cells in the primitive streak undergo epithelial-mesenchymal transformation and the resulting mesenchymal cells migrate out laterally to form mesoderm and definitive endoderm across the entire embryonic cylinder. The mechanisms underlying mesoderm and endoderm specification, migration, and allocation are poorly understood. In this study, we focused on the function of mouse Cripto, a member of the EGF-CFC gene family that is highly expressed in the primitive streak and migrating mesoderm cells on embryonic day 6.5. Conditional inactivation of Cripto during gastrulation leads to varied defects in mesoderm and endoderm development. Mutant embryos display accumulation of mesenchymal cells around the shortened primitive streak indicating a functional requirement of Cripto during the formation of mesoderm layer in gastrulation. In addition, some mutant embryos showed poor formation and abnormal allocation of definitive endoderm cells on embryonic day 7.5. Consistently, many mutant embryos that survived to embryonic day 8.5 displayed defects in ventral closure of the gut endoderm causing cardia bifida. Detailed analyses revealed that both the Fgf8-Fgfr1 pathway and p38 MAP kinase activation are partially affected by the loss of Cripto function. These results demonstrate a critical role for Cripto during mouse gastrulation, especially in mesoderm and endoderm formation and allocation.


Subject(s)
Endoderm/metabolism , Epidermal Growth Factor/physiology , Gastrulation , Gene Expression Regulation, Developmental , Membrane Glycoproteins/physiology , Mesoderm/metabolism , Neoplasm Proteins/physiology , Alleles , Animals , Epidermal Growth Factor/genetics , Fibroblast Growth Factor 8/metabolism , Gene Expression Profiling , Membrane Glycoproteins/genetics , Mice , Mutation , Neoplasm Proteins/genetics , Primitive Streak/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Dev Dyn ; 239(7): 2110-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20549719

ABSTRACT

The formation of mammalian secondary palate requires a series of developmental events such as growth, elevation, and fusion. Despite recent advances in the field of palate development, the process of palate elevation remains poorly understood. The current consensus on palate elevation is that the distal end of the vertical palatal shelf corresponds to the medial edge of the elevated horizontal palatal shelf. We provide evidence suggesting that the prospective medial edge of the vertical palate is located toward the interior side (the side adjacent to the tongue), instead of the distal end, of the vertical palatal shelf and that the horizontal palatal axis is generated through palatal outgrowth from the side of the vertical palatal shelf rather than rotating the pre-existing vertical axis orthogonally. Because palate elevation represents a classic example of embryonic tissue re-orientation, our findings here may also shed light on the process of tissue re-orientation in general.


Subject(s)
Mesoderm/embryology , Palate/embryology , Animals , Mesoderm/cytology , Mice , Palate/cytology
18.
Cell Tissue Res ; 333(1): 29-38, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18470539

ABSTRACT

Cleft palate is a common birth defect that involves disruptions in multiple developmental steps such as growth, differentiation, elevation, and fusion. Medial edge epithelial (MEE) differentiation is essential for palate fusion. An important question is whether the MEE differentiation that occurs during fusion is induced by palate shelf contact or is programmed intrinsically by the palate shelf itself. Here, we report that the loss of Zfhx1a function in mice leads to a cleft palate phenotype that is mainly attributable to a delay in palate elevation. Zfhx1a encodes a transcription regulatory protein that modulates several signaling pathways including those activated by members of the transforming growth factor-beta (TGF-beta) superfamily. Loss of Zfhx1a function in mice leads to a complete cleft palate with 100% penetrance. Zfhx1a mutant palatal shelves display normal cell differentiation and proliferation and are able to fuse in an in vitro culture system. The only defect detected was a delay of 24-48 h in palatal shelf elevation. Using the Zfhx1a mutant as a model, we studied the relationship between MEE differentiation and palate contact/adhesion. We found that down-regulation of Jag2 expression in the MEE cells, a key differentiation event establishing palate fusion competence, was independent of palate contact/adhesion. Moreover, the expression of several key factors essential for fusion, such as TGF-beta3 and MMP13, was also down-regulated at embryonic stage 16.5 in a contact-independent manner, suggesting that differentiation of the medial edge epithelium was largely programmed through an intrinsic mechanism within the palate shelf.


Subject(s)
Epithelial Cells/cytology , Epithelium/embryology , Homeodomain Proteins/analysis , Palate/cytology , Palate/embryology , Transcription Factors/analysis , Animals , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Cleft Palate/embryology , Cleft Palate/genetics , Gene Expression Regulation, Developmental , In Situ Hybridization , Mice , Mice, Mutant Strains
19.
Int J Dev Biol ; 51(2): 167-72, 2007.
Article in English | MEDLINE | ID: mdl-17294368

ABSTRACT

Cleft palate is a common birth defect caused by disruptions in secondary palate development. Anterior-posterior (A-P) regional specification plays a critical role in palate development and fusion. Previous studies have shown that at the molecular level, the anterior palate can be defined by the expression of Shox-2 and the posterior palate by Meox-2 expression in certain mouse strains. Here, we have extended previous studies by performing a more detailed analysis of these genes during mouse palate development. We found that the expression patterns of Shox-2 and Meox-2 are dynamic during palate development. At embryonic day 12.5 (E12.5), Shox-2 expression is localized to the anterior end and its expression domain covers less than 25% of the length of the palate shelf. The Shox-2 expression domain then gradually expands towards the posterior end and ultimately occupies more than 60% of the palate shelf by E14.5. The expansion of the Shox-2 domain may involve induction of Shox2 expression in additional cells. Reciprocally, the Meox-2 expression domain at E12.5 covers a large portion of the palate shelf, a region more than 70% of the entire palate, but then regresses to the posterior 25% by E14.5. This regression is likely caused by the repression of Meox-2 expression in certain Meox2 expressing cells, rather than the cessation of cell proliferation. Therefore, certain Meox-2 positive "primitive posterior cells" are differentiated/converted into Shox-2 positive "definitive anterior cells" during A-P regional specification.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , MSX1 Transcription Factor/genetics , Palate, Soft/embryology , Animals , Cell Division , Cleft Palate/genetics , Humans , Mice , Mice, Inbred C57BL , Palate, Soft/cytology , Palate, Soft/physiology
20.
Development ; 133(17): 3341-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16887819

ABSTRACT

Malformations in secondary palate fusion will lead to cleft palate, a common human birth defect. Palate fusion involves the formation and subsequent degeneration of the medial edge epithelial seam. The cellular mechanisms underlying seam degeneration have been a major focus in the study of palatogenesis. Three mechanisms have been proposed for seam degeneration: lateral migration of medial edge epithelial cells; epithelial-mesenchymal trans-differentiation; and apoptosis of medial edge epithelial cells. However, there is still a great deal of controversy over these proposed mechanisms. In this study, we established a [Rosa26<-->C57BL/6] chimeric culture system, in which a Rosa26-originated ;blue' palatal shelf was paired with a C57BL/6-derived ;white' palatal shelf. Using this organ culture system, we observed the migration of medial edge epithelial cells to the nasal side, but not to the oral side. We also observed an anteroposterior migration of medial edge epithelial cells, which may play an important role in posterior palate fusion. To examine epithelial-mesenchymal transdifferentiation during palate fusion, we bred a cytokeratin 14-Cre transgenic line into the R26R background. In situ hybridization showed that the Cre transgene is expressed exclusively in the epithelium. However, beta-galactosidase staining gave extensive signals in the palatal mesenchymal region during and after palate fusion, demonstrating the occurrence of an epithelial-mesenchymal transdifferentiation mechanism during palate fusion. Finally, we showed that Apaf1 mutant mouse embryos are able to complete palate fusion without DNA fragmentation-mediated programmed cell death, indicating that this is not essential for palate fusion in vivo.


Subject(s)
Apoptosis/physiology , Cell Differentiation/physiology , Cell Movement/physiology , Epithelial Cells/physiology , Palate/embryology , Animals , Chimera , Epithelial Cells/cytology , In Situ Hybridization , Integrases/genetics , Keratins/genetics , Mesoderm/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Palate/cytology
SELECTION OF CITATIONS
SEARCH DETAIL