Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Eur J Anaesthesiol ; 36(6): 411-417, 2019 06.
Article in English | MEDLINE | ID: mdl-30950904

ABSTRACT

BACKGROUND: Supraglottic airway devices are commonly used in general anaesthesia in children. The AuraGain is a newly developed disposable second-generation supraglottic airway device with an inflatable cuff and a gastric port. OBJECTIVE: To confirm our hypothesis that the oropharyngeal leak pressure (OLP) of the AuraGain would be noninferior to that of the i-gel in paediatric patients. DESIGN: A single-blinded, randomised, noninferiority, clinical trial. SETTING: Single-centre trial from January to March 2017. PATIENTS: One hundred paediatric patients (American Society of Anesthesiologists' physical status 1 to 3), aged up to 12 years old, and body weight of 5 to 30 kg requiring a supraglottic airway for elective surgery with an expected surgery time of less than 2 h under general anaesthesia. INTERVENTION: The patients were randomised to allocation to the AuraGain group or the i-gel group. The device size (1.5 to 2.5) used in each group was based on the manufacturer's recommendation. MAIN OUTCOME MEASURES: The primary outcome measure was OLP immediately after insertion, with a predefined noninferiority margin of 3 cmH2O. RESULTS: The OLP immediately after insertion was lower for the AuraGain than for the i-gel (17.1 vs. 23.0 cmH2O; mean difference: -5.9 cmH2O; 95% confidence interval: -8.5 to -3.3 cmH2O; P = 0.98 and <0.001 for noninferiority and superiority, respectively). The first-attempt success rate (AuraGain, 96% vs. i-gel, 90%; P = 0.44) was comparable between the devices. The incidence of blood staining was lower (AuraGain, 6% vs. i-gel, 0%; P = 0.012) and time to insertion was shorter (AuraGain 21.3 vs. i-gel, 17.1 s; P < 0.001) for the i-gel compared with the AuraGain. CONCLUSION: Our noninferiority hypothesis was not adopted. The OLP of the i-gel may be higher than that of the AuraGain, but this superiority hypothesis should be investigated in future trials. The i-gel demonstrated a shorter time to successful placement and lower incidence of blood staining than the AuraGain. TRIAL REGISTRATION: University Hospital Medical Information Network identifier, UMIN000024222.


Subject(s)
Anesthesia, General/instrumentation , Elective Surgical Procedures/adverse effects , Intraoperative Complications/epidemiology , Laryngeal Masks/adverse effects , Oropharynx/injuries , Anesthesia, General/adverse effects , Anesthesia, General/methods , Child, Preschool , Disposable Equipment , Equipment Design , Female , Hemorrhage/epidemiology , Hemorrhage/etiology , Humans , Incidence , Infant , Intraoperative Complications/etiology , Male , Pharyngeal Diseases/epidemiology , Pharyngeal Diseases/etiology , Pressure
2.
Paediatr Anaesth ; 25(8): 824-828, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25880338

ABSTRACT

BACKGROUND: The Alexis(®) wound retractor is composed of an inner- and outer-ring, with the inner-ring placed inside the abdominal cavity. This placement can constrict the internal organs and large vessels, especially in children, which can lead to hypotension. Our study aimed to investigate the relation between the incidence of hypotension and insertion of the wound retractor during lower abdominal surgery in pediatric patients. METHODS: We retrospectively examined 161 pediatric patients (<4 years old), who underwent lower abdominal surgery, regardless of abdominal retractor type. Hypotension was defined as a decrease in systolic blood pressure of >15% from baseline. We used logistic regression to predict hypotension using the following nine parameters: age, height, weight, American Society of Anesthesiologists physical status (ASA-PS), sex, laterality of the operation (unilateral or bilateral), presence or absence of caudal block, wound length, and type of retractor (retractors with or without an inner-ring). RESULTS: Logistic multivariate regression analysis revealed that a retractor with an inner-ring use (odds ratio 3.28, 95% CI 1.41-7.63, P = 0.006) and younger age (odds ratio 1.07 per month, 95% CI 1.02-1.12, P = 0.010) were independent risk factors associated with hypotension following lower abdominal surgery in this age group. CONCLUSIONS: Patient age and use of a wound retractor with an inner-ring are independent risk factors associated with hypotension during insertion of the abdominal retractor in lower abdominal surgery.


Subject(s)
Abdomen/surgery , Digestive System Surgical Procedures/adverse effects , Digestive System Surgical Procedures/instrumentation , Hypotension/etiology , Age Factors , Child, Preschool , Digestive System Surgical Procedures/methods , Equipment Design , Female , Humans , Infant , Male , Retrospective Studies , Risk Factors
3.
J Cell Biol ; 186(3): 393-407, 2009 Aug 10.
Article in English | MEDLINE | ID: mdl-19667129

ABSTRACT

Expression of the p16(Ink4a) tumor suppressor gene, a sensor of oncogenic stress, is up-regulated by a variety of potentially oncogenic stimuli in cultured primary cells. However, because p16(Ink4a) expression is also induced by tissue culture stress, physiological mechanisms regulating p16(Ink4a) expression remain unclear. To eliminate any potential problems arising from tissue culture-imposed stress, we used bioluminescence imaging for noninvasive and real-time analysis of p16(Ink4a) expression under various physiological conditions in living mice. In this study, we show that oncogenic insults such as ras activation provoke epigenetic derepression of p16(Ink4a) expression through reduction of DNMT1 (DNA methyl transferase 1) levels as a DNA damage response in vivo. This pathway is accelerated in the absence of p53, indicating that p53 normally holds the p16(Ink4a) response in check. These results unveil a backup tumor suppressor role for p16(Ink4a) in the event of p53 inactivation, expanding our understanding of how p16(Ink4a) expression is regulated in vivo.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Models, Animal , Gene Expression Profiling , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
4.
Antioxid Redox Signal ; 11(10): 2585-93, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19619006

ABSTRACT

Thioredoxin binding protein 2 (TBP2) plays a regulatory role in lipid metabolism and immune regulation. We previously reported the effect of TBP2 loss-of-function on lipid metabolism using TBP2 knockout (TBP2KO) mice. In this study, we employed TBP2 transgenic (TBP2TG) mice to analyze the in vivo effect of TBP2 gain-of-function. We revealed a decrease in the percentage of hepatic natural killer T (NKT) cells in TBP2KO mice and an increase in the percentage of hepatic NKT cells in TBP2TG mice. The TBP2KO mice were resistant to concanavalin A (ConA)-induced hepatitis, but they were highly susceptible to other types of hepatitis. TBP2 modulates lipid metabolism as well as NKT cell activity. Moreover, TBP2 expression was increased significantly in klotho-deficient mice, which exhibit a syndrome resembling aging human phenotypes. TBP2 may play multiple roles in lipid metabolism, innate immunity, and aging.


Subject(s)
Carrier Proteins/metabolism , Immunity, Innate/immunology , Lipid Metabolism , Liver/immunology , Natural Killer T-Cells/immunology , Thioredoxins/metabolism , Aging/physiology , Animals , Antigens, CD1d/genetics , Antigens, CD1d/immunology , Carrier Proteins/genetics , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A , Female , Glucuronidase/genetics , Glucuronidase/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Insulin/metabolism , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/immunology , Klotho Proteins , Liver/pathology , Liver/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Natural Killer T-Cells/cytology , Thioredoxins/genetics
5.
Diabetes ; 58(6): 1321-32, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19258434

ABSTRACT

OBJECTIVE: Components of insulin/IGF-1 receptor-mediated signaling pathways in pancreatic beta-cells have been implicated in the development of diabetes, in part through the regulation of beta-cell mass in vivo. Studies in vitro have shown that the protein Ras homolog enriched in brain (Rheb) plays a key role as a positive upstream regulator of the mammalian target of rapamycin complex 1 (mTORC1) pathway in integrating inputs from nutrients and growth factors for cell growth. Our objective was to investigate the role of the mTORC1 pathway in the regulation of beta-cell mass in vivo. RESEARCH DESIGN AND METHODS: We generated transgenic mice that overexpress Rheb in beta-cells. We examined the activation of the mTORC1 pathway and its effects on beta-cell mass, on glucose metabolism, and on protection against hyperglycemia. RESULTS: Immunoblots of islet extracts revealed that the phosphorylation levels of ribosomal protein S6 and eukaryotic initiation factor 4E binding protein 1, downstream effectors for mTORC1, were upregulated in transgenic beta-cells. Immunostaining of the pancreatic sections with anti-phospho-S6 antibody confirmed upregulation of the mTORC1 pathway in beta-cells in vivo. The mice showed improved glucose tolerance with higher insulin secretion. This arose from increased beta-cell mass accompanied by increased cell size. The mice also exhibited resistance to hyperglycemia induced by streptozotocin and obesity. CONCLUSIONS: Activation of the mTORC1 pathway by Rheb led to increased beta-cell mass in this mouse model without producing obvious unfavorable effects, giving a potential approach for the treatment of beta-cell failure and diabetes.


Subject(s)
Diabetes Mellitus, Experimental/prevention & control , Hyperglycemia/prevention & control , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Monomeric GTP-Binding Proteins/genetics , Neuropeptides/genetics , Transcription Factors/genetics , Animals , Cell Division , Diabetes Mellitus, Experimental/physiopathology , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/physiology , Glucose Tolerance Test , Humans , Insulin-Secreting Cells/transplantation , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Transgenic , Multiprotein Complexes , Proteins , Ras Homolog Enriched in Brain Protein , TOR Serine-Threonine Kinases , Up-Regulation
7.
Proc Natl Acad Sci U S A ; 105(18): 6720-4, 2008 May 06.
Article in English | MEDLINE | ID: mdl-18436651

ABSTRACT

Controlled proteolytic degradation of specialized junctional structures, corneodesmosomes, by epidermal proteases is an essential process for physiological desquamation of the skin. Corneodesmosin (CDSN) is an extracellular component of corneodesmosomes and, although considerable debate still exists, genetic studies have suggested that the CDSN gene in the major psoriasis-susceptibility locus (PSORS1) may be responsible for susceptibility to psoriasis, a human skin disorder characterized by excessive growth and aberrant differentiation of keratinocytes. CDSN is also expressed in the inner root sheath of hair follicles, and a heterozygous nonsense mutation of the CDSN gene in humans is associated with scalp-specific hair loss of poorly defined etiology. Here, we have investigated the pathogenetic roles of CDSN loss of function in the development of skin diseases by generating a mouse strain with targeted deletion of the Cdsn gene. Cdsn-deficient mouse skin showed detachment of the stratum corneum from the underlying granular layer and/or detachment within the upper granular layers due to the disrupted integrity of the corneodesmosomes. When grafted onto immunodeficient mice, Cdsn-deficient skin showed rapid hair loss together with epidermal abnormalities resembling psoriasis. These results underscore the essential roles of CDSN in hair physiology and suggest functional relevance of CDSN gene polymorphisms to psoriasis susceptibility.


Subject(s)
Gene Deletion , Gene Targeting , Glycoproteins/genetics , Hair/physiology , Skin Physiological Phenomena/genetics , Animals , Desmosomes/metabolism , Desmosomes/pathology , Glycoproteins/deficiency , Hair/growth & development , Mice , Phenotype , Psoriasis/pathology , Skin Abnormalities/genetics , Skin Abnormalities/ultrastructure , Skin Transplantation
8.
Proc Natl Acad Sci U S A ; 104(37): 14789-94, 2007 Sep 11.
Article in English | MEDLINE | ID: mdl-17766434

ABSTRACT

Tamalin is a scaffold protein that interacts with metabotropic glutamate receptors and the kinase-deficient neurotrophin TrkCT1 receptor and forms a protein complex with multiple protein-trafficking and intracellular signaling molecules. In culture, tamalin promotes intracellular trafficking of group 1 metabotropic glutamate receptors through its interaction with guanine nucleotide exchange factor cytohesins and causes actin reorganization and membrane ruffling via the TrkCT1/cytohesin-2 signaling mechanism. However, how tamalin serves its physiological function in vivo has remained elusive. In this study, we generated tamalin knockout (Tam(-/-) KO) mice and investigated behavioral alterations resulting from their deficiency in functional tamalin. Targeted deletion of functional tamalin altered neither the overall brain architecture nor the general behavior of the mice under ordinary conditions. However, Tam(-/-) KO mice showed a decrease in sensitivity to acute morphine-induced hyperlocomotion and morphine analgesic effects in the hot-plate test. Furthermore, tamalin deficiency impaired the ability of the animals to show conditioned place preference after repeated morphine administration and to display locomotor sensitization by chronic cocaine treatment. Upon in vivo microdialysis analysis of the nucleus accumbens, Tam(-/-) KO and wild-type mice showed no genotypic differences in their response patterns of extracellular dopamine and glutamate before or after morphine administration. These results demonstrate that the tamalin scaffold protein plays a unique role in both acute and adaptive behavioral responses to morphine and cocaine and could regulate common neural substrates implicated in drugs of abuse.


Subject(s)
Analgesia/methods , Behavior, Addictive/metabolism , Carrier Proteins/genetics , Cocaine/pharmacology , Locomotion/drug effects , Morphine/pharmacology , Animals , Cocaine/administration & dosage , Crosses, Genetic , Dopamine/metabolism , Dose-Response Relationship, Drug , Glutamic Acid/metabolism , Heterozygote , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Locomotion/physiology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/administration & dosage , Nucleus Accumbens/drug effects
9.
Proc Natl Acad Sci U S A ; 104(38): 15034-9, 2007 Sep 18.
Article in English | MEDLINE | ID: mdl-17848507

ABSTRACT

Although the role of p21(Waf1/Cip1) gene expression is well documented in various cell culture studies, its in vivo roles are poorly understood. To gain further insight into the role of p21(Waf1/Cip1) gene expression in vivo, we attempted to visualize the dynamics of p21(Waf1/Cip1) gene expression in living animals. In this study, we established a transgenic mice line (p21-p-luc) expressing the firefly luciferase under the control of the p21(Waf1/Cip1) gene promoter. In conjunction with a noninvasive bioluminescent imaging technique, p21-p-luc mice enabled us to monitor the endogenous p21(Waf1/Cip1) gene expression in vivo. By monitoring and quantifying the p21(Waf1/Cip1) gene expression repeatedly in the same mouse throughout its entire lifespan, we were able to unveil the dynamics of p21(Waf1/Cip1) gene expression in the aging process. We also applied this system to chemically induced skin carcinogenesis and found that the levels of p21(Waf1/Cip1) gene expression rise dramatically in benign skin papillomas, suggesting that p21(Waf1/Cip1) plays a preventative role(s) in skin tumor formation. Surprisingly, moreover, we found that the level of p21(Waf1/Cip1) expression strikingly increased in the hair bulb and oscillated with a 3-week period correlating with hair follicle cycle progression. Notably, this was accompanied by the expression of p63 but not p53. This approach, together with the analysis of p21(Waf1/Cip1) knockout mice, has uncovered a novel role for the p21(Waf1/Cip1) gene in hair development. These data illustrate the unique utility of bioluminescence imaging in advancing our understanding of the timing and, hence, likely roles of specific gene expression in higher eukaryotes.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/analysis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Luminescent Measurements/methods , Animals , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Firefly Luciferin/metabolism , Fluorescent Antibody Technique , Gene Expression , Luciferases/genetics , Luciferases/metabolism , Luminescent Agents/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction
10.
J Exp Med ; 204(9): 2031-8, 2007 Sep 03.
Article in English | MEDLINE | ID: mdl-17682067

ABSTRACT

Trafficking of immune cells is controlled by directed migration of relevant cells toward chemotactic signals. Actin cytoskeleton undergoes continuous remodeling and serves as machinery for cell migration. The mDia family of formins and the Wiskott-Aldrich syndrome protein (WASP)-Arp2/3 system are two major actin nucleating-polymerizing systems in mammalian cells, with the former producing long straight actin filaments and the latter producing branched actin meshwork. Although much is known about the latter, the physiological functions of mDia proteins are unclear. We generated mice deficient in one mDia isoform, mDia1. Although mDia1(-/-) mice were born and developed without apparent abnormality, mDia1(-/-) T lymphocytes exhibited impaired trafficking to secondary lymphoid organs in vivo and showed reduced chemotaxis, little actin filament formation, and impaired polarity in response to chemotactic stimuli in vitro. Similarly, mDia1(-/-) thymocytes showed reduced chemotaxis and impaired egression from the thymus. These results suggest that mDia1 plays a distinct role in chemotaxis in T lymphocyte trafficking.


Subject(s)
Actins/metabolism , Carrier Proteins/metabolism , Cell Movement , T-Lymphocytes/cytology , Animals , Cell Count , Cell Movement/drug effects , Cell Polarity/drug effects , Chemokines/pharmacology , Formins , Immunity, Cellular/drug effects , Lymphoid Tissue/cytology , Lymphoid Tissue/drug effects , Lymphoid Tissue/immunology , Mice , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
11.
Dev Cell ; 13(2): 298-304, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17681139

ABSTRACT

Periodic formation of somites is controlled by the segmentation clock, where the oscillator Hes7 regulates cyclic expression of the Notch modulator Lunatic fringe. Here, we show that Hes7 also regulates cyclic expression of the Fgf signaling inhibitor Dusp4 and links Notch and Fgf oscillations in phase. Strikingly, inactivation of Notch signaling abolishes the propagation but allows the initiation of Hes7 oscillation. By contrast, transient inactivation of Fgf signaling abolishes the initiation, whereas sustained inactivation abolishes both the initiation and propagation of Hes7 oscillation. We thus propose that Hes7 oscillation is initiated by Fgf signaling and propagated/maintained anteriorly by Notch signaling.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Biological Clocks , Cleavage Stage, Ovum/metabolism , Fibroblast Growth Factors/metabolism , Receptors, Notch/metabolism , Signal Transduction , Somites/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Biological Clocks/drug effects , Cleavage Stage, Ovum/drug effects , Fibroblast Growth Factors/antagonists & inhibitors , Gene Expression Regulation, Developmental/drug effects , Mice , Mice, Knockout , Models, Biological , Mutation/genetics , Pyrroles/pharmacology , Receptors, Notch/antagonists & inhibitors , Signal Transduction/drug effects , Somites/drug effects , Triglycerides/pharmacology , gamma-Aminobutyric Acid/analogs & derivatives , gamma-Aminobutyric Acid/pharmacology
12.
EMBO J ; 26(6): 1649-59, 2007 Mar 21.
Article in English | MEDLINE | ID: mdl-17332747

ABSTRACT

The mouse homeobox gene Otx2 plays essential roles at each step and in every tissue during head development. We have previously identified a series of enhancers that are responsible for driving the Otx2 expression in these contexts. Among them the AN enhancer, existing 92 kb 5' upstream, directs Otx2 expression in anterior neuroectoderm (AN) at the headfold stage. Analysis of the enhancer mutant Otx2(DeltaAN/-) indicated that Otx2 expression under the control of this enhancer is essential to the development of AN. This study demonstrates that the AN enhancer is promoter-dependent and regulated by acetylated YY1. YY1 binds to both the AN enhancer and promoter region. YY1 is acetylated in the anterior head, and only acetylated YY1 can bind to the sequence in the enhancer. Moreover, YY1 binding to both of these two sites is essential to Otx2 expression in AN. These YY1 binding sites are highly conserved in AN enhancers in tetrapods, coelacanth and skate, suggesting that establishment of the YY1 regulation coincides with that of OTX2 function in AN development in an ancestral gnathostome.


Subject(s)
Brain/embryology , Ectoderm/metabolism , Gene Expression Regulation, Developmental/physiology , Otx Transcription Factors/metabolism , YY1 Transcription Factor/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Developmental/genetics , Luciferases , Mice , Mice, Transgenic , Molecular Sequence Data , NIH 3T3 Cells , Promoter Regions, Genetic/genetics , Sequence Homology , YY1 Transcription Factor/genetics
13.
Nihon Rinsho ; 65(1): 57-62, 2007 Jan.
Article in Japanese | MEDLINE | ID: mdl-17233416

ABSTRACT

Adjuvant analgesics are drugs that have primary indications other than pain but are analgesic in selected circumstances. Antidepressants, anticonvulsants, local anesthetics, and NMDA receptor antagonists are drugs used in the treatment of neuropathic cancer pain. Assessment of pain is very important in selecting appropriate adjuvant analgesics. The assessment includes visual analog scale for pain intensity, McGill Pain Questionaire for quality of pain, and the location of pain. It is also important to assess the effectiveness of immediate release opioids. Most neuropathic pain is thought to be refractory to opioids. In Japan, effective adjuvant analgesics such as gabapentin and pregabalin are not available. The main adjuvant analgesics are still tricyclic antidepressants such as amytriptylin and amoxapin, and anticonvulsants such as carbamazepin and clonazepam. Another problem is that morphine is the only rescue drug available for the assessment of opioid responsiveness since morphine is the only opioid with an immediately release form among the strong opioids available in Japan which are morphine, oxycodone, and fentanyl. Adjuvant analgesics also have side effects such as constipation and sleepiness, which may augment the side effects of morphine and may impair the QOL of cancer patients with neuropathic pain. There is a need to improve the systems of development and importation of adjuvant analgesics.


Subject(s)
Anticonvulsants/administration & dosage , Antidepressive Agents, Tricyclic/administration & dosage , Neoplasms/physiopathology , Pain/drug therapy , Humans , Japan , Pain/diagnosis
14.
Proc Natl Acad Sci U S A ; 103(51): 19350-5, 2006 Dec 19.
Article in English | MEDLINE | ID: mdl-17159156

ABSTRACT

Otx2 is a paired type homeobox gene that plays essential roles in each step and site of head development in vertebrates. In the mouse, Otx2 expression in the anterior neuroectoderm is regulated primarily by two distinct enhancers: anterior neuroectoderm (AN) and forebrain/midbrain (FM) enhancers at 92 kb and 75 kb 5'of the Otx2 locus, respectively. The AN enhancer has activity in the entire anterior neuroectoderm at headfold and early somite stages, whereas the FM enhancer is subsequently active in the future caudal forebrain and midbrain ectoderm. In tetrapods, both AN and FM enhancers are conserved, whereas the AN region is missing in teleosts, despite overt Otx2 expression in the anterior neuroectoderm. Here, we show that zebrafish and fugu FM regions drive expression not only in the forebrain and midbrain but also in the anterior neuroectoderm at headfold stage. The analysis of coelacanth and skate genomic Otx2 orthologues suggests that the utilization of the two enhancers, AN and FM, is an ancestral condition. In contrast, the AN enhancer has been specifically lost in the teleost lineage with a compensatory establishment of AN activity within the FM enhancer. Furthermore, the AN activity in the fish FM enhancer was established by recruiting upstream factors different from those that direct the tetrapod AN enhancer, yet zebrafish FM enhancer is active in both mouse and zebrafish anterior neuroectoderm at the headfold stage.


Subject(s)
Brain/embryology , Ectoderm/metabolism , Evolution, Molecular , Morphogenesis/genetics , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , Phylogeny , Vertebrates/embryology , Vertebrates/genetics , Animals , Base Sequence , Brain/metabolism , Cloning, Molecular , Embryo, Mammalian/metabolism , Embryo, Nonmammalian , Enhancer Elements, Genetic/genetics , In Situ Hybridization , Mice , Molecular Sequence Data , Takifugu , Vertebrates/metabolism , Zebrafish
15.
Development ; 133(20): 3993-4004, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16971467

ABSTRACT

Fez and Fez-like (Fezl) are zinc-finger genes that encode transcriptional repressors expressed in overlapping domains of the forebrain. By generating Fez;Fezl-deficient mice we found that a redundant function of Fez and Fezl is required for the formation of diencephalon subdivisions. The caudal forebrain can be divided into three transverse subdivisions: prethalamus (also called ventral thalamus), thalamus (dorsal thalamus) and pretectum. Fez;Fezl-deficient mice showed a complete loss of prethalamus and a strong reduction of the thalamus at late gestation periods. Genetic marker analyses revealed that during early diencephalon patterning in Fez;Fezl-deficient mice, the rostral diencephalon (prospective prethalamus) did not form and the caudal diencephalon (prospective thalamus and pretectum) expanded rostrally. Fez;Fezl-deficient mice also displayed defects in the formation of the zona limitans intrathalamica (ZLI), which is located on the boundary between the prethalamus and thalamus. Fez and Fezl are expressed in the region rostral to the rostral limit of Irx1 expression, which marks the prospective position of the ZLI. Transgene-mediated misexpression of Fezl or Fez caudal to the ZLI repressed the caudal diencephalon fate and affected the formation of the Shh-expressing ZLI. These data indicate that Fez and Fezl repress the caudal diencephalon fate in the rostral diencephalon, and ZLI formation probably depends on Fez/Fezl-mediated formation of diencephalon subdivisions.


Subject(s)
Body Patterning/genetics , DNA-Binding Proteins/physiology , Diencephalon/embryology , Nerve Tissue Proteins/physiology , Animals , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Diencephalon/abnormalities , Diencephalon/anatomy & histology , Enhancer Elements, Genetic , Gene Expression , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Fingers
16.
Development ; 132(21): 4719-29, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16207754

ABSTRACT

The cell population and the activity of the organizer change during the course of development. We addressed the mechanism of mouse node development via an analysis of the node/notochord enhancer (NE) of Foxa2. We first identified the core element (CE) of the enhancer, which in multimeric form drives gene expression in the node. The CE was activated in Wnt/beta-catenin-treated P19 cells with a time lag, and this activation was dependent on two separate sequence motifs within the CE. These same motifs were also required for enhancer activity in transgenic embryos. We identified the Tead family of transcription factors as binding proteins for the 3' motif. Teads and their co-factor YAP65 activated the CE in P19 cells, and binding of Tead to CE was essential for enhancer activity. Inhibition of Tead activity by repressor-modified Tead compromised NE enhancer activation and notochord development in transgenic mouse embryos. Furthermore, manipulation of Tead activity in zebrafish embryos led to altered expression of foxa2 in the embryonic shield. These results suggest that Tead activates the Foxa2 enhancer core element in the mouse node in cooperation with a second factor that binds to the 5' element, and that a similar mechanism also operates in the zebrafish shield.


Subject(s)
DNA-Binding Proteins/physiology , Enhancer Elements, Genetic , Hepatocyte Nuclear Factor 3-beta/genetics , Notochord/metabolism , Nuclear Proteins/physiology , Transcription Factors/physiology , Zebrafish Proteins/physiology , Animals , Embryo, Mammalian , Embryo, Nonmammalian , Forkhead Transcription Factors/physiology , Mice , Mice, Transgenic , Organizers, Embryonic/growth & development , TEA Domain Transcription Factors , Wnt Proteins/physiology , Zebrafish
17.
Development ; 132(11): 2535-46, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15857913

ABSTRACT

The transcriptional activity of LIM-homeodomain (LIM-HD) proteins is regulated by their interactions with various factors that bind to the LIM domain. We show that reduced expression of single-stranded DNA-binding protein 1 (Ssdp1), which encodes a co-factor of LIM domain interacting protein 1 (Ldb1), in the mouse mutant headshrinker (hsk) disrupts anterior head development by partially mimicking Lim1 mutants. Although the anterior visceral endoderm and the anterior definitive endoderm, which together comprise the head organizer, were able to form normally in Ssdp1(hsk/hsk) mutants, development of the prechordal plate was compromised. Head development is partially initiated in Ssdp1(hsk/hsk) mutants, but neuroectoderm tissue anterior to the midbrain-hindbrain boundary is lost, without a concomitant increase in apoptosis. Cell proliferation is globally reduced in Ssdp1(hsk/hsk) mutants, and approximately half also exhibit smaller body size, similar to the phenotype observed in Lim1 and Ldb1 mutants. We also show that Ssdp1 contains an activation domain and is able to enhance transcriptional activation through a Lim1-Ldb1 complex in transfected cells, and that Ssdp1 interacts genetically with Lim1 and Ldb1 in both head development and body growth. These results suggest that Ssdp1 regulates the development of late head organizer tissues and body growth by functioning as an essential activator component of a Lim1 complex through interaction with Ldb1.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Head/embryology , Mice/embryology , Morphogenesis , Multiprotein Complexes/metabolism , Animals , Blotting, Northern , Cell Proliferation , DNA Primers , DNA-Binding Proteins/genetics , Genotype , Homeodomain Proteins/metabolism , In Situ Hybridization , In Situ Hybridization, Fluorescence , In Situ Nick-End Labeling , LIM Domain Proteins , LIM-Homeodomain Proteins , Mice, Transgenic , Mutation/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors , Transgenes/genetics
18.
Dev Dyn ; 232(2): 282-92, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15614767

ABSTRACT

Sonic hedgehog (Shh) is a secreted molecule that is thought to regulate tissue growth and patterning in vertebrate embryos. Although it has been reported that Gli transcription factors mediate Shh signaling to the nucleus, little is known about developmental target genes of Gli. In the previous genetic study, we showed that Shh is required for Fgf15 expression in the diencephalon and midbrain. Here, we examined whether Fgf15 is a direct target of Shh signaling through Gli. Shh was expressed in the midline cells and Fgf15 in the medial region of the diencephalon/midbrain by the seven-somite stage. The Fgf15 expression domain coincided with that of Gli1 and overlapped with that of Gli2 at this stage. Fgf15 expression in the diencephalon/midbrain was greatly reduced in the seven-somite Shh mutant embryos. Transgenic analysis showed that the 3.6-kb 5'-flanking region of the Fgf15 gene is sufficient for induction of Fgf15 in the medial/ventral diencephalon/midbrain. Luciferase assay showed that the 3.6-kb Fgf15 enhancer/promoter was activated by Gli2. A Gli-binding site was located 1 kb upstream of the transcription start site and was required for expression in the medial/ventral diencephalon/midbrain in transgenic embryos and for activation in luciferase assay. These findings indicate that Fgf15 is directly regulated by Shh signaling through Gli proteins.


Subject(s)
Diencephalon/embryology , Fibroblast Growth Factors/biosynthesis , Gene Expression Regulation, Developmental , Mesencephalon/embryology , Oncogene Proteins/biosynthesis , Trans-Activators/metabolism , Transcription Factors/biosynthesis , Amino Acid Motifs , Animals , Base Sequence , Binding Sites , Brain/metabolism , Cell Line , Cell Nucleus/metabolism , Cloning, Molecular , Enhancer Elements, Genetic , Genes, Reporter , Glutathione Transferase/metabolism , Hedgehog Proteins , In Situ Hybridization , Kruppel-Like Transcription Factors , Luciferases/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Mutagenesis , Mutation , Nucleic Acid Hybridization , Plasmids/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcription, Genetic , Transgenes , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2 , beta-Galactosidase/metabolism
19.
Development ; 131(14): 3307-17, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15201223

ABSTRACT

We have identified cis-regulatory sequences acting on Otx2 expression in epiblast (EP) and anterior neuroectoderm (AN) at about 90 kb 5' upstream. The activity of the EP enhancer is found in the inner cell mass at E3.5 and the entire epiblast at E5.5. The AN enhancer activity is detected initially at E7.0 and ceases by E8.5; it is found later in the dorsomedial aspect of the telencephalon at E10.5. The EP enhancer includes multiple required domains over 2.3 kb, and the AN enhancer is an essential component of the EP enhancer. Mutants lacking the AN enhancer have demonstrated that these cis-sequences indeed regulate Otx2 expression in EP and AN. At the same time, our analysis indicates that another EP and AN enhancer must exist outside of the -170 kb to +120 kb range. In Otx2DeltaAN/- mutants, in which one Otx2 allele lacks the AN enhancer and the other allele is null, anteroposterior axis forms normally and anterior neuroectoderm is normally induced. Subsequently, however, forebrain and midbrain are lost, indicating that Otx2 expression under the AN enhancer functions to maintain anterior neuroectoderm once induced. Furthermore, Otx2 under the AN enhancer cooperates with Emx2 in diencephalon development. The AN enhancer region is conserved among mouse, human and Xenopus; moreover, the counterpart region in Xenopus exhibited an enhancer activity in mouse anterior neuroectoderm.


Subject(s)
Brain/embryology , Ectoderm/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Mesencephalon/embryology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Prosencephalon/embryology , Trans-Activators/biosynthesis , Trans-Activators/genetics , Alleles , Animals , Base Sequence , Body Patterning , Chromosomes, Artificial, Bacterial , Enhancer Elements, Genetic , Gene Deletion , Genotype , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Mutagenesis , Mutation , Otx Transcription Factors , Phenotype , Polymerase Chain Reaction , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Time Factors , Transcription Factors , Transgenes
20.
Development ; 131(14): 3319-31, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15201224

ABSTRACT

Otx2 expression in the forebrain and midbrain was found to be regulated by two distinct enhancers (FM and FM2) located at 75 kb 5' upstream and 115 kb 3' downstream. The activities of these two enhancers were absent in anterior neuroectoderm earlier than E8.0; however, at E9.5 their regions of activity spanned the entire mesencephalon and diencephalon with their caudal limits at the boundary with the metencephalon or isthmus. In telencephalon, activities were found only in the dorsomedial aspect. Potential binding sites of OTX and TCF were essential to FM activity, and TCF sites were also essential to FM2 activity. The FM2 enhancer appears to be unique to rodent; however, the FM enhancer region is deeply conserved in gnathostomes. Studies of mutants lacking FM or FM2 enhancer demonstrated that these enhancers indeed regulate Otx2 expression in forebrain and midbrain. Development of mesencephalic and diencephalic regions was differentially regulated in a dose-dependent manner by the cooperation between Otx1 and Otx2 under FM and FM2 enhancers: the more caudal the structure the higher the OTX dose requirement. At E10.5 Otx1-/-Otx2DeltaFM/DeltaFM mutants, in which Otx2 expression under the FM2 enhancer remained, exhibited almost complete loss of the entire diencephalon and mesencephalon; the telencephalon did, however, develop.


Subject(s)
Homeodomain Proteins , Mesencephalon/embryology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Prosencephalon/embryology , Trans-Activators/biosynthesis , Trans-Activators/genetics , Alleles , Animals , Base Sequence , Binding Sites , Conserved Sequence , Diencephalon/embryology , Ectoderm/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , Metencephalon/embryology , Mice , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Mutation , Otx Transcription Factors , RNA/metabolism , Sequence Homology, Nucleic Acid , Signal Transduction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...