Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
6.
Nat Commun ; 9(1): 2704, 2018 07 13.
Article in English | MEDLINE | ID: mdl-30006544

ABSTRACT

Formation of the lymphatic system requires the coordinated expression of several key regulators: vascular endothelial growth factor C (VEGFC), its receptor FLT4, and a key transcriptional effector, PROX1. Yet, how expression of these signaling components is regulated remains poorly understood. Here, using a combination of genetic and molecular approaches, we identify the transcription factor hematopoietically expressed homeobox (HHEX) as an upstream regulator of VEGFC, FLT4, and PROX1 during angiogenic sprouting and lymphatic formation in vertebrates. By analyzing zebrafish mutants, we found that hhex is necessary for sprouting angiogenesis from the posterior cardinal vein, a process required for lymphangiogenesis. Furthermore, studies of mammalian HHEX using tissue-specific genetic deletions in mouse and knockdowns in cultured human endothelial cells reveal its highly conserved function during vascular and lymphatic development. Our findings that HHEX is essential for the regulation of the VEGFC/FLT4/PROX1 axis provide insights into the molecular regulation of lymphangiogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Lymphangiogenesis/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Base Sequence , Blood Vessels/cytology , Blood Vessels/growth & development , Blood Vessels/metabolism , Cell Line , Embryo, Mammalian , Embryo, Nonmammalian , Endothelial Cells/cytology , Endothelial Cells/metabolism , Homeodomain Proteins/metabolism , Humans , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Mice , Neovascularization, Physiologic/genetics , Repressor Proteins/deficiency , Signal Transduction , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Zebrafish , Zebrafish Proteins/deficiency , Zebrafish Proteins/metabolism
7.
Pediatrics ; 139(5)2017 May.
Article in English | MEDLINE | ID: mdl-28557748

ABSTRACT

The "7 Great Achievements in Pediatric Research" campaign noted discoveries in the past 40 years that have improved child and adult health in the United States and around the globe. This article predicts the next 7 great pediatric research advancements, including new immunizations, cancer immunotherapy, genomic discoveries, identification of early antecedents of adult health, impact of specific social-environmental influences on biology and health, quality improvement science, and implementation and dissemination research to reduce global poverty. It is an extraordinary time of new research tools that include electronic health records, technological ability to manage big data and measure "omics," and new functional and structural imaging modalities. These tools will discern mechanisms leading to health and disease with new prevention targets and cures. This article further discusses the challenges and opportunities to accelerate these exciting pediatric research discoveries to improve the lives of children and the adults they will become.


Subject(s)
Biomedical Research/trends , Pediatrics/trends , Achievement , Child , Delivery of Health Care/standards , Delivery of Health Care/trends , Genomics/trends , Global Health/trends , Humans , Immunization/trends , Immunotherapy/trends , Neoplasms/therapy , Poverty/prevention & control , Poverty/trends , Quality Improvement
8.
Genes Dev ; 30(1): 78-91, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26728554

ABSTRACT

Unlike clustered HOX genes, the role of nonclustered homeobox gene family members in hematopoiesis and leukemogenesis has not been extensively studied. Here we found that the hematopoietically expressed homeobox gene Hhex is overexpressed in acute myeloid leukemia (AML) and is essential for the initiation and propagation of MLL-ENL-induced AML but dispensable for normal myelopoiesis, indicating a specific requirement for Hhex for leukemic growth. Loss of Hhex leads to expression of the Cdkn2a-encoded tumor suppressors p16(INK4a) and p19(ARF), which are required for growth arrest and myeloid differentiation following Hhex deletion. Mechanistically, we show that Hhex binds to the Cdkn2a locus and directly interacts with the Polycomb-repressive complex 2 (PRC2) to enable H3K27me3-mediated epigenetic repression. Thus, Hhex is a potential therapeutic target that is specifically required for AML stem cells to repress tumor suppressor pathways and enable continued self-renewal.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Epigenesis, Genetic , Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/physiopathology , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Gene Deletion , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Inbred C57BL , Protein Binding , Transcription Factors/genetics
9.
PLoS One ; 11(1): e0146806, 2016.
Article in English | MEDLINE | ID: mdl-26784346

ABSTRACT

Elucidating the molecular mechanisms involved in the differentiation of stem cells to hepatic cells is critical for both understanding normal developmental processes as well as for optimizing the generation of functional hepatic cells for therapy. We performed in vitro differentiation of mouse embryonic stem cells (mESCs) with a null mutation in the homeobox gene Hhex and show that Hhex(-/-) mESCs fail to differentiate from definitive endoderm (Sox17(+/)Foxa2(+)) to hepatic endoderm (Alb(+)/Dlk(+)). In addition, hepatic culture elicited a >7-fold increase in Vegfa mRNA expression in Hhex(-/-) cells compared to Hhex(+/+) cells. Furthermore, we identified VEGFR2(+)/ALB(+/)CD34(-) in early Hhex(+/+) hepatic cultures. These cells were absent in Hhex(-/-) cultures. Finally, through manipulation of Hhex and Vegfa expression, gain and loss of expression experiments revealed that Hhex shares an inverse relationship with the activity of the Vegf signaling pathway in supporting hepatic differentiation. In summary, our results suggest that Hhex represses Vegf signaling during hepatic differentiation of mouse ESCs allowing for cell-type autonomous regulation of Vegfr2 activity independent of endothelial cells.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Hepatocytes/cytology , Homeodomain Proteins/genetics , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/metabolism , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , HMGB Proteins/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocytes/metabolism , Homeodomain Proteins/metabolism , Mice , Mutation , SOXF Transcription Factors/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
Blood ; 125(5): 803-14, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25472970

ABSTRACT

The hematopoietically expressed homeobox gene, Hhex, is a transcription factor that is important for development of definitive hematopoietic stem cells (HSCs) and B cells, and that causes T-cell leukemia when overexpressed. Here, we have used an Hhex inducible knockout mouse model to study the role of Hhex in adult hematopoiesis. We found that loss of Hhex was tolerated in HSCs and myeloid lineages, but resulted in a progressive loss of B lymphocytes in the circulation. This was accompanied by a complete loss of B-cell progenitors in the bone marrow and of transitional B-cell subsets in the spleen. In addition, transplantation and in vitro culture experiments demonstrated an almost complete failure of Hhex-null HSCs to contribute to lymphoid lineages beyond the common lymphoid precursor stage, including T cells, B cells, NK cells, and dendritic cells. Gene expression analysis of Hhex-deleted progenitors demonstrated deregulated expression of a number of cell cycle regulators. Overexpression of one of these, cyclin D1, could rescue the B-cell developmental potential of Hhex-null lymphoid precursors. Thus, Hhex is a key regulator of early lymphoid development, functioning, at least in part, via regulation of the cell cycle.


Subject(s)
Cell Cycle Proteins/genetics , Cyclin D1/genetics , Hematopoiesis/genetics , Homeodomain Proteins/genetics , Lymphopoiesis/genetics , Precursor Cells, B-Lymphoid/pathology , Transcription Factors/genetics , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cell Cycle Proteins/immunology , Cell Differentiation , Cell Proliferation , Cyclin D1/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Gene Deletion , Gene Expression Regulation , Genetic Complementation Test , Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Homeodomain Proteins/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Lymphocyte Count , Lymphocyte Depletion , Lymphopoiesis/immunology , Mice , Mice, Knockout , Precursor Cells, B-Lymphoid/immunology , Spleen/immunology , Spleen/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transcription Factors/deficiency , Transcription Factors/immunology , Transcription, Genetic
11.
Cell Mol Gastroenterol Hepatol ; 1(5): 550-569, 2015 09 01.
Article in English | MEDLINE | ID: mdl-26740970

ABSTRACT

BACKGROUND & AIMS: Perturbations in pancreatic ductal bicarbonate secretion cause chronic pancreatitis. The physiologic mechanism of ductal secretion is known, but its transcriptional control is not. We determine the role of the transcription factor hematopoietically expressed homeobox protein (Hhex) in ductal secretion and pancreatitis. METHODS: We derived mice with pancreas-specific, Cremediated Hhex gene ablation to determine the requirement of Hhex in the pancreatic duct in early life and in adult stages. Histologic and immunostaining analyses were used to detect the presence of pathology. Pancreatic primary ductal cells were isolated to discover differentially expressed transcripts upon acute Hhex ablation on a cell autonomous level. RESULTS: Hhex protein was detected throughout the embryonic and adult ductal trees. Ablation of Hhex in pancreatic progenitors resulted in postnatal ductal ectasia associated with acinar-to-ductal metaplasia, a progressive phenotype that ultimately resulted in chronic pancreatitis. Hhex ablation in adult mice, however, did not cause any detectable pathology. Ductal ectasia in young mice did not result from perturbation of expression of Hnf6, Hnf1ß, or the primary cilia genes. RNA-seq analysis of Hhex-ablated pancreatic primary ductal cells showed mRNA levels of the G-protein coupled receptor natriuretic peptide receptor 3 (Npr3), implicated in paracrine signaling, up-regulated by 4.70-fold. CONCLUSIONS: Although Hhex is dispensable for ductal cell function in the adult, ablation of Hhex in pancreatic progenitors results in pancreatitis. Our data highlight the critical role of Hhex in maintaining ductal homeostasis in early life and support ductal hypersecretion as a novel etiology of pediatric chronic pancreatitis.

12.
Genes Dev ; 28(8): 829-34, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24736842

ABSTRACT

The homeodomain transcription factor HHEX (hematopoietically expressed homeobox) has been repeatedly linked to type 2 diabetes mellitus (T2DM) using genome-wide association studies. We report here that within the adult endocrine pancreas, Hhex is selectively expressed in the somatostatin-secreting δ cell. Using two mouse models with Hhex deficiency in the endocrine pancreas, we show that Hhex is required for δ-cell differentiation. Decreased somatostatin levels in Hhex-deficient islets cause disrupted paracrine inhibition of insulin release from ß cells. These findings identify Hhex as the first transcriptional regulator specifically required for islet δ cells and suggest compromised paracrine control as a contributor to T2DM.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Homeodomain Proteins/metabolism , Somatostatin-Secreting Cells/cytology , Somatostatin-Secreting Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Homeodomain Proteins/genetics , Mice , Paracrine Communication/physiology , Transcription Factors/deficiency , Transcription Factors/genetics
13.
Pediatr Res ; 75(1-2): 120-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24192700

ABSTRACT

The developmental stages of each foregut organ are intimately linked to the development of the other foregut organs such that the ultimate function of any one foregut organ, such as the metabolic function of the liver, depends on organizational changes associated with the maturation of multiple foregut organs. These changes include: (i) proliferation of the intrahepatic bile ducts and hepatoblasts within the liver coinciding with parenchymal expansion, (ii) elongation of extrahepatic bile ducts, which allows for proper gallbladder (GB) formation, and (iii) duodenal elongation and rotation, which coincides with all of the above to connect the intrahepatic, extrahepatic, and pancreatic ductal systems with the intestine. It is well established that cross talk between endodermal and mesenchymal components of the foregut occurs, particularly regarding the vascularization of developing organs. Furthermore, genetic mutations in mesenchymal and hepatic compartments of the developing foregut result in similar foregut pathologies: hypoplastic liver, absence of GB, biliary atresia (intrahepatic and/or extrahepatic), and failure of gut elongation and rotation. Finally, these shared pathologies can be linked to deficiencies in genes specific to the septum transversum mesenchyme (Hes1, Hlx, and Foxf1) or liver (Hhex and Hnf6), illustrating the complexity of such cross talk.


Subject(s)
Digestive System/metabolism , Endoderm/metabolism , Mesoderm/metabolism , Signal Transduction , Transcription Factors/metabolism , Anemia/embryology , Anemia/genetics , Anemia/metabolism , Animals , Biliary Atresia/embryology , Biliary Atresia/genetics , Biliary Atresia/metabolism , Cell Differentiation , Cell Lineage , Cell Proliferation , Digestive System/embryology , Gene Expression Regulation, Developmental , Humans , Organogenesis , Transcription Factors/genetics
14.
J Intensive Care Med ; 29(4): 225-8, 2014.
Article in English | MEDLINE | ID: mdl-23753227

ABSTRACT

Sedation monitoring is essential in pediatric patients on ventilatory support to achieve comfort and safety. The COMFORT scale was designed and validated to assess the level of sedation in intubated pediatric patients. However, it remains unreliable in pharmacologically paralyzed patients. The SNAP II index is calculated using an algorithm that incorporates high-frequency (80-420 Hz) electroencephalogram (EEG) components, known to be useful in discriminating between awake and unconscious states, unlike other measurements that only include low-frequency EEG segments such as the bispectral index score. Previous studies suggested that the SNAP II index is a reliable and sensitive indicator of the level of consciousness in adult patients. Despite its potential, no data are currently available in the pediatric critically ill population on ventilatory support. This is the first pilot study assessing the potential application of the SNAP II index in critically ill pediatric patients by comparing it to the commonly used COMFORT scale.


Subject(s)
Consciousness Monitors , Critical Care/methods , Hypnotics and Sedatives , Monitoring, Physiologic/methods , Respiration, Artificial , Child , Child, Preschool , Electroencephalography , Female , Humans , Infant , Male , Pilot Projects , Prospective Studies
15.
J Diabetes Sci Technol ; 6(1): 48-57, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22401322

ABSTRACT

BACKGROUND: The practice of glycemic control with intravenous insulin in critically ill patients has brought clinical focus on understanding the effects of hypoglycemia, especially in children. Very little is published on the impact of hypoglycemia in this population. We aimed to review the existing literature on hypoglycemia in critically ill neonates and children. METHODS: We performed a systematic review of the literature up to August 2011 using PubMed, Ovid MEDLINE and ISI Web of Science using the search terms "hypoglycemia or hypoglyc*" and "critical care or intensive care or critical illness". Articles were limited to "all child (0-18 years old)" and "English". RESULTS: A total of 513 articles were identified and 132 were included for review. Hypoglycemia is a significant concern among pediatric and neonatal intensivists. Its definition is complicated by the use of a biochemical measure (i.e., blood glucose) for a pathophysiologic problem (i.e., neuroglycopenia). Based on associated outcomes, we suggest defining hypoglycemia as <40-45 mg/dl in neonates and <60-65 mg/dl in children. Below the suggested threshold values, hypoglycemia is associated with worse neurological outcomes, increased intensive care unit stay, and increased mortality. Disruptions in carbohydrate metabolism increase the risk of hypoglycemia incritically ill children. Prevention of hypoglycemia, especially in the setting of intravenous insulin use, will be best accomplished by the combination of accurate measuring techniques, frequent or continuous glucose monitoring, and computerized insulin titration protocols. CONCLUSION: Studies on hypoglycemia in critically ill children have focused on spontaneous hypoglycemia. With the current practice of maintaining blood glucose within a narrow range with intravenous insulin, the risk factors and outcomes associated with insulin-induced hypoglycemia should be rigorously studied to prevent hypoglycemia and potentially improve outcomes of critically ill children.


Subject(s)
Critical Illness/epidemiology , Hypoglycemia/epidemiology , Intensive Care Units, Pediatric/statistics & numerical data , Adolescent , Child , Child, Preschool , Humans , Hypoglycemia/diagnosis , Hypoglycemia/etiology , Hypoglycemia/prevention & control , Infant , Infant, Newborn , Prognosis , Risk Factors
16.
Pediatr Crit Care Med ; 11(6): 690-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20625333

ABSTRACT

OBJECTIVES: To determine the prevalence of hypoglycemia in critically ill nondiabetic children and the association of hypoglycemia with mortality and worsening organ function in critically ill children. DESIGN: Retrospective cohort study with matched-cohort analysis. SETTING: Academic pediatric intensive care unit. PATIENTS: A total of 899 nondiabetic patients <18 yrs old admitted to the pediatric intensive care unit for >1 day with at least one blood glucose measurement. Forty-two patients with a blood glucose level of <50 mg/dL (<2.8 mmol/L) were matched with 126 nonhypoglycemic patients. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Hypoglycemia, based on point-of-care blood glucose measurements, occurred in 2.2% (<40 mg/dL [<2.2 mmol/L]) to 7.5% (<60 mg/dL [<3.3 mmol/L]) of the patients. Hypoglycemia was more common in patients on mechanical ventilation and/or vasopressor support. Severity of hypoglycemia correlated with an increased mortality rate. The highest odds ratio of mortality was 4.49 (95% confidence interval [CI], 1.69-11.96; p < .01) at a blood glucose level of <40 mg/dL (<2.2 mmol/L). In the matched analysis, hypoglycemia was an independent risk factor for mortality. The unadjusted, covariate-adjusted, and propensity score-adjusted odds ratios of mortality were 3.69 (95% CI, 1.78-7.68; p < .01), 4.16 (95% CI, 1.53-11.32; p < .01), and 8.45 (95% CI, 1.75-40.86; p < .01), respectively. Hypoglycemia was associated with worsening organ function in the covariate-adjusted model (odds ratio, 2.37; 95% CI, 1.12-5.01; p = .02) but not in the unadjusted and propensity-score adjusted models. CONCLUSIONS: Hypoglycemia is common in critically ill children. It is associated with increased mortality rates in critically ill nondiabetic children. Our data suggest that hypoglycemia is also associated with worsening organ function. Hypoglycemia may merely be a marker of severity of illness. Further investigations are needed to establish the mortality risk with hypoglycemia due to insulin compared to spontaneous hypoglycemia.


Subject(s)
Critical Illness/mortality , Hypoglycemia/mortality , Blood Glucose/analysis , Chi-Square Distribution , Child , Female , Humans , Intensive Care Units, Pediatric , Male , Point-of-Care Systems , Prevalence , Regression Analysis , Respiration, Artificial , Retrospective Studies , Risk Factors , Severity of Illness Index , Statistics, Nonparametric , Vasoconstrictor Agents/administration & dosage
17.
Blood ; 116(8): 1254-62, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20472829

ABSTRACT

The development and emergence of the hematopoietic stem cell involves a series of tightly regulated molecular events that are not well characterized. The hematopoietically expressed homeobox (Hhex) gene, a member of the homeobox gene family, is an essential regulator of embryogenesis and hematopoietic progenitor development. To investigate the role of Hhex in hematopoiesis we adapted a murine embryonic stem (ES) cell coculture system, in which ES cells can differentiate into CD41(+) and CD45(+) hematopoietic progenitors in vitro. Our results show that in addition to delayed hemangioblast development, Hhex(-/-) ES-derived progeny accumulate as CD41(+) and CD41(+)c-kit(+) cells, or the earliest definitive hematopoietic progenitors. In addition, Hhex(-/-) ES-derived progeny display a significantly reduced ability to develop into mature CD45(+) hematopoietic cells. The observed reduction in hematopoietic maturation was accompanied by reduced proliferation, because Hhex(-/-) CD41(+)CD45(-)c-kit(+) hematopoietic progenitors accumulated in the G(2) phase of the cell cycle. Thus, Hhex is a critical regulator of hematopoietic development and is necessary for the maturation and proliferation of the earliest definitive hematopoietic progenitors.


Subject(s)
Embryo, Mammalian/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation/physiology , Hematopoiesis , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , Apoptosis , Blotting, Western , Cell Cycle , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Colony-Forming Units Assay , Embryo, Mammalian/metabolism , Leukocyte Common Antigens/metabolism , Mice , Mice, Knockout , Platelet Membrane Glycoprotein IIb/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism
18.
J Pediatr Endocrinol Metab ; 20(10): 1115-24, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18051930

ABSTRACT

AIM: To identify patients aged 10-30 years with probable hyperglycemic hyperosmolar syndrome (HHS), to describe demographic and clinical profiles, and to attempt to assess risk factors for poor outcomes. STUDY DESIGN: Retrospective cohort study (medical records review). SETTING: A 944-bed tertiary care teaching and research hospital and a 425-bed affiliated facility. PATIENTS: 10-30 year-old patients with a primary or secondary discharge diagnosis of HHS or diabetic ketoacidosis (DKA). Patients with a serum glucose >600 mg/dl in the absence of significant ketoacidosis (possible HHS) were profiled. Further stratification based on measured or calculated serum osmolarity >320 mOsm/kg (probable HHS) was undertaken. INTERVENTIONS: Patients received treatment for hyperglycemic crises, consisting primarily of fluids, electrolyte replacement and insulin. MEASUREMENTS AND MAIN RESULTS: Of the 629 admissions, 10 with a diagnosis of HHS and 33 with a diagnosis of DKA met the initial study criteria for HHS. 60% were African Americans and 89% were new-onset diabetics. From this group, 20 admissions had serum osmolarity > or =320 mOsm/kg. Fisher's exact test and Pearson coefficients were used to examine associations between risk factor and poor outcomes and correlations between admission data and length of hospital stay, respectively. Serious complications occurred in four patients (including two deaths, 10% mortality) and were limited to those with unreversed shock over the first 24 hours of admission and who received <40 ml/kg of intravenous fluids over the first 6 hours of treatment. CONCLUSIONS: HHS was underdiagnosed in this population and occurred disproportionately in African Americans. Serious complications occurred exclusively in those with unreversed shock and inadequate fluid resuscitation.


Subject(s)
Diabetes Mellitus, Type 2/complications , Hyperglycemia/diagnosis , Adolescent , Adult , Child , Cohort Studies , Diabetic Ketoacidosis/blood , Diabetic Ketoacidosis/diagnosis , Diabetic Ketoacidosis/etiology , Humans , Hyperglycemia/blood , Hyperglycemia/etiology , Osmolar Concentration , Population Groups , Retrospective Studies , Syndrome
19.
Dev Biol ; 308(2): 355-67, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17580084

ABSTRACT

Hhex is required for early development of the liver. A null mutation of Hhex results in a failure to form the liver bud and embryonic lethality. Therefore, Hhex null mice are not informative as to whether this gene is required during later stages of hepatobiliary morphogenesis. To address this question, we derived Hhex conditional null mice using the Cre-loxP system and two different Cre transgenics (Foxa3-Cre and Alfp-Cre). Deletion of Hhex in the hepatic diverticulum (Foxa3-Cre;Hhex(d2,3/-)) led to embryonic lethality and resulted in a small and cystic liver with loss of Hnf4alpha and Hnf6 expression in early hepatoblasts. In addition, the gall bladder was absent and the extrahepatic bile duct could not be identified. Loss of Hhex in the embryonic liver (Alfp-Cre;Hhex(d2,3/-)) caused irregular development of intrahepatic bile ducts and an absence of Hnf1beta in many (cystic) biliary epithelial cells, which resulted in a slow, progressive form of polycystic liver disease in adult mice. Thus, we have shown that Hhex is required during multiple stages of hepatobiliary development. The altered expression of Hnf4alpha, Hnf6 and Hnf1beta in Hhex conditional null mice suggests that Hhex is an essential component of the genetic networks regulating hepatoblast differentiation and intrahepatic bile duct morphogenesis.


Subject(s)
Bile Ducts/embryology , Genes, Homeobox , Hepatocytes/cytology , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , Bile Ducts/growth & development , Bile Ducts/metabolism , Bile Ducts, Extrahepatic/embryology , Bile Ducts, Extrahepatic/growth & development , Bile Ducts, Extrahepatic/metabolism , Bile Ducts, Intrahepatic/embryology , Bile Ducts, Intrahepatic/growth & development , Bile Ducts, Intrahepatic/metabolism , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 6/genetics , Hepatocytes/metabolism , Homeodomain Proteins/physiology , Liver/abnormalities , Liver/embryology , Liver/growth & development , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Biological , Transcription Factors/deficiency , Transcription Factors/physiology
20.
Biochem Biophys Res Commun ; 346(2): 535-45, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16764824

ABSTRACT

Hhex encodes a homeodomain-containing protein that functions as both a transcriptional repressor and activator, and is necessary for normal embryonic development. We previously reported that a null mutation of Hhex leads to abnormalities in vasculogenesis and have focused on identifying the transcriptional targets of Hhex necessary for vascular development. Here we report that the expression of ESM-1, a cysteine-rich protein expressed in the endothelium, is increased in Hhex(-/-) embryos. Overexpression of Hhex in endothelial cells down-regulates ESM-1. The results from transient cotransfection assay, electrophoretic-mobility shift assay, site-directed mutagenesis, and chromatin immunoprecipitation assay demonstrate that Hhex can directly bind to and repress ESM-1 via an evolutionarily conserved Hhex response element (HRE) 1. These findings indicate that ESM-1 is a direct target of Hhex and that Hhex functions as a transcriptional repressor of ESM-1. We speculate that Hhex-mediated repression of ESM-1 is critical for the normal function of the vascular endothelium and for tumor vasculogenesis.


Subject(s)
Endothelial Cells/physiology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/physiology , Neoplasm Proteins/physiology , Proteoglycans/physiology , Response Elements/genetics , Transcription Factors/physiology , Animals , Base Sequence , Cells, Cultured , Embryo, Mammalian/metabolism , Genes, Reporter , Homeodomain Proteins/genetics , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Neoplasm Proteins/genetics , Promoter Regions, Genetic , Proteoglycans/genetics , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...