Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Pain Symptom Manage ; 58(2): 344-354, 2019 08.
Article in English | MEDLINE | ID: mdl-30922704

ABSTRACT

Vietnamese Americans are a heterogeneous population with a rich, shared experience and historical and cultural influences from Asia and Europe. Societal upheaval resulting from the Vietnam War and varied immigration patterns to the U.S. and levels of acculturation layer complexity to this resilient population. These experiences influence how the communities as a whole and how the family as a unit approach health care issues, their attitudes toward serious illness and care at the end of life. Challenges with caring for this population include lack of resources and training to provide culturally sensitive care, lack of appropriate advance care planning, and lack of interpreters or culture-specific care programs. All contribute to poor end-of-life care. An understanding of how these complexities interplay may help clinicians provide compassionate and patient-centric care to these patients, their families, and their supporting communities. This article provides an overview of culturally effective care for seriously ill Vietnamese American patients and makes recommendations for potential strategies for providing respectful end-of-life care.


Subject(s)
Asian , Culturally Competent Care , Respect , Terminal Care , Advance Care Planning , Hospice Care , Humans
2.
Development ; 144(5): 866-875, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28246214

ABSTRACT

miR-1 is a small noncoding RNA molecule that modulates gene expression in heart and skeletal muscle. Loss of Drosophila miR-1 produces defects in somatic muscle and embryonic heart development, which have been partly attributed to miR-1 directly targeting Delta to decrease Notch signaling. Here, we show that overexpression of miR-1 in the fly wing can paradoxically increase Notch activity independently of its effects on Delta. Analyses of potential miR-1 targets revealed that miR-1 directly regulates the 3'UTR of the E3 ubiquitin ligase Nedd4 Analysis of embryonic and adult fly heart revealed that the Nedd4 protein regulates heart development in Drosophila Larval fly hearts overexpressing miR-1 have profound defects in actin filament organization that are partially rescued by concurrent overexpression of Nedd4. These results indicate that miR-1 and Nedd4 act together in the formation and actin-dependent patterning of the fly heart. Importantly, we have found that the biochemical and genetic relationship between miR-1 and the mammalian ortholog Nedd4-like (Nedd4l) is evolutionarily conserved in the mammalian heart, potentially indicating a role for Nedd4L in mammalian postnatal maturation. Thus, miR-1-mediated regulation of Nedd4/Nedd4L expression may serve to broadly modulate the trafficking or degradation of Nedd4/Nedd4L substrates in the heart.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , MicroRNAs/metabolism , Ubiquitin-Protein Ligases/metabolism , 3' Untranslated Regions , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Body Patterning , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Green Fluorescent Proteins/metabolism , Heart/physiology , Nedd4 Ubiquitin Protein Ligases , Phenotype , Phosphorylation , Protein Transport , Receptors, Notch/metabolism , Signal Transduction , Ubiquitination , Wings, Animal/metabolism
3.
J Biol Chem ; 289(20): 14263-71, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24719334

ABSTRACT

MicroRNA (miRNA) maturation is regulated by interaction of particular miRNA precursors with specific RNA-binding proteins. Following their biogenesis, mature miRNAs are incorporated into the RNA-induced silencing complex (RISC) where they interact with mRNAs to negatively regulate protein production. However, little is known about how mature miRNAs are regulated at the level of their activity. To address this, we screened for proteins differentially bound to the mature form of the miR-1 or miR-133 miRNA families. These muscle-enriched, co-transcribed miRNA pairs cooperate to suppress smooth muscle gene expression in the heart. However, they also have opposing roles, with the miR-1 family, composed of miR-1 and miR-206, promoting myogenic differentiation, whereas miR-133 maintains the progenitor state. Here, we describe a physical interaction between TDP-43, an RNA-binding protein that forms aggregates in the neuromuscular disease, amyotrophic lateral sclerosis, and the miR-1, but not miR-133, family. Deficiency of the TDP-43 Drosophila ortholog enhanced dmiR-1 activity in vivo. In mammalian cells, TDP-43 limited the activity of both miR-1 and miR-206, but not the miR-133 family, by disrupting their RISC association. Consistent with TDP-43 dampening miR-1/206 activity, protein levels of the miR-1/206 targets, IGF-1 and HDAC4, were elevated in TDP-43 transgenic mouse muscle. This occurred without corresponding Igf-1 or Hdac4 mRNA increases and despite higher miR-1 and miR-206 expression. Our findings reveal that TDP-43 negatively regulates the activity of the miR-1 family of miRNAs by limiting their bioavailability for RISC loading and suggest a processing-independent mechanism for differential regulation of miRNA activity.


Subject(s)
DNA-Binding Proteins/metabolism , MicroRNAs/metabolism , RNA-Induced Silencing Complex/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Argonaute Proteins/metabolism , Drosophila melanogaster/genetics , Humans , Male , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Protein Binding
4.
Nat Cell Biol ; 13(10): 1244-51, 2011 Aug 14.
Article in English | MEDLINE | ID: mdl-21841793

ABSTRACT

Cellular decisions of self-renewal or differentiation arise from integration and reciprocal titration of numerous regulatory networks. Notch and Wnt/ß-catenin signalling often intersect in stem and progenitor cells and regulate each other transcriptionally. The biological outcome of signalling through each pathway often depends on the context and timing as cells progress through stages of differentiation. Here, we show that membrane-bound Notch physically associates with unphosphorylated (active) ß-catenin in stem and colon cancer cells and negatively regulates post-translational accumulation of active ß-catenin protein. Notch-dependent regulation of ß-catenin protein did not require ligand-dependent membrane cleavage of Notch or the glycogen synthase kinase-3ß-dependent activity of the ß-catenin destruction complex. It did, however, require the endocytic adaptor protein Numb and lysosomal activity. This study reveals a previously unrecognized function of Notch in negatively titrating active ß-catenin protein levels in stem and progenitor cells.


Subject(s)
Colonic Neoplasms/metabolism , Embryonic Stem Cells/metabolism , Protein Processing, Post-Translational , Receptor, Notch1/metabolism , beta Catenin/metabolism , Animals , Cell Differentiation , Cell Proliferation , Colonic Neoplasms/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HT29 Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , RNA Interference , Receptor, Notch1/genetics , Signal Transduction , Transfection , beta Catenin/deficiency , beta Catenin/genetics
5.
J Cell Biol ; 193(7): 1181-96, 2011 Jun 27.
Article in English | MEDLINE | ID: mdl-21690310

ABSTRACT

Unraveling the gene regulatory networks that govern development and function of the mammalian heart is critical for the rational design of therapeutic interventions in human heart disease. Using the Drosophila heart as a platform for identifying novel gene interactions leading to heart disease, we found that the Rho-GTPase Cdc42 cooperates with the cardiac transcription factor Tinman/Nkx2-5. Compound Cdc42, tinman heterozygous mutant flies exhibited impaired cardiac output and altered myofibrillar architecture, and adult heart-specific interference with Cdc42 function is sufficient to cause these same defects. We also identified K(+) channels, encoded by dSUR and slowpoke, as potential effectors of the Cdc42-Tinman interaction. To determine whether a Cdc42-Nkx2-5 interaction is conserved in the mammalian heart, we examined compound heterozygous mutant mice and found conduction system and cardiac output defects. In exploring the mechanism of Nkx2-5 interaction with Cdc42, we demonstrated that mouse Cdc42 was a target of, and negatively regulated by miR-1, which itself was negatively regulated by Nkx2-5 in the mouse heart and by Tinman in the fly heart. We conclude that Cdc42 plays a conserved role in regulating heart function and is an indirect target of Tinman/Nkx2-5 via miR-1.


Subject(s)
Drosophila Proteins/physiology , Drosophila/physiology , Heart/physiology , Homeodomain Proteins/physiology , MicroRNAs/physiology , Repressor Proteins/physiology , Trans-Activators/physiology , Transcription Factors/physiology , cdc42 GTP-Binding Protein/metabolism , Animals , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Heart Diseases/genetics , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Humans , Mice , Myocardial Contraction/genetics , Myocardium/metabolism , Myocytes, Cardiac/cytology , Repressor Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/physiology , rho GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/physiology
6.
Dev Cell ; 20(4): 497-510, 2011 Apr 19.
Article in English | MEDLINE | ID: mdl-21497762

ABSTRACT

Many molecular pathways involved in heart disease have their roots in evolutionarily ancient developmental programs that depend critically on gene dosage and timing. MicroRNAs (miRNAs) modulate gene dosage posttranscriptionally, and among these, the muscle-specific miR-1 is particularly important for developing and maintaining somatic/skeletal and cardiac muscle. To identify pathways regulated by miR-1, we performed a forward genetic screen in Drosophila using wing-vein patterning as a biological assay. We identified several unexpected genes that genetically interacted with dmiR-1, one of which was kayak, encodes a developmentally regulated transcription factor. Additional studies directed at this genetic relationship revealed a previously unappreciated function of dmiR-1 in regulating the polarity of cardiac progenitor cells. The mammalian ortholog of kayak, c-Fos, was dysregulated in hearts of gain- or loss-of-function miR-1 mutant mice in a stress-dependent manner. These findings illustrate the power of Drosophila-based screens to find points of intersection between miRNAs and conserved pathways in mammals.


Subject(s)
Cell Polarity/genetics , Drosophila/genetics , Genome-Wide Association Study , MicroRNAs/genetics , Myocardium/cytology , Animals , Cell Polarity/physiology , Drosophila/embryology , Mice , Mice, Transgenic , MicroRNAs/metabolism , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
J Biol Chem ; 283(51): 35845-52, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-18852265

ABSTRACT

Myocardin (MYOCD) is a transcriptional co-activator that promotes cardiac or smooth muscle gene programs through its interaction with myocyte-enhancing factor (MEF2) or serum-response factor (SRF). Isoforms of MYOCD with a truncated amino terminus show increased activity when compared with those with the full-length amino terminus, but how this is achieved remains unknown. We identified a rare human sequence variation in MYOCD in a patient with congenital heart disease that resulted in a missense mutation at codon 259 (K259R). This variation created a hypomorphic cardiac isoform with impaired SRF binding and transactivation capacity but did not impair the smooth muscle isoform of MYOCD, which lacks the amino terminus. Consistent with differential effects of the amino terminus on the K259R mutation, we found that the cardiac-specific amino terminus acted in an autoinhibitory fashion to bind MYOCD via specific negatively charged residues and thereby repressed SRF-dependent MYOCD activity. This effect was exaggerated in the MYOCD-K259R mutant. The amino terminus was sufficient to impair MYOCD-dependent fibroblast conversion into smooth muscle cells as well as cardiomyocyte hypertrophy. These findings identify a novel mechanism that regulates levels of MYOCD-dependent activation of the SRF genetic program differentially in cardiac and smooth muscle.


Subject(s)
Heart Defects, Congenital/metabolism , Mutation, Missense , Myocytes, Cardiac/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Animals , COS Cells , Chlorocebus aethiops , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Humans , Myocytes, Cardiac/pathology , Nuclear Proteins/genetics , Protein Structure, Tertiary/genetics , Rats , Rats, Sprague-Dawley , Trans-Activators/genetics , Transcriptional Activation/genetics
8.
Proc Natl Acad Sci U S A ; 104(35): 14008-13, 2007 Aug 28.
Article in English | MEDLINE | ID: mdl-17715064

ABSTRACT

Embryonic organs attain their final dimensions through the generation of proper cell number and size, but the control mechanisms remain obscure. Here, we establish Gridlock (Grl), a Hairy-related basic helix-loop-helix (bHLH) transcription factor, as a negative regulator of cardiomyocyte proliferative growth in zebrafish embryos. Mutations in grl cause an increase in expression of a group of immediate-early growth genes, myocardial genes, and development of hyperplastic hearts. Conversely, cardiomyocytes with augmented Grl activity have diminished cell volume and fail to divide, resulting in a marked reduction in heart size. Both bHLH domain and carboxyl region are required for Grl negative control of myocardial proliferative growth. These Grl-induced cardiac effects are counterbalanced by the transcriptional activator Gata5 but not Gata4, which promotes cardiomyocyte expansion in the embryo. Biochemical analyses show that Grl forms a complex with Gata5 through the carboxyl region and can repress Gata5-mediated transcription via the bHLH domain. Hence, our studies suggest that Grl regulates embryonic heart growth via opposing Gata5, at least in part through their protein interactions in modulating gene expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , GATA5 Transcription Factor/genetics , Heart/growth & development , Myocytes, Cardiac/physiology , Zebrafish Proteins/genetics , Animals , Cell Division , Embryonic Development , Gene Expression Regulation, Developmental , Genes, Reporter , Heart/anatomy & histology , Heart/embryology , Molecular Sequence Data , Myocytes, Cardiac/cytology , Zebrafish
9.
Biochem Biophys Res Commun ; 345(1): 446-52, 2006 Jun 23.
Article in English | MEDLINE | ID: mdl-16682003

ABSTRACT

Notch signaling is central to cell differentiation, organ development, and apoptosis. Upon ligand binding, the Notch intracellular domain (NotchIC) translocates to the nucleus to interact with its DNA-binding partner, RBP-Jkappa. The NotchIC-RBP-Jkappa complex activates target genes, such as those encoding the Hrt and Hes families of basic-helix-loop-helix (bHLH) transcriptional repressors. Hrt transcripts are enriched in the developing cardiovascular system, and mice lacking Hrt2 have cardiac malformations. Here we show that Hrt2 and Hes1 interact with RBP-Jkappa to negatively regulate Notch-dependent activation of Hrt and Hes expression. The bHLH domain of Hrt2 was necessary for this interaction, and disrupting the protein complex abrogated the negative autoregulation. The interaction did not interfere with the formation or DNA-binding of the NotchIC-RBP-Jkappa complex, indicating direct inhibition by Hrt and Hes as co-repressors. These findings suggest a novel mechanism for negative feedback on Notch signaling that requires RBP-Jkappa to interact physically with Hrt and Hes.


Subject(s)
Arabidopsis Proteins/metabolism , DNA-Binding Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Animals , COS Cells , Chlorocebus aethiops , Down-Regulation/physiology , HeLa Cells , Humans
10.
Nature ; 437(7056): 270-4, 2005 Sep 08.
Article in English | MEDLINE | ID: mdl-16025100

ABSTRACT

Calcification of the aortic valve is the third leading cause of heart disease in adults. The incidence increases with age, and it is often associated with a bicuspid aortic valve present in 1-2% of the population. Despite the frequency, neither the mechanisms of valve calcification nor the developmental origin of a two, rather than three, leaflet aortic valve is known. Here, we show that mutations in the signalling and transcriptional regulator NOTCH1 cause a spectrum of developmental aortic valve anomalies and severe valve calcification in non-syndromic autosomal-dominant human pedigrees. Consistent with the valve calcification phenotype, Notch1 transcripts were most abundant in the developing aortic valve of mice, and Notch1 repressed the activity of Runx2, a central transcriptional regulator of osteoblast cell fate. The hairy-related family of transcriptional repressors (Hrt), which are activated by Notch1 signalling, physically interacted with Runx2 and repressed Runx2 transcriptional activity independent of histone deacetylase activity. These results suggest that NOTCH1 mutations cause an early developmental defect in the aortic valve and a later de-repression of calcium deposition that causes progressive aortic valve disease.


Subject(s)
Aortic Valve/abnormalities , Heart Valve Diseases/genetics , Mutation/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Adult , Animals , Aortic Valve/pathology , Base Sequence , COS Cells , Calcinosis/genetics , Child , Chromosomes, Human, Pair 9/genetics , Core Binding Factor Alpha 1 Subunit , DNA Mutational Analysis , Female , Gene Expression Regulation, Developmental , Heart Valve Diseases/congenital , Heart Valve Diseases/pathology , Heart Valve Diseases/physiopathology , Humans , In Situ Hybridization , Lod Score , Male , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pedigree , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Notch1 , Receptors, Cell Surface/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/chemistry
11.
J Biol Chem ; 279(52): 54937-43, 2004 Dec 24.
Article in English | MEDLINE | ID: mdl-15485867

ABSTRACT

Combinatorial actions of transcription factors in multiprotein complexes dictate gene expression profiles in cardiac development and disease. The Hairy-related transcription factor (HRT) family of basic helix-loop-helix proteins is composed of transcriptional repressors highly expressed in the cardiovascular system. However, it has remained unclear whether HRT proteins modulate gene expression driven by cardiac transcriptional activators. Here, we have shown that HRT proteins inhibit cardiac gene transcription by interfering with GATA transcription factors that are implicated in cardiac development and hypertrophy. HRT proteins inhibited GATA-dependent transcriptional activation of cardiac gene promoters such as the atrial natriuretic factor (ANF) promoter. Adenovirus-mediated expression of Hrt2 suppressed mRNA expression of ANF and other cardiac-specific genes in cultured cardiomyocytes. Among various signaling molecules implicated in cardiomyocyte growth, constitutively active Akt1/protein kinase B alpha relieved Hrt2-mediated inhibition of GATA-dependent transcription. HRT proteins physically interacted with GATA proteins, and the basic domain of HRT was critical for physical association as well as transcriptional inhibition. These results suggest that HRT proteins may regulate specific sets of cardiac genes by modulating the function of GATA proteins and other cardiac transcriptional activators in a signal-dependent manner.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression/physiology , Myocytes, Cardiac/metabolism , Repressor Proteins/physiology , Signal Transduction , Transcription Factors/physiology , Animals , Atrial Natriuretic Factor/genetics , Basic Helix-Loop-Helix Transcription Factors , COS Cells , Cardiomegaly , Cells, Cultured , Chlorocebus aethiops , DNA-Binding Proteins/genetics , GATA4 Transcription Factor , Gene Deletion , Gene Expression/drug effects , Helix-Loop-Helix Motifs , Humans , Luciferases/genetics , Mice , Myocytes, Cardiac/chemistry , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , RNA, Messenger/analysis , Rats , Recombinant Fusion Proteins , Repressor Proteins/genetics , Transcription Factors/genetics , Transfection
12.
Nature ; 424(6947): 443-7, 2003 Jul 24.
Article in English | MEDLINE | ID: mdl-12845333

ABSTRACT

Congenital heart defects (CHDs) are the most common developmental anomaly and are the leading non-infectious cause of mortality in newborns. Only one causative gene, NKX2-5, has been identified through genetic linkage analysis of pedigrees with non-syndromic CHDs. Here, we show that isolated cardiac septal defects in a large pedigree were linked to chromosome 8p22-23. A heterozygous G296S missense mutation of GATA4, a transcription factor essential for heart formation, was found in all available affected family members but not in any control individuals. This mutation resulted in diminished DNA-binding affinity and transcriptional activity of Gata4. Furthermore, the Gata4 mutation abrogated a physical interaction between Gata4 and TBX5, a T-box protein responsible for a subset of syndromic cardiac septal defects. Conversely, interaction of Gata4 and TBX5 was disrupted by specific human TBX5 missense mutations that cause similar cardiac septal defects. In a second family, we identified a frame-shift mutation of GATA4 (E359del) that was transcriptionally inactive and segregated with cardiac septal defects. These results implicate GATA4 as a genetic cause of human cardiac septal defects, perhaps through its interaction with TBX5.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heart Defects, Congenital/genetics , Mutation/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Xenopus Proteins , Animals , Binding Sites , COS Cells , Chromosome Mapping , Chromosomes, Human, Pair 8/genetics , DNA/genetics , DNA/metabolism , DNA Mutational Analysis , DNA-Binding Proteins/chemistry , Electrophoretic Mobility Shift Assay , Female , Frameshift Mutation/genetics , GATA4 Transcription Factor , HeLa Cells , Heart Defects, Congenital/physiopathology , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/metabolism , Humans , Male , Mice , Pedigree , Precipitin Tests , Protein Binding , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/genetics , Transcription Factors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...