Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
PLoS Biol ; 22(3): e3002565, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38527087

ABSTRACT

K+ channels regulate morphogens to scale adult fins, but little is known about what regulates the channels and how they control morphogen expression. Using the zebrafish pectoral fin bud as a model for early vertebrate fin/limb development, we found that K+ channels also scale this anatomical structure, and we determined how one K+-leak channel, Kcnk5b, integrates into its developmental program. From FLIM measurements of a Förster Resonance Energy Transfer (FRET)-based K+ sensor, we observed coordinated decreases in intracellular K+ levels during bud growth, and overexpression of K+-leak channels in vivo coordinately increased bud proportions. Retinoic acid, which can enhance fin/limb bud growth, decreased K+ in bud tissues and up-regulated regulator of calcineurin (rcan2). rcan2 overexpression increased bud growth and decreased K+, while CRISPR-Cas9 targeting of rcan2 decreased growth and increased K+. We observed similar results in the adult caudal fins. Moreover, CRISPR targeting of Kcnk5b revealed that Rcan2-mediated growth was dependent on the Kcnk5b. We also found that Kcnk5b enhanced depolarization in fin bud cells via Na+ channels and that this enhanced depolarization was required for Kcnk5b-enhanced growth. Lastly, Kcnk5b-induced shha transcription and bud growth required IP3R-mediated Ca2+ release and CaMKK activity. Thus, we provide a mechanism for how retinoic acid via rcan2 can regulate K+-channel activity to scale a vertebrate appendage via intercellular Ca2+ signaling.


Subject(s)
Calcium , Zebrafish , Animals , Zebrafish/genetics , Calcium/metabolism , Tretinoin , Animal Fins/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Gene Expression Regulation, Developmental
2.
Dev Biol ; 478: 89-101, 2021 10.
Article in English | MEDLINE | ID: mdl-34048735

ABSTRACT

Inosine monophosphate dehydrogenase (IMPDH) catalyzes the rate-limiting step in de novo guanine nucleotide biosynthesis. Its activity is negatively regulated by the binding of GTP. IMPDH can form a membraneless subcellular structure termed the cytoophidium in response to certain changes in the metabolic status of the cell. The polymeric form of IMPDH, which is the subunit of the cytoophidium, has been shown to be more resistant to the inhibition by GTP at physiological concentrations, implying a functional correlation between cytoophidium formation and the upregulation of GTP biosynthesis. Herein we demonstrate that zebrafish IMPDH1b and IMPDH2 isoforms can assemble abundant cytoophidium in most of cultured cells under stimuli, while zebrafish IMPDH1a shows distinctive properties of forming the cytoophidium in different cell types. Point mutations that disrupt cytoophidium structure in mammalian models also prevent the aggregation of zebrafish IMPDHs. In addition, we discover the presence of the IMPDH cytoophidium in various tissues of larval and adult fish under normal growth conditions. Our results reveal that polymerization and cytoophidium assembly of IMPDH can be a regulatory machinery conserved among vertebrates, and with specific physiological purposes.


Subject(s)
Cytoplasmic Structures/ultrastructure , IMP Dehydrogenase/chemistry , Zebrafish Proteins/chemistry , Zebrafish/metabolism , Animals , Cell Line , Cytoplasmic Structures/chemistry , Gene Expression , Guanosine Triphosphate/biosynthesis , Guanosine Triphosphate/metabolism , Humans , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Isoenzymes/chemistry , Isoenzymes/genetics , Point Mutation , Up-Regulation , Zebrafish/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
3.
Exp Cell Res ; 405(2): 112684, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34129847

ABSTRACT

Cytidine triphosphate synthase (CTPS) catalyzes the rate-limiting step of de novo CTP biosynthesis. An intracellular structure of CTPS, the cytoophidium, has been found in many organisms including prokaryotes and eukaryotes. Formation of the cytoophidium has been suggested to regulate the activity and stability of CTPS and may participate in certain physiological events. Herein, we demonstrate that both CTPS1a and CTPS1b in zebrafish are able to form the cytoophidium in cultured cells. A point mutation, H355A, abrogates cytoophidium assembly of zebrafish CTPS1a and CTPS1b. In addition, we show the presence of CTPS cytoophidia in multiple tissues of larval and adult fish under normal conditions, while treatment with a CTPS inhibitor 6-diazo-5-oxo-l-norleucine (DON) can induce more cytoophidia in some tissues. Our findings reveal that forming the CTPS cytoophidium is a natural phenomenon of zebrafish and provide valuable information for future research on the physiological importance of this intracellular structure in vertebrates.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Cytidine Triphosphate/metabolism , Eukaryota/cytology , Prokaryotic Cells/cytology , Animals , Cell Line , Nitric Oxide Synthase/metabolism , Zebrafish
4.
Development ; 141(18): 3529-39, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25183871

ABSTRACT

Canonical ß-catenin-dependent Wnt signal transduction is important for several biological phenomena, such as cell fate determination, cell proliferation, stem cell maintenance and anterior-posterior axis formation. The hallmark of canonical Wnt signaling is the translocation of ß-catenin into the nucleus where it activates gene transcription. However, the mechanisms regulating ß-catenin nuclear localization are poorly understood. We show that Simplet/Fam53B (Smp) is required for Wnt signaling by positively regulating ß-catenin nuclear localization. In the zebrafish embryo, the loss of smp blocks the activity of two ß-catenin-dependent reporters and the expression of Wnt target genes, and prevents nuclear accumulation of ß-catenin. Conversely, overexpression of smp increases ß-catenin nuclear localization and transcriptional activity in vitro and in vivo. Expression of mutant Smp proteins lacking either the nuclear localization signal or the ß-catenin interaction domain reveal that the translocation of Smp into the nucleus is essential for ß-catenin nuclear localization and Wnt signaling in vivo. We also provide evidence that mammalian Smp is involved in regulating ß-catenin nuclear localization: the protein colocalizes with ß-catenin-dependent gene expression in mouse intestinal crypts; siRNA knockdown of Smp reduces ß-catenin nuclear localization and transcriptional activity; human SMP mediates ß-catenin transcriptional activity in a dose-dependent manner; and the human SMP protein interacts with human ß-catenin primarily in the nucleus. Thus, our findings identify the evolutionary conserved SMP protein as a regulator of ß-catenin-dependent Wnt signal transduction.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation, Developmental/physiology , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Blotting, Western , Humans , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Luciferases , Mice , Mice, Transgenic , RNA Interference , RNA, Small Interfering/genetics , Wnt Proteins/genetics
5.
Elife ; 102021 04 08.
Article in English | MEDLINE | ID: mdl-33830014

ABSTRACT

The increase in activity of the two-pore potassium-leak channel Kcnk5b maintains allometric juvenile growth of adult zebrafish appendages. However, it remains unknown how this channel maintains allometric growth and how its bioelectric activity is regulated to scale these anatomical structures. We show the activation of Kcnk5b is sufficient to activate several genes that are part of important development programs. We provide in vivo transplantation evidence that the activation of gene transcription is cell autonomous. We also show that Kcnk5b will induce the expression of different subsets of the tested developmental genes in different cultured mammalian cell lines, which may explain how one electrophysiological stimulus can coordinately regulate the allometric growth of diverse populations of cells in the fin that use different developmental signals. We also provide evidence that the post-translational modification of serine 345 in Kcnk5b by calcineurin regulates channel activity to scale the fin. Thus, we show how an endogenous bioelectric mechanism can be regulated to promote coordinated developmental signaling to generate and scale a vertebrate appendage.


Organs, limbs, fins and tails are made of multiple tissues whose growth is controlled by specific signals and genetic programmes. All these different cell populations must work together during development or regeneration to form a complete structure that is the right size in relation to the rest of the body. Growing evidence suggests that this synchronicity might be down to electric signals, which are created by movements of charged particles in and out of cells. In particular, previous work has identified two factors that control the development of fins in fish: the Kcnk5b potassium-leak channel, which allows positive ions to cross the cell membrane; and an enzyme called calcineurin, which can modify the activity of proteins. Kcnk5b and calcineurin seem to play similar roles in the proportional growth of the fins in relation to the body, but exactly how was unknown. To investigate this question, Yi et al. used genetically modified zebrafish to show how the Kcnk5b channel could control genes responsible for appendage growth. However, their tests on different cell types revealed that potassium movement through the Kcnk5b channel leads to different sets of developmental genes being turned on, depending on the tissue type of the cell. This could explain how one type of signal (in this case, movement of ions) can coordinate the growth of a wide range of tissues that use different combinations of developmental genes to form. Kcnk5b therefore appears to coordinate the regulation of the various combinations of genes needed for different fin tissues to develop, so that every component grows in a proportional, synchronized manner. Yi et al. also showed that calcineurin can modify the Kcnk5b channel to control its activity. In turn, this affects the movement of potassium ions across the membrane, changing electrical activity and, as a consequence, the proportional growth of the fin. Further work should explore how Kcnk5b and calcineurin link to other signals that regulate the size of fins and limbs. Ultimately, a finer understanding of the molecules controlling the growth of body parts will be useful in fields such as regenerative medicine or stem cell biology, which attempt to build organs for clinical therapies.


Subject(s)
Animal Fins/metabolism , Calcineurin/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Potassium/metabolism , Transcription, Genetic , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animal Fins/embryology , Animal Fins/growth & development , Animals , Animals, Genetically Modified , Calcineurin/genetics , Female , HEK293 Cells , HeLa Cells , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Ion Channel Gating , Male , Membrane Potentials , Morphogenesis , Phosphorylation , Potassium Channels, Tandem Pore Domain/genetics , Protein Processing, Post-Translational , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics
6.
Acta Pharm Sin B ; 11(10): 3206-3219, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34729310

ABSTRACT

The TEA domain (TEAD) family proteins (TEAD1‒4) are essential transcription factors that control cell differentiation and organ size in the Hippo pathway. Although the sequences and structures of TEAD family proteins are highly conserved, each TEAD isoform has unique physiological and pathological functions. Therefore, the development and discovery of subtype selective inhibitors for TEAD protein will provide important chemical probes for the TEAD-related function studies in development and diseases. Here, we identified a novel TEAD1/3 covalent inhibitor (DC-TEADin1072) with biochemical IC50 values of 0.61 ± 0.02 and 0.58 ± 0.12 µmol/L against TEAD1 and TEAD3, respectively. Further chemical optimization based on DC-TEAD in 1072 yielded a selective TEAD3 inhibitor DC-TEAD3in03 with the IC50 value of 0.16 ± 0.03 µmol/L, which shows 100-fold selectivity over other TEAD isoforms in activity-based protein profiling (ABPP) assays. In cells, DC-TEAD3in03 showed selective inhibitory effect on TEAD3 in GAL4-TEAD (1-4) reporter assays with the IC50 value of 1.15 µmol/L. When administered to zebrafish juveniles, experiments showed that DC-TEAD3in03 reduced the growth rate of zebrafish caudal fins, indicating the importance of TEAD3 activity in controlling proportional growth of vertebrate appendages.

7.
Dev Biol ; 325(2): 329-40, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19014929

ABSTRACT

Two hallmarks of vertebrate epimorphic regeneration are a significant increase in the proliferation of normally quiescent cells and a re-activation of genes that are active during embryonic development. It is unclear what the molecular determinants are that regulate these events and how they are coordinated. Zebrafish have the ability to regenerate several compound structures by regulating cell proliferation and gene transcription. We report that fam53b/simplet (smp) regulates both cell proliferation and the transcription of specific genes. In situ hybridization and quantitative RT-PCR experiments showed that amputation of zebrafish hearts and fins resulted in strong up-regulation of the smp gene. In regenerating adult fin, smp expression remained strong in the distal mesenchyme which later expanded to the basal layers of the distal epidermis and distal tip epithelium. Morpholino knockdown of smp reduced regenerative outgrowth by decreasing cell proliferation as measured by BrdU incorporation and histone H3 phosphorylation. In addition, smp knockdown increased the expression of msxb, msxc, and shh, as well as the later formation of ectopic bone. Taken together, these data indicate a requirement for smp in fin regeneration through control of cell proliferation, the regulation of specific genes and proper bone patterning.


Subject(s)
Cell Proliferation , Extremities/physiology , Transcription Factors/physiology , Zebrafish Proteins/physiology , Zebrafish/physiology , Animals , Animals, Genetically Modified , Epidermis/growth & development , Epidermis/physiology , Extremities/growth & development , Gene Expression Regulation , Mesoderm/growth & development , Mesoderm/physiology , Myocardium/metabolism , Osteogenesis/physiology , Regeneration , Transcription Factors/genetics , Zebrafish Proteins/genetics
8.
Adv Exp Med Biol ; 695: 184-214, 2010.
Article in English | MEDLINE | ID: mdl-21222207

ABSTRACT

There are several animal model organisms that have the ability to regenerate severe injuries by stimulating local cells to restore damaged and lost organs and appendages. In this chapter, we will describe how various vertebrate animals regenerate different structures (central nervous system, heart and appendages) as well as detail specific cellular and molecular features concerning the regeneration of these structures.


Subject(s)
Regeneration , Vertebrates , Animals , Central Nervous System , Extremities , Stem Cells
9.
Circ Res ; 98(8): 1089-97, 2006 Apr 28.
Article in English | MEDLINE | ID: mdl-16556869

ABSTRACT

In response to stress signals, postnatal cardiomyocytes undergo hypertrophic growth accompanied by activation of a fetal gene program, assembly of sarcomeres, and cellular enlargement. We show that hypertrophic signals stimulate the expression and transcriptional activity of myocardin, a cardiac and smooth muscle-specific coactivator of serum response factor (SRF). Consistent with a role for myocardin as a transducer of hypertrophic signals, forced expression of myocardin in cardiomyocytes is sufficient to substitute for hypertrophic signals and induce cardiomyocyte hypertrophy and the fetal cardiac gene program. Conversely, a dominant-negative mutant form of myocardin, which retains the ability to associate with SRF but is defective in transcriptional activation, blocks cardiomyocyte hypertrophy induced by hypertrophic agonists such as phenylephrine and leukemia inhibitory factor. Myocardin-dependent hypertrophy can also be partially repressed by histone deacetylase 5, a transcriptional repressor of myocardin. These findings identify myocardin as a nuclear effector of hypertrophic signaling pathways that couples stress signals to a transcriptional program for postnatal cardiac growth and remodeling.


Subject(s)
Cardiomegaly/physiopathology , Muscle Cells/cytology , Nuclear Proteins/genetics , Trans-Activators/genetics , Animals , Cardiomegaly/genetics , Cells, Cultured , Disease Models, Animal , Heart/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Muscle Cells/metabolism , Myocardium/cytology , Myocardium/metabolism , Nuclear Proteins/metabolism , Rats , Serum Response Factor/physiology , Trans-Activators/metabolism
10.
Dev Cell ; 46(1): 85-101.e8, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29974866

ABSTRACT

Neural stem cells (NSCs) constitute an endogenous reservoir for neurons that could potentially be harnessed for regenerative therapies in disease contexts such as neurodegeneration. However, in Alzheimer's disease (AD), NSCs lose plasticity and thus possible regenerative capacity. We investigate how NSCs lose their plasticity in AD by using starPEG-heparin-based hydrogels to establish a reductionist 3D cell-instructive neuro-microenvironment that promotes the proliferative and neurogenic ability of primary and induced human NSCs. We find that administration of AD-associated Amyloid-ß42 causes classical neuropathology and hampers NSC plasticity by inducing kynurenic acid (KYNA) production. Interleukin-4 restores NSC proliferative and neurogenic ability by suppressing the KYNA-producing enzyme Kynurenine aminotransferase (KAT2), which is upregulated in APP/PS1dE9 mouse model of AD and in postmortem human AD brains. Thus, our culture system enables a reductionist investigation of regulation of human NSC plasticity for the identification of potential therapeutic targets for intervention in AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Cell Plasticity/physiology , Interleukin-4/metabolism , Neural Stem Cells/cytology , Neurogenesis/physiology , Adult , Aged, 80 and over , Alzheimer Disease , Animals , Brain/metabolism , Cell Proliferation/physiology , Cells, Cultured , Disease Models, Animal , Female , Humans , Kynurenic Acid/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Neural Stem Cells/physiology , Neurons/cytology , Transaminases/metabolism , Transcriptional Activation/genetics , Young Adult
11.
J Clin Invest ; 113(11): 1535-49, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15173880

ABSTRACT

Environmental stresses converge on the mitochondria that can trigger or inhibit cell death. Excitable, postmitotic cells, in response to sublethal noxious stress, engage mechanisms that afford protection from subsequent insults. We show that reoxygenation after prolonged hypoxia reduces the reactive oxygen species (ROS) threshold for the mitochondrial permeability transition (MPT) in cardiomyocytes and that cell survival is steeply negatively correlated with the fraction of depolarized mitochondria. Cell protection that exhibits a memory (preconditioning) results from triggered mitochondrial swelling that causes enhanced substrate oxidation and ROS production, leading to redox activation of PKC, which inhibits glycogen synthase kinase-3beta (GSK-3beta). Alternatively, receptor tyrosine kinase or certain G protein-coupled receptor activation elicits cell protection (without mitochondrial swelling or durable memory) by inhibiting GSK-3beta, via protein kinase B/Akt and mTOR/p70(s6k) pathways, PKC pathways, or protein kinase A pathways. The convergence of these pathways via inhibition of GSK-3beta on the end effector, the permeability transition pore complex, to limit MPT induction is the general mechanism of cardiomyocyte protection.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Ion Channels/antagonists & inhibitors , Signal Transduction/physiology , Animals , Glycogen Synthase Kinase 3 beta , Hypoxia/metabolism , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Myocytes, Cardiac , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Respiratory Burst/physiology
12.
Int J Cardiol ; 241: 379-386, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28377185

ABSTRACT

In vitro generation of cardiomyocytes (CMs) from human cells opens the possibility to develop patient-specific therapies to various cardiomyopathies. By establishing the in vitro reprograming methods that produce human CMs, we learn about what is involved in the development of specific CM subtypes. In this review, we summarize the latest achievements in CM generation technologies, emphasizing the differentiation methods of specific CM subtypes. We also relate the biological properties and functions of the in vitro-generated CMs to those of their in vivo counterparts. Furthermore, we describe the main problem of current CM derivation methods - maturation of CMs. We subsequently discuss biochemical and physical stimuli that are used to overcome the maturation problems of in vitro-derived CMs. As a result, a more holistic approach with controllable environment and timing of specific stimuli for creation of more mature engineered heart tissues is described as well. Finally, we propose a novel approach in which enhancing energy transfer mechanisms in the immature CMs might help to overcome the current hurdle of incomplete in vitro differentiation.


Subject(s)
Cell Differentiation/physiology , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Action Potentials/physiology , Cells, Cultured , Humans
13.
Sci Rep ; 7(1): 12959, 2017 10 11.
Article in English | MEDLINE | ID: mdl-29021554

ABSTRACT

Microtubule-associated TAU protein is a pathological hallmark in Alzheimer's disease (AD), where hyperphosphorylation of TAU generates neurofibrillary tangles. To investigate the effects of TAU in a regenerative adult vertebrate brain system, we generated a cre/lox-based transgenic model of zebrafish that chronically expresses human TAUP301L, which is a variant of human TAU protein that forms neurofibrillary tangles in mouse models and humans. Interestingly, we found that although chronic and abundant expression of TAUP301L starting from early embryonic development led to hyperphosphorylation, TAUP301L did not form oligomers and neurofibrillary tangles, and did not cause elevated apoptosis and microglial activation, which are classical symptoms of tauopathies in mammals. Additionally, TAUP301L neither increased neural stem cell proliferation nor activated the expression of regenerative factor Interleukin-4, indicating that TAUP301L toxicity is prevented in the adult zebrafish brain. By combining TAUP301L expression with our established Aß42 toxicity model, we found that Aß42 ceases to initiate neurofibrillary tangle formation by TAUP301L, and TAUP301L does not exacerbate the toxicity of Aß42. Therefore, our results propose a cellular mechanism that protects the adult zebrafish brain against tauopathies, and our model can be used to understand how TAU toxicity can be prevented in humans.


Subject(s)
Aging/metabolism , Brain/metabolism , Mutant Proteins/metabolism , Neurofibrillary Tangles/metabolism , Zebrafish/metabolism , tau Proteins/metabolism , Amyloid beta-Peptides/toxicity , Animals , Animals, Genetically Modified , Behavior, Animal , Cell Death , Humans , Inflammation/pathology , Larva/metabolism , Models, Biological , Nerve Regeneration/drug effects , Neurons/metabolism , Phenotype , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Protein Multimerization , Stem Cells/metabolism
14.
Naunyn Schmiedebergs Arch Pharmacol ; 390(8): 857-862, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28451724

ABSTRACT

Inhibitor-1 (I-1) modulates protein phosphatase 1 (PP1) activity and thereby counteracts the phosphorylation by kinases. I-1 is downregulated and deactivated in failing hearts, but whether its role is beneficial or detrimental remains controversial, and opposing therapeutic strategies have been proposed. Overactivity of Ca2+/calmodulin-dependent protein kinase II (CaMKII) with hyperphosphorylation of ryanodine receptors (RyR2) at the CaMKII-site is recognized to be central for heart failure and arrhythmias. Using an I-1-deficient mouse line as well as transfected cell lines, we investigated the effects of acute and chronic modulation of I-1 on CaMKII activity and RyR2 phosphorylation. We demonstrate that I-1 acutely modulates CaMKII by regulating PP1 activity. However, while ablation of I-1 should thus limit CaMKII-activation, we unexpectedly found exaggerated CaMKII-activation under ß-adrenergic stress upon chronic loss of I-1 in knockout mice. We unraveled that this is due to chronic upregulation of the exchange protein activated by cAMP (EPAC) leading to augmented CaMKII activation, and using computational modeling validated that an increase in EPAC expression can indeed explain our experimental findings. Interestingly, at the level of RyR2, the increase in PP1 activity more than outweighed the increase in CaMKII activity, resulting in reduced RyR phosphorylation at Ser-2814. Exaggerated CaMKII activation due to counterregulatory mechanisms upon loss of I-1 is an important caveat with respect to suggested therapeutic I-1-inhibition, as CaMKII overactivity has been heavily implicated in several cardiac pathologies.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Myocardium/metabolism , Proteins/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Adrenergic beta-1 Receptor Agonists , Animals , Dobutamine , Dogs , Echocardiography, Stress , Guanine Nucleotide Exchange Factors/metabolism , Mice, Knockout , Myocytes, Cardiac/metabolism , Phosphorylation , Protein Phosphatase 1/metabolism , Rats , Stress, Physiological
15.
Circulation ; 108(23): 2926-33, 2003 Dec 09.
Article in English | MEDLINE | ID: mdl-14623810

ABSTRACT

BACKGROUND: Although beta-adrenergic receptor (AR) blockade therapy is beneficial in the treatment of heart failure, little is known regarding the transcriptional mechanisms underlying this salutary action. METHODS AND RESULTS: In the present study, we screened mice overexpressing Gsalpha, beta1AR, beta2AR, or protein kinase A to test if a common genomic pathway exists in different models with enhanced beta-adrenergic signaling. In mice overexpressing Gsalpha, differentially expressed genes were identified by mRNA profiling. In addition to well-known markers of cardiac hypertrophy (atrial natriuretic factor, CARP, and beta-myosin heavy chain), uncoupling protein 2 (UCP2), a protein involved in the control of mitochondrial membrane potential, and four-and-a-half LIM domain protein-1 (FHL1), a member of the LIM protein family, were predicted to be upregulated. Upregulation of these genes was confirmed by quantitative reverse transcriptase-polymerase chain reaction at all time points tested during the development of cardiomyopathy in mice overexpressing Gsalpha. In mice overexpressing beta1AR, beta2AR, or protein kinase A, increased UCP2 and FHL1 expression was also observed at the onset of cardiomyopathy. BetaAR blockade treatment reversed the cardiomyopathy and suppressed the increased expression of UCP2 and FHL1 in mice overexpressing Gsalpha. CONCLUSIONS: UCP2 and FHL1 are important candidate genes that correlate with the development of betaAR-induced cardiomyopathy in different mouse models with enhanced betaAR signaling. In addition to preserving cardiac function, betaAR blockade treatment also prevents the genomic regulation that correlates with the onset of heart failure.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Cardiomyopathies/genetics , Epinephrine/physiology , Gene Expression Regulation , Heart Failure/prevention & control , Receptors, Adrenergic, beta/physiology , Adrenergic beta-Antagonists/pharmacology , Animals , Cardiomyopathies/drug therapy , Cardiomyopathies/etiology , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic AMP-Dependent Protein Kinases/genetics , GTP-Binding Protein alpha Subunits, Gs/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/genetics , Gene Expression Profiling , Gene Expression Regulation/drug effects , Heart Failure/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Ion Channels , Membrane Transport Proteins/biosynthesis , Membrane Transport Proteins/genetics , Mice , Mice, Transgenic , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Models, Animal , Receptors, Adrenergic, beta/biosynthesis , Receptors, Adrenergic, beta/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Uncoupling Protein 2
16.
J Vis Exp ; (88)2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24961304

ABSTRACT

The zebrafish is an important model to understand the cell and molecular biology of organ and appendage regeneration. However, molecular strategies to employ reverse genetics have not yet been adequately developed to assess gene function in regeneration or tissue homeostasis during larval stages after zebrafish embryogenesis, and several tissues within the zebrafish larva are difficult to target. Intraventricular injections of gene-specific morpholinos offer an alternative method for the current inability to genomically target zebrafish genes in a temporally controlled manner at these stages. This method allows for complete dispersion and subsequent incorporation of the morpholino into various tissues throughout the body, including structures that were formerly impossible to reach such as those in the larval caudal fin, a structure often used to noninvasively research tissue regeneration. Several genes activated during larval finfold regeneration are also present in regenerating adult vertebrate tissues, so the larva is a useful model to understand regeneration in adults. This morpholino dispersion method allows for the quick and easy identification of genes required for the regeneration of larval tissues as well as other physiological phenomena regulating tissue homeostasis after embryogenesis. Therefore, this delivery method provides a currently needed strategy for temporal control to the evaluation of gene function after embryogenesis. 


Subject(s)
Morpholinos/administration & dosage , Morpholinos/genetics , Regeneration/genetics , Transfection/methods , Zebrafish/physiology , Animals , Drug Administration Routes , Fluorescein/chemistry , Heart Ventricles , Larva , Zebrafish/embryology , Zebrafish/genetics
17.
Dev Cell ; 28(5): 573-87, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24561038

ABSTRACT

Vertebrates develop organs and appendages in a proportionally coordinated manner, and animals that regenerate them do so to the same dimensions as the original structures. Coordinated proportional growth involves controlled regulation between allometric and isometric growth programs, but it is unclear what executes this control. We show that calcineurin inhibition results in continued allometric outgrowth of regenerating fins beyond their original dimensions. Calcineurin inhibition also maintains allometric growth of juvenile fins and induces it in adult fins. Furthermore, calcineurin activity is low when the regeneration rate is highest, and its activity increases as the rate decreases. Growth measurements and morphometric analysis of proximodistal asymmetry indicate that calcineurin inhibition shifts fin regeneration from a distal growth program to a proximal program. This shift is associated with the promotion of retinoic acid signaling. Thus, we identified a calcineurin-mediated mechanism that operates as a molecular switch between position-associated isometric and allometric growth programs.


Subject(s)
Animal Fins/growth & development , Calcineurin/metabolism , Regeneration/physiology , Tretinoin/metabolism , Zebrafish/growth & development , Animal Fins/anatomy & histology , Animal Fins/metabolism , Animals , Blotting, Western , Immunoenzyme Techniques , Immunosuppressive Agents/pharmacology , In Situ Hybridization , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Regeneration/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tacrolimus/pharmacology , Zebrafish/anatomy & histology , Zebrafish/metabolism
18.
Am J Physiol Heart Circ Physiol ; 292(2): H838-45, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17012357

ABSTRACT

Mutations in myosin heavy chain (MyHC) can cause hypertrophic cardiomyopathy (HCM) that is characterized by hypertrophy, histopathology, contractile dysfunction, and sudden death. The signaling pathways involved in the pathology of HCM have not been elucidated, and an unresolved question is whether blocking hypertrophic growth in HCM may be maladaptive or beneficial. To address these questions, a mouse model of HCM was crossed with an antihypertrophic mouse model of constitutive activated glycogen synthase kinase-3beta (caGSK-3beta). Active GSK-3beta blocked cardiac hypertrophy in both male and female HCM mice. However, doubly transgenic males (HCM/GSK-3beta) demonstrated depressed contractile function, reduced sarcoplasmic (endo) reticulum Ca(2+)-ATPase (SERCA) expression, elevated atrial natriuretic factor (ANF) expression, and premature death. In contrast, female HCM/GSK-3beta double transgenic mice exhibited similar cardiac histology, function, and survival to their female HCM littermates. Remarkably, dietary modification from a soy-based diet to a casein-based diet significantly improved survival in HCM/GSK-3beta males. These findings indicate that activation of GSK-3beta is sufficient to limit cardiac growth in this HCM model and the consequence of caGSK-3beta was sexually dimorphic. Furthermore, these results show that blocking hypertrophy by active GSK-3beta in this HCM model is not therapeutic.


Subject(s)
Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/physiopathology , Glycogen Synthase Kinase 3/metabolism , Myosin Heavy Chains/metabolism , Ventricular Remodeling , Actins/metabolism , Animals , Atrial Natriuretic Factor/metabolism , Calcium-Binding Proteins/metabolism , Cardiomyopathy, Hypertrophic/diet therapy , Cardiomyopathy, Hypertrophic/pathology , Crosses, Genetic , Dietary Proteins/administration & dosage , Disease Models, Animal , Female , Fibrosis , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Myocardial Contraction , Myosin Heavy Chains/genetics , Phosphorylation , RNA, Messenger/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sex Factors , Time Factors
19.
Cell ; 110(4): 479-88, 2002 Aug 23.
Article in English | MEDLINE | ID: mdl-12202037

ABSTRACT

The heart responds to stress signals by hypertrophic growth, which is accompanied by activation of the MEF2 transcription factor and reprogramming of cardiac gene expression. We show here that class II histone deacetylases (HDACs), which repress MEF2 activity, are substrates for a stress-responsive kinase specific for conserved serines that regulate MEF2-HDAC interactions. Signal-resistant HDAC mutants lacking these phosphorylation sites are refractory to hypertrophic signaling and inhibit cardiomyocyte hypertrophy. Conversely, mutant mice lacking the class II HDAC, HDAC9, are sensitized to hypertrophic signals and exhibit stress-dependent cardiomegaly. Thus, class II HDACs act as signal-responsive suppressors of the transcriptional program governing cardiac hypertrophy and heart failure.


Subject(s)
Cardiomegaly/enzymology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Histone Deacetylases/deficiency , Repressor Proteins , Signal Transduction/genetics , Stress, Physiological/enzymology , Transcription Factors/metabolism , Transcriptional Activation/physiology , Aging/genetics , Aging/metabolism , Aging/pathology , Animals , Calcineurin/genetics , Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 1 , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA-Binding Proteins/genetics , Enzyme Inhibitors/pharmacology , Heart/embryology , Histone Deacetylase 2 , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Hypertension/complications , Hypertension/pathology , Hypertension/physiopathology , MEF2 Transcription Factors , Male , Mice , Mice, Knockout , Mutation/genetics , Myocardium/enzymology , Myocardium/pathology , Myogenic Regulatory Factors , Phosphorylation , Phosphotransferases/genetics , Phosphotransferases/metabolism , Stress, Physiological/genetics , Stress, Physiological/physiopathology , Transcription Factors/genetics
20.
J Biol Chem ; 278(31): 28930-7, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12761226

ABSTRACT

Postnatal cardiac myocytes respond to stress signals by hypertrophic growth and activation of a fetal gene program. Recently, we showed that class II histone deacetylases (HDACs) suppress cardiac hypertrophy, and mice lacking the class II HDAC, HDAC9, are sensitized to hypertrophic signals. To further define the roles of HDACs in cardiac hypertrophy, we analyzed the effects of HDAC inhibitors on the responsiveness of primary cardiomyocytes to hypertrophic agonists. Paradoxically, HDAC inhibitors imposed a dose-dependent blockade to hypertrophy and fetal gene activation. We conclude that distinct HDACs play positive or negative roles in the control of cardiomyocyte hypertrophy. HDAC inhibitors are currently being tested in clinical trials as anti-cancer agents. Our results suggest that these inhibitors may also hold promising clinical value as therapeutics for cardiac hypertrophy and heart failure.


Subject(s)
Cardiomegaly/prevention & control , Enzyme Inhibitors/administration & dosage , Histone Deacetylase Inhibitors , Adenylate Kinase/metabolism , Animals , Animals, Newborn , Atrial Natriuretic Factor/analysis , Atrial Natriuretic Factor/genetics , Cells, Cultured , DNA/analysis , Dose-Response Relationship, Drug , Fetus , Fluorescent Antibody Technique, Indirect , Gene Expression/drug effects , Histone Deacetylases/physiology , Leucine/metabolism , Myocardium/chemistry , Myocardium/pathology , Rats , Rats, Sprague-Dawley , Tritium , Ventricular Myosins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL