Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cell Stem Cell ; 29(5): 840-855.e7, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35395180

ABSTRACT

Hypoplastic left heart syndrome (HLHS) is a severe congenital heart disease with 30% mortality from heart failure (HF) in the first year of life, but the cause of early HF remains unknown. Induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CM) from patients with HLHS showed that early HF is associated with increased apoptosis, mitochondrial respiration defects, and redox stress from abnormal mitochondrial permeability transition pore (mPTP) opening and failed antioxidant response. In contrast, iPSC-CM from patients without early HF showed normal respiration with elevated antioxidant response. Single-cell transcriptomics confirmed that early HF is associated with mitochondrial dysfunction accompanied with endoplasmic reticulum (ER) stress. These findings indicate that uncompensated oxidative stress underlies early HF in HLHS. Importantly, mitochondrial respiration defects, oxidative stress, and apoptosis were rescued by treatment with sildenafil to inhibit mPTP opening or TUDCA to suppress ER stress. Together these findings point to the potential use of patient iPSC-CM for modeling clinical heart failure and the development of therapeutics.


Subject(s)
Heart Defects, Congenital , Heart Failure , Induced Pluripotent Stem Cells , Antioxidants/metabolism , Heart Defects, Congenital/metabolism , Heart Failure/metabolism , Humans , Mitochondrial Permeability Transition Pore , Myocytes, Cardiac/metabolism , Oxidative Stress
2.
Biomolecules ; 10(8)2020 08 14.
Article in English | MEDLINE | ID: mdl-32823934

ABSTRACT

Background: Cilia are actin based cellular protrusions conserved from algae to complex multicellular organisms like Homo sapiens. Respiratory motile cilia line epithelial cells of the tracheobronchial tree, beat in a synchronous, metachronal wave, moving inhaled pollutants and pathogens cephalad. Their role in both congenital disorders like primary ciliary dyskinesia (PCD) to acquired disorders like chronic obstructive pulmonary disease (COPD) continues to evolve. In this current body of work we outline a protocol optimized to reciliate human nasal epithelial cells and mouse tracheal cells in vitro. Using this protocol, we knocked down known cilia genes, as well as use a small molecule inhibitor of Notch, N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl Ester (DAPT), to assess the effect of these on ciliogenesis in order to show the validity of our protocol. Methods: Tracheas were harvested from wild-type, adult C57B6 mice, pronase digested and sloughed off epithelial cells grown to confluence in stationary culture on rat-tail collagen coated wells. Upon reaching confluence, collagen was digested and cells placed suspension culture protocol to reciliate the cells. Using this suspension culture protocol, we employed siRNA gene knockdown to assay gene functions required for airway ciliogenesis. Knock down of Dynein axonemal heavy chain 5 (Dnah5), a ciliary structural protein, was confirmed using immunostaining. Mouse tracheal cells were treated in suspension with varying doses of DAPT, an inhibitor of Notch, with the purpose of evaluating its effect and dose response on ciliogenesis. The optimum dose was then used on reciliating human nasal epithelial cells. Results: siRNA knockdown of Foxj1 prevented ciliation, consistent with its role as a master regulator of motile cilia. Knockdown of Dnai1 and Dnah5 resulted in immotile cilia, and Cand1 knockdown, a centrosome protein known to regulate centrosome amplification, inhibited airway ciliogenesis. Dnah5 knockdown was confirmed with significantly decreased immunostaining of cilia for this protein. Inhibiting Notch signaling by inhibiting gamma secretase with DAPT enhanced the percentage of ciliation, and resulted in longer cilia that beat with higher frequency in both mouse and human airway epithelia. Conclusions: Modifying existing reciliation protocols to suit both human nasal epithelial and mouse tracheal tissue, we have shown that knockdown of known cilia-related genes have the expected effects. Additionally, we have demonstrated the optimal dosage for significantly improving reciliation of airway epithelia using DAPT. Given that cilia length and function are significantly compromised in COPD, these findings open up interesting avenues for further exploration.


Subject(s)
Cilia/metabolism , Dipeptides/pharmacology , Nose/cytology , Trachea/cytology , Animals , Axonemal Dyneins/genetics , Cell Culture Techniques , Cell Differentiation/drug effects , Cells, Cultured , Cilia/drug effects , Cilia/genetics , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Forkhead Transcription Factors/genetics , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Nose/drug effects , Trachea/drug effects , Trachea/metabolism , Transcription Factors/genetics
3.
Commun Biol ; 3(1): 312, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32546759

ABSTRACT

The recent recovery of mutations in vesicular trafficking genes causing congenital heart disease (CHD) revealed an unexpected role for the endocytic pathway. We now show that mice with a C4232R missense mutation in Low density lipoprotein receptor related protein 1 (LRP1) exhibit atrioventricular septal defects with double outlet right ventricle. Lrp1m/m mice exhibit shortened outflow tracts (OFT) and dysmorphic hypocellular cushions with reduced proliferation and increased apoptosis. Lrp1m/m embryonic fibroblasts show decreased cell motility and focal adhesion turnover associated with retention of mutant LRP1 in endoplasmic reticulum and reduced LRP1 expression. Conditional deletion of Lrp1 in cardiac neural crest cells (CNC) replicates the full CHD phenotype. Cushion explants showed defective cell migration, with gene expression analysis indicating perturbation of Wnt and other signaling pathways. Thus, LRP1 function in CNCs is required for normal OFT development with other cell lineages along the CNC migratory path playing a supporting role.


Subject(s)
Heart Defects, Congenital/genetics , Heart/embryology , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Mutation, Missense , Neural Crest/cytology , Animals , Cell Lineage , Cell Movement/genetics , Female , Gene Expression Regulation, Developmental , Heart/diagnostic imaging , Heart Defects, Congenital/pathology , Heart Septal Defects/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardium/cytology
4.
Biomolecules ; 8(4)2018 11 14.
Article in English | MEDLINE | ID: mdl-30441852

ABSTRACT

Our previous work identified a 12-amino acid peptide that targets the heart, termed cardiac targeting peptide (CTP). We now quantitatively assess the bio-distribution of CTP, show a clinical application with the imaging of the murine heart, and study its mechanisms of transduction. Bio-distribution studies of cyanine5.5-N-Hydroxysuccinimide (Cy5.5) labeled CTP were undertaken in wild-type mice. Cardiac targeting peptide was labeled with Technetium 99m (99mTc) using the chelator hydrazino-nicotinamide (HYNIC), and imaging performed using micro-single photon emission computerized tomography/computerized tomography (SPECT/CT). Human-induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMCs) were incubated with dual-labeled CTP, and imaged using confocal microscopy. TriCEPs technology was utilized to study the mechanism of transduction. Bio-distribution studies showed peak uptake of CTP at 15 min. 99mTc-HYNIC-CTP showed heart-specific uptake. Robust transduction of beating human iPSC-derived CMCs was seen. TriCEPs experiments revealed five candidate binding partners for CTP, with Kcnh5 being felt to be the most likely candidate as it showed a trend towards being competed out by siRNA knockdown. Transduction efficiency was enhanced by increasing extracellular potassium concentration, and with Quinidine, a Kcnh5 inhibitor, that blocks the channel in an open position. We demonstrate that CTP transduces the normal heart as early as 15 min. 99mTc-HYNIC-CTP targets the normal murine heart with substantially improved targeting compared with 99mTc Sestamibi. Cardiac targeting peptide's transduction ability is not species limited and has human applicability. Cardiac targeting peptide appears to utilize Kcnh5 to gain cell entry, a phenomenon that is affected by pre-treatment with Quinidine and changes in potassium levels.


Subject(s)
Myocardium/metabolism , Peptides/metabolism , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Transduction, Genetic , Animals , Humans , Induced Pluripotent Stem Cells/metabolism , Ligands , Mice , Myocytes, Cardiac/metabolism , RNA, Small Interfering/metabolism , Technetium/chemistry , Tissue Distribution , Transferrin/metabolism
5.
Methods Cell Biol ; 132: 109-26, 2016.
Article in English | MEDLINE | ID: mdl-26928541

ABSTRACT

We describe methods based on live cell fluorescent microscopy and mass spectrometry to characterize the mechanism of endosomal cAMP production and its regulation using the parathyroid hormone (PTH) type 1 receptor as a prime example. These methods permit to measure rapid changes of cAMP levels in response to PTH, kinetics of endosomal ligand-receptor interaction, pH changes associated with receptor trafficking, and to identify the endosomal receptor interactome.


Subject(s)
Cyclic AMP/biosynthesis , Receptor, Parathyroid Hormone, Type 1/metabolism , Second Messenger Systems , Amino Acid Sequence , Endocytosis , Endosomes/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Protein Transport , Receptor, Parathyroid Hormone, Type 1/chemistry
6.
Neurobiol Dis ; 73: 70-82, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25281317

ABSTRACT

Current research on Parkinson's disease (PD) pathogenesis requires relevant animal models that mimic the gradual and progressive development of neuronal dysfunction and degeneration that characterizes the disease. Polymorphisms in engrailed 1 (En1), a homeobox transcription factor that is crucial for both the development and survival of mesencephalic dopaminergic neurons, are associated with sporadic PD. This suggests that En1 mutant mice might be a promising candidate PD model. Indeed, a mouse that lacks one En1 allele exhibits decreased mitochondrial complex I activity and progressive midbrain dopamine neuron degeneration in adulthood, both features associated with PD. We aimed to further characterize the disease-like phenotype of these En1(+/-) mice with a focus on early neurodegenerative changes that can be utilized to score efficacy of future disease modifying studies. We observed early terminal defects in the dopaminergic nigrostriatal pathway in En1(+/-) mice. Several weeks before a significant loss of dopaminergic neurons in the substantia nigra could be detected, we found that striatal terminals expressing high levels of dopaminergic neuron markers TH, VMAT2, and DAT were dystrophic and swollen. Using transmission electron microscopy, we identified electron dense bodies consistent with abnormal autophagic vacuoles in these terminal swellings. In line with these findings, we detected an up-regulation of the mTOR pathway, concurrent with a downregulation of the autophagic marker LC3B, in ventral midbrain and nigral dopaminergic neurons of the En1(+/-) mice. This supports the notion that autophagic protein degradation is reduced in the absence of one En1 allele. We imaged the nigrostriatal pathway using the CLARITY technique and observed many fragmented axons in the medial forebrain bundle of the En1(+/-) mice, consistent with axonal maintenance failure. Using in vivo electrochemistry, we found that nigrostriatal terminals in the dorsal striatum were severely deficient in dopamine release and reuptake. Our findings support a progressive retrograde degeneration of En1(+/-) nigrostriatal neurons, akin to what is suggested to occur in PD. We suggest that using the En1(+/-) mice as a model will provide further key insights into PD pathogenesis, and propose that axon terminal integrity and function can be utilized to estimate dopaminergic neuron health and efficacy of experimental PD therapies.


Subject(s)
Corpus Striatum/metabolism , Corpus Striatum/pathology , Homeodomain Proteins/genetics , Nerve Degeneration/etiology , Parkinson Disease , Substantia Nigra/pathology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Autophagy/genetics , Disease Models, Animal , Disease Progression , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Dopaminergic Neurons/ultrastructure , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homovanillic Acid/metabolism , Mice , Mice, Transgenic , Parkinson Disease/complications , Parkinson Disease/genetics , Parkinson Disease/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Substantia Nigra/metabolism , TOR Serine-Threonine Kinases/metabolism , Time Factors , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
7.
Methods Mol Biol ; 1066: 121-9, 2013.
Article in English | MEDLINE | ID: mdl-23955739

ABSTRACT

Förster resonance energy transfer (FRET) is a proximity-dependent quantum effect that allows the measurement of protein interactions and conformational changes which are invisible to traditional forms of fluorescence or electron microscopy. However, FRET experiments often have difficulty detecting interactions that are transient and localized or occur in low abundance against a large background. This protocol describes a method of improving on the sensitivity and quantifiability of FRET experiments by using time-specific detection to isolate FRET-mediated acceptor emission from cross-talk excitation and all other sources of nonspecific fluorescence background.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Protein Interaction Mapping/methods , Cell Communication/physiology , Cell Line , HEK293 Cells , Humans , Protein Conformation , Receptors, G-Protein-Coupled/metabolism
8.
J Biol Chem ; 288(39): 27849-60, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-23935101

ABSTRACT

The vasopressin type 2 receptor (V2R) is a critical G protein-coupled receptor (GPCR) for vertebrate physiology, including the balance of water and sodium ions. It is unclear how its two native hormones, vasopressin (VP) and oxytocin (OT), both stimulate the same cAMP/PKA pathway yet produce divergent antinatriuretic and antidiuretic effects that are either strong (VP) or weak (OT). Here, we present a new mechanism that differentiates the action of VP and OT on V2R signaling. We found that vasopressin, as opposed to OT, continued to generate cAMP and promote PKA activation for prolonged periods after ligand washout and receptor internalization in endosomes. Contrary to the classical model of arrestin-mediated GPCR desensitization, arrestins bind the VP-V2R complex yet extend rather than shorten the generation of cAMP. Signaling is instead turned off by the endosomal retromer complex. We propose that this mechanism explains how VP sustains water and Na(+) transport in renal collecting duct cells. Together with recent work on the parathyroid hormone receptor, these data support the existence of a novel "noncanonical" regulatory pathway for GPCR activation and response termination, via the sequential action of ß-arrestin and the retromer complex.


Subject(s)
Arrestins/metabolism , Gene Expression Regulation , Receptors, Vasopressin/metabolism , Signal Transduction , Animals , Antidiuretic Agents/pharmacology , Aquaporin 2/metabolism , Cell Membrane/metabolism , Cyclic AMP/metabolism , Dogs , Endosomes/metabolism , HEK293 Cells , Humans , Kidney/metabolism , Ligands , Madin Darby Canine Kidney Cells , Oxytocin/chemistry , Phosphorylation , Receptors, G-Protein-Coupled/metabolism , Sodium/metabolism , beta-Arrestins
9.
Methods Enzymol ; 522: 337-63, 2013.
Article in English | MEDLINE | ID: mdl-23374192

ABSTRACT

We describe optical and microscopy methods based on Förster resonance energy transfer, fluorescence recovery after photobleaching, and imaging cross-correlation spectroscopy that permit to determine kinetic and dynamic properties of key reactions involved G protein-coupled receptor (GPCR) signaling from the initial ligand binding step to the generation of the second messenger, cAMP. Well suited to determine rate-limiting reactions taking place along a GPCR signaling cascade in live cells, these techniques have also uncovered new concepts in GPCR signaling as well as many interesting mechanistic subtleties by which GPCRs transmit neurotransmitter and hormone signals into cells.


Subject(s)
Cyclic AMP/metabolism , Fluorescence Resonance Energy Transfer/methods , Heterotrimeric GTP-Binding Proteins/metabolism , Parathyroid Hormone/metabolism , Receptors, Parathyroid Hormone/metabolism , Signal Transduction/genetics , Arrestins/genetics , Arrestins/metabolism , Gene Expression , HEK293 Cells , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Kinetics , Ligands , Microscopy, Confocal , Microscopy, Fluorescence , Photobleaching , Protein Binding , Protein Stability , Receptors, Parathyroid Hormone/genetics
10.
Proc Natl Acad Sci U S A ; 110(4): 1530-5, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23297229

ABSTRACT

G protein-coupled receptors (GPCRs) participate in ubiquitous transmembrane signal transduction processes by activating heterotrimeric G proteins. In the current "canonical" model of GPCR signaling, arrestins terminate receptor signaling by impairing receptor-G-protein coupling and promoting receptor internalization. However, parathyroid hormone receptor type 1 (PTHR), an essential GPCR involved in bone and mineral metabolism, does not follow this conventional desensitization paradigm. ß-Arrestins prolong G protein (G(S))-mediated cAMP generation triggered by PTH, a process that correlates with the persistence of arrestin-PTHR complexes on endosomes and which is thought to be associated with prolonged physiological calcemic and phosphate responses. This presents an inescapable paradox for the current model of arrestin-mediated receptor-G-protein decoupling. Here we show that PTHR forms a ternary complex that includes arrestin and the Gßγ dimer in response to PTH stimulation, which in turn causes an accelerated rate of G(S) activation and increases the steady-state levels of activated G(S), leading to prolonged generation of cAMP. This work provides the mechanistic basis for an alternative model of GPCR signaling in which arrestins contribute to sustaining the effect of an agonist hormone on the receptor.


Subject(s)
Arrestins/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism , Arrestins/chemistry , Cyclic AMP/biosynthesis , Fluorescence Resonance Energy Transfer , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein gamma Subunits/chemistry , HEK293 Cells , Humans , Kinetics , Models, Biological , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Parathyroid Hormone/metabolism , Parathyroid Hormone/pharmacology , Receptor, Parathyroid Hormone, Type 1/chemistry , Receptors, G-Protein-Coupled/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction , beta-Arrestins
11.
Trends Pharmacol Sci ; 33(8): 423-31, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22709554

ABSTRACT

The classical model of arrestin-mediated desensitization of cell-surface G-protein-coupled receptors (GPCRs) is thought to be universal. However, this paradigm is incompatible with recent reports that the parathyroid hormone (PTH) receptor (PTHR), a crucial GPCR for bone and mineral ion metabolism, sustains G(S) activity and continues to generate cAMP for prolonged periods after ligand washout; during these periods the receptor is observed mainly in endosomes, associated with the bound ligand, G(S) and ß-arrestins. In this review we discuss possible molecular mechanisms underlying sustained signaling by the PTHR, including modes of signal generation and attenuation within endosomes, as well as the biological relevance of such non-canonical signaling.


Subject(s)
Receptors, Parathyroid Hormone/chemistry , Signal Transduction , Animals , Arrestins/metabolism , Cyclic AMP/metabolism , Endosomes/metabolism , GTP-Binding Proteins/metabolism , Humans , Protein Conformation , Receptors, Parathyroid Hormone/metabolism , beta-Arrestins
12.
Proc Natl Acad Sci U S A ; 109(19): 7433-8, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22538810

ABSTRACT

Parathyroid hormone (PTH), the major calcium-regulating hormone, and norepinephrine (NE), the principal neurotransmitter of sympathetic nerves, regulate bone remodeling by activating distinct cell-surface G protein-coupled receptors in osteoblasts: the parathyroid hormone type 1 receptor (PTHR) and the ß(2)-adrenergic receptor (ß(2)AR), respectively. These receptors activate a common cAMP/PKA signal transduction pathway mediated through the stimulatory heterotrimeric G protein. Activation of ß(2)AR via the sympathetic nervous system decreases bone formation and increases bone resorption. Conversely, daily injection of PTH (1-34), a regimen known as intermittent (i)PTH treatment, increases bone mass through the stimulation of trabecular and cortical bone formation and decreases fracture incidences in severe cases of osteoporosis. Here, we show that iPTH has no osteoanabolic activity in mice lacking the ß(2)AR. ß(2)AR deficiency suppressed both iPTH-induced increase in bone formation and resorption. We showed that the lack of ß(2)AR blocks expression of iPTH-target genes involved in bone formation and resorption that are regulated by the cAMP/PKA pathway. These data implicate an unexpected functional interaction between PTHR and ß(2)AR, two G protein-coupled receptors from distinct families, which control bone formation and PTH anabolism.


Subject(s)
Bone and Bones/drug effects , Parathyroid Hormone/pharmacology , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Absorptiometry, Photon , Anabolic Agents/metabolism , Anabolic Agents/pharmacology , Animals , Bone Density/drug effects , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Female , Femur/drug effects , Femur/metabolism , Fluoresceins , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Osteogenesis/drug effects , Osteogenesis/genetics , Parathyroid Hormone/metabolism , Receptor, Parathyroid Hormone, Type 1/genetics , Receptors, Adrenergic, beta-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , X-Ray Microtomography
13.
Proc Natl Acad Sci U S A ; 108(43): 17767-72, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21990347

ABSTRACT

The sympathetic nervous system suppresses bone mass by mechanisms that remain incompletely elucidated. Using cell-based and murine genetics approaches, we show that this activity of the sympathetic nervous system requires osteopontin (OPN), a cytokine and one of the major members of the noncollagenous extracellular matrix proteins of bone. In this work, we found that the stimulation of the sympathetic tone by isoproterenol increased the level of OPN expression in the plasma and bone and that mice lacking OPN (OPN-KO) suppressed the isoproterenol-induced bone loss by preventing reduced osteoblastic and enhanced osteoclastic activities. In addition, we found that OPN is necessary for changes in the expression of genes related to bone resorption and bone formation that are induced by activation of the sympathetic tone. At the cellular level, we showed that intracellular OPN modulated the capacity of the ß2-adrenergic receptor to generate cAMP with a corresponding modulation of cAMP-response element binding (CREB) phosphorylation and associated transcriptional events inside the cell. Our results indicate that OPN plays a critical role in sympathetic tone regulation of bone mass and that this OPN regulation is taking place through modulation of the ß2-adrenergic receptor/cAMP signaling system.


Subject(s)
Bone and Bones/physiology , Osteopontin/metabolism , Sympathetic Nervous System/physiology , Analysis of Variance , Animals , Bone and Bones/metabolism , Cyclic AMP/metabolism , Fluorescence Resonance Energy Transfer , Isoproterenol/pharmacology , Mice , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteopontin/deficiency , Receptors, Adrenergic, beta-2/metabolism , Sympathetic Nervous System/drug effects
14.
Nat Chem Biol ; 7(5): 278-84, 2011 May.
Article in English | MEDLINE | ID: mdl-21445058

ABSTRACT

The generation of cAMP by G protein-coupled receptors (GPCRs) and its termination are currently thought to occur exclusively at the plasma membrane of cells. Under existing models of receptor regulation, this signal is primarily restricted by desensitization of the receptors through their binding to ß-arrestins. However, this paradigm is not consistent with recent observations that the parathyroid hormone receptor type 1 (PTHR) continues to stimulate cAMP production even after receptor internalization, as ß-arrestins are known to rapidly bind and internalize activated PTHR. Here we show that binding to ß-arrestin1 prolongs rather than terminates the generation of cAMP by PTHR, and that cAMP generation correlates with the persistence of arrestin-receptor complexes on endosomes. PTHR signaling is instead turned off by the retromer complex, which regulates the movement of internalized receptor from endosomes to the Golgi apparatus. Thus, binding by the retromer complex regulates the sustained generation of cAMP triggered by an internalized GPCR.


Subject(s)
Cyclic AMP/metabolism , Endosomes/metabolism , Golgi Apparatus/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Arrestins/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Models, Biological , Receptor, Parathyroid Hormone, Type 1/genetics , Signal Transduction , Time Factors , beta-Arrestins
15.
Nat Chem Biol ; 5(10): 734-42, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19701185

ABSTRACT

Cell signaling mediated by the G protein-coupled parathyroid hormone receptor type 1 (PTHR) is fundamental to bone and kidney physiology. It has been unclear how the two ligand systems--PTH, endocrine and homeostatic, and PTH-related peptide (PTHrP), paracrine--can effectively operate with only one receptor and trigger different durations of the cAMP responses. Here we analyze the ligand response by measuring the kinetics of activation and deactivation for each individual reaction step along the PTHR signaling cascade. We found that during the time frame of G protein coupling and cAMP production, PTHrP(1-36) action was restricted to the cell surface, whereas PTH(1-34) had moved to internalized compartments where it remained associated with the PTHR and Galpha(s), potentially as a persistent and active ternary complex. Such marked differences suggest a mechanism by which PTH and PTHrP induce differential responses, and these results indicate that the central tenet that cAMP production originates exclusively at the cell membrane must be revised.


Subject(s)
Cyclic AMP/biosynthesis , Endocytosis/physiology , Receptor, Parathyroid Hormone, Type 1/physiology , Signal Transduction/physiology , Animals , Bone Resorption/metabolism , Cell Line , Cell Membrane/metabolism , Fluorescence Resonance Energy Transfer , GTP-Binding Proteins/metabolism , Humans , Kinetics , Ligands , Mice , Microscopy, Confocal , Osteoblasts/metabolism , Parathyroid Hormone/metabolism , Protein Conformation , Protein Transport , Receptor, Parathyroid Hormone, Type 1/agonists , Receptor, Parathyroid Hormone, Type 1/metabolism
16.
Mol Endocrinol ; 23(5): 590-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19196832

ABSTRACT

Many biochemical pathways are driven by G protein-coupled receptors, cell surface proteins that convert the binding of extracellular chemical, sensory, and mechanical stimuli into cellular signals. Their interaction with various ligands triggers receptor activation that typically couples to and activates heterotrimeric G proteins, which in turn control the propagation of secondary messenger molecules (e.g. cAMP) involved in critically important physiological processes (e.g. heart beat). Successful transfer of information from ligand binding events to intracellular signaling cascades involves a dynamic interplay between ligands, receptors, and G proteins. The development of Förster resonance energy transfer and bioluminescence resonance energy transfer-based methods has now permitted the kinetic analysis of initial steps involved in G protein-coupled receptor-mediated signaling in live cells and in systems as diverse as neurotransmitter and hormone signaling. The direct measurement of ligand efficacy at the level of the receptor by Förster resonance energy transfer is also now possible and allows intrinsic efficacies of clinical drugs to be linked with the effect of receptor polymorphisms.


Subject(s)
GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Animals , Fluorescence Resonance Energy Transfer , GTP-Binding Proteins/chemistry , Humans , Kinetics , Luminescent Measurements , Models, Biological , Receptors, G-Protein-Coupled/chemistry
17.
Mol Biol Cell ; 19(7): 2696-707, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18434598

ABSTRACT

Recent work indicates that mitogen-activated protein kinase kinase (MEK)1 signaling at the G2/M cell cycle transition unlinks the contiguous mammalian Golgi apparatus and that this regulates cell cycle progression. Here, we sought to determine the role in this pathway of Golgi reassembly protein (GRASP)55, a Golgi-localized target of MEK/extracellular signal-regulated kinase (ERK) phosphorylation at mitosis. In support of the hypothesis that GRASP55 is inhibited in late G2 phase, causing unlinking of the Golgi ribbon, we found that HeLa cells depleted of GRASP55 show a fragmented Golgi similar to control cells arrested in G2 phase. In the absence of GRASP55, Golgi stack length is shortened but Golgi stacking, compartmentalization, and transport seem normal. Absence of GRASP55 was also sufficient to suppress the requirement for MEK1 in the G2/M transition, a requirement that we previously found depends on an intact Golgi ribbon. Furthermore, mimicking mitotic phosphorylation of GRASP55 by using aspartic acid substitutions is sufficient to unlink the Golgi apparatus in a gene replacement assay. Our results implicate MEK1/ERK regulation of GRASP55-mediated Golgi linking as a control point in cell cycle progression.


Subject(s)
Golgi Apparatus/metabolism , Membrane Proteins/physiology , CDC2 Protein Kinase/metabolism , Cell Cycle , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Golgi Matrix Proteins , HeLa Cells , Humans , Image Processing, Computer-Assisted , Membrane Proteins/metabolism , Microscopy, Electron, Transmission , Mitosis , Models, Biological , Phosphorylation , Transfection
18.
Mol Biol Cell ; 18(2): 594-604, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17182854

ABSTRACT

Two controversies have emerged regarding the signaling pathways that regulate Golgi disassembly at the G(2)/M cell cycle transition. The first controversy concerns the role of mitogen-activated protein kinase activator mitogen-activated protein kinase kinase (MEK)1, and the second controversy concerns the participation of Golgi structure in a novel cell cycle "checkpoint." A potential simultaneous resolution is suggested by the hypothesis that MEK1 triggers Golgi unlinking in late G(2) to control G(2)/M kinetics. Here, we show that inhibition of MEK1 by RNA interference or by using the MEK1/2-specific inhibitor U0126 delayed the passage of synchronized HeLa cells into M phase. The MEK1 requirement for normal mitotic entry was abrogated if Golgi proteins were dispersed before M phase by treatment of cells with brefeldin A or if GRASP65, which links Golgi stacks into a ribbon network, was depleted. Imaging revealed that unlinking of the Golgi apparatus begins before M phase, is independent of cyclin-dependent kinase 1 activation, and requires MEK signaling. Furthermore, expression of the GRASP family member GRASP55 after alanine substitution of its MEK1-dependent mitotic phosphorylation sites inhibited both late G(2) Golgi unlinking and the G(2)/M transition. Thus, MEK1 plays an in vivo role in Golgi reorganization, which regulates cell cycle progression.


Subject(s)
Golgi Apparatus/enzymology , MAP Kinase Kinase 1/physiology , Mitosis , Cell Division/genetics , G2 Phase/genetics , Golgi Matrix Proteins , HeLa Cells , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , Membrane Proteins/metabolism , Mitosis/genetics , Phosphorylation , RNA Interference
19.
Biochemistry ; 44(36): 12188-95, 2005 Sep 13.
Article in English | MEDLINE | ID: mdl-16142917

ABSTRACT

Four recombinant mutants of human fetal hemoglobin [Hb F (alpha2gamma2)] with amino acid substitutions at the position 43 of the gamma-chain, rHb (gammaD43L), rHb (gammaD43E), rHb (gammaD43W), and rHb (gammaD43R), have been expressed in our Escherichia coli expression system and used to investigate their inhibitory effect on the polymerization of deoxygenated sickle cell hemoglobin (Hb S). Oxygen-binding studies show that rHb (gammaD43E), rHb (gammaD43W), and rHb (gammaD43R) exhibit higher oxygen affinity than human normal adult hemoglobin (Hb A), Hb F, or rHb (gammaD43L), and all four rHbs are cooperative in binding O2. Proton nuclear magnetic resonance (NMR) studies of these four rHbs indicate that the quaternary and tertiary structures around the heme pockets are similar to those of Hb F in both deoxy (T) and liganded (R) states. Solution light-scattering experiments indicate that these mutants remain mostly tetrameric in the liganded (R) state. In equimolar mixtures of Hb S and each of the four rHb mutants (gammaD43L, gammaD43E, gammaD43R, and gammaD43W), the solubility (Csat) of each of the pairs of Hbs is higher than that of a similar mixture of Hb S and Hb A, as measured by dextran-Csat experiments. Furthermore, the Csat values for Hb S/rHb (gammaD43L), Hb S/rHb (gammaD43E), and Hb S/rHb (gammaD43R) mixtures are substantially higher than that for Hb S/Hb F. The results suggest that these three mutants of Hb F are more effective than Hb F in inhibiting the polymerization of deoxy-Hb S in equimolar mixtures.


Subject(s)
Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Hemoglobin, Sickle/chemistry , Hemoglobin, Sickle/metabolism , Mutation/genetics , Dextrans , Fetal Hemoglobin/pharmacology , Humans , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Oxygen/metabolism , Protein Structure, Quaternary/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...