Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(10): 1661-1675.e16, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35483373

ABSTRACT

ß-arrestins bind G protein-coupled receptors to terminate G protein signaling and to facilitate other downstream signaling pathways. Using single-molecule fluorescence resonance energy transfer imaging, we show that ß-arrestin is strongly autoinhibited in its basal state. Its engagement with a phosphopeptide mimicking phosphorylated receptor tail efficiently releases the ß-arrestin tail from its N domain to assume distinct conformations. Unexpectedly, we find that ß-arrestin binding to phosphorylated receptor, with a phosphorylation barcode identical to the isolated phosphopeptide, is highly inefficient and that agonist-promoted receptor activation is required for ß-arrestin activation, consistent with the release of a sequestered receptor C tail. These findings, together with focused cellular investigations, reveal that agonism and receptor C-tail release are specific determinants of the rate and efficiency of ß-arrestin activation by phosphorylated receptor. We infer that receptor phosphorylation patterns, in combination with receptor agonism, synergistically establish the strength and specificity with which diverse, downstream ß-arrestin-mediated events are directed.


Subject(s)
Phosphopeptides , Receptors, G-Protein-Coupled , Phosphopeptides/metabolism , Phosphorylation , Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 1/metabolism , beta-Arrestins/metabolism
2.
Mol Cell ; 82(16): 3089-3102.e7, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35931084

ABSTRACT

The ß2-adrenergic receptor (ß2AR), a prototypic G-protein-coupled receptor (GPCR), is a powerful driver of bronchorelaxation, but the effectiveness of ß-agonist drugs in asthma is limited by desensitization and tachyphylaxis. We find that during activation, the ß2AR is modified by S-nitrosylation, which is essential for both classic desensitization by PKA as well as desensitization of NO-based signaling that mediates bronchorelaxation. Strikingly, S-nitrosylation alone can drive ß2AR internalization in the absence of traditional agonist. Mutant ß2AR refractory to S-nitrosylation (Cys265Ser) exhibits reduced desensitization and internalization, thereby amplifying NO-based signaling, and mice with Cys265Ser mutation are resistant to bronchoconstriction, inflammation, and the development of asthma. S-nitrosylation is thus a central mechanism in ß2AR signaling that may be operative widely among GPCRs and targeted for therapeutic gain.


Subject(s)
Asthma , Animals , Asthma/chemically induced , Asthma/genetics , Mice , Signal Transduction
3.
Nat Chem Biol ; 19(6): 687-694, 2023 06.
Article in English | MEDLINE | ID: mdl-36646958

ABSTRACT

G protein-coupled receptors (GPCRs) selectively activate at least one of the four families of heterotrimeric G proteins, but the mechanism of coupling selectivity remains unclear. Structural studies emphasize structural complementarity of GPCRs and nucleotide-free G proteins, but selectivity is likely to be determined by transient intermediate-state complexes that exist before nucleotide release. Here we study coupling to nucleotide-decoupled G protein variants that can adopt conformations similar to receptor-bound G proteins without releasing nucleotide, and are therefore able to bypass intermediate-state complexes. We find that selectivity is degraded when nucleotide release is not required for GPCR-G protein complex formation, to the extent that most GPCRs interact with most nucleotide-decoupled G proteins. These findings demonstrate the absence of absolute structural incompatibility between noncognate receptor-G protein pairs, and are consistent with the hypothesis that transient intermediate states are partly responsible for coupling selectivity.


Subject(s)
Heterotrimeric GTP-Binding Proteins , Receptors, G-Protein-Coupled , Receptors, G-Protein-Coupled/metabolism , Protein Conformation , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Article in English | MEDLINE | ID: mdl-33990469

ABSTRACT

G protein-coupled receptors (GPCRs) are gatekeepers of cellular homeostasis and the targets of a large proportion of drugs. In addition to their signaling activity at the plasma membrane, it has been proposed that their actions may result from translocation and activation of G proteins at endomembranes-namely endosomes. This could have a significant impact on our understanding of how signals from GPCR-targeting drugs are propagated within the cell. However, little is known about the mechanisms that drive G protein movement and activation in subcellular compartments. Using bioluminescence resonance energy transfer (BRET)-based effector membrane translocation assays, we dissected the mechanisms underlying endosomal Gq trafficking and activity following activation of Gq-coupled receptors, including the angiotensin II type 1, bradykinin B2, oxytocin, thromboxane A2 alpha isoform, and muscarinic acetylcholine M3 receptors. Our data reveal that GPCR-promoted activation of Gq at the plasma membrane induces its translocation to endosomes independently of ß-arrestin engagement and receptor endocytosis. In contrast, Gq activity at endosomes was found to rely on both receptor endocytosis-dependent and -independent mechanisms. In addition to shedding light on the molecular processes controlling subcellular Gq signaling, our study provides a set of tools that will be generally applicable to the study of G protein translocation and activation at endosomes and other subcellular organelles, as well as the contribution of signal propagation to drug action.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques/methods , Endocytosis/physiology , Endosomes/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Receptors, G-Protein-Coupled/physiology , HEK293 Cells , Humans , Rho Guanine Nucleotide Exchange Factors/physiology , Signal Transduction/physiology , beta-Arrestins/physiology
5.
J Magn Reson Imaging ; 2023 Oct 03.
Article in English | MEDLINE | ID: mdl-37787109

ABSTRACT

BACKGROUND: 1 H-magnetic resonance spectroscopy (1 H-MRS) may provide a direct index for the testing of medicines for neuroprotection and drug mechanisms in multiple sclerosis (MS) through measures of total N-acetyl-aspartate (tNAA), total creatine (tCr), myo-inositol (mIns), total-choline (tCho), and glutamate + glutamine (Glx). Neurometabolites may be associated with clinical disability with evidence that baseline neuroaxonal integrity is associated with upper limb function and processing speed in secondary progressive MS (SPMS). PURPOSE: To assess the effect on neurometabolites from three candidate drugs after 96-weeks as seen by 1 H-MRS and their association with clinical disability in SPMS. STUDY-TYPE: Longitudinal. POPULATION: 108 participants with SPMS randomized to receive neuroprotective drugs amiloride [mean age 55.4 (SD 7.4), 61% female], fluoxetine [55.6 (6.6), 71%], riluzole [54.6 (6.3), 68%], or placebo [54.8 (7.9), 67%]. FIELD STRENGTH/SEQUENCE: 3-Tesla. Chemical-shift-imaging 2D-point-resolved-spectroscopy (PRESS), 3DT1. ASSESSMENT: Brain metabolites in normal appearing white matter (NAWM) and gray matter (GM), brain volume, lesion load, nine-hole peg test (9HPT), and paced auditory serial addition test were measured at baseline and at 96-weeks. STATISTICAL TESTS: Paired t-test was used to analyze metabolite changes in the placebo arm over 96-weeks. Metabolite differences between treatment arms and placebo; and associations between baseline metabolites and upper limb function/information processing speed at 96-weeks assessed using multiple linear regression models. P-value<0.05 was considered statistically significant. RESULTS: In the placebo arm, tCho increased in GM (mean difference = -0.32 IU) but decreased in NAWM (mean difference = 0.13 IU). Compared to placebo, in the fluoxetine arm, mIns/tCr was lower (ß = -0.21); in the riluzole arm, GM Glx (ß = -0.25) and Glx/tCr (ß = -0.29) were reduced. Baseline tNAA(ß = 0.22) and tNAA/tCr (ß = 0.23) in NAWM were associated with 9HPT scores at 96-weeks. DATA CONCLUSION: 1 H-MRS demonstrated altered membrane turnover over 96-weeks in the placebo group. It also distinguished changes in neuro-metabolites related to gliosis and glutaminergic transmission, due to fluoxetine and riluzole, respectively. Data show tNAA is a potential marker for upper limb function. LEVEL OF EVIDENCE: 1 TECHNICAL EFFICACY: Stage 4.

6.
Protein Expr Purif ; 203: 106215, 2023 03.
Article in English | MEDLINE | ID: mdl-36535546

ABSTRACT

Apyrase from potato (Solanum tuberosum) is a divalent metal ion-dependent enzyme that catalyzes the hydrolysis of nucleoside di- and tri-phosphates with broad substrate specificity. The enzyme is widely used to manipulate nucleotide levels such as in the G protein-coupled receptor (GPCR) field where it is used to deplete guanine nucleotides to stabilize nucleotide-free ternary agonist-GPCR-G protein complexes. Potato apyrase is available commercially as the native enzyme purified from potatoes or as a recombinant protein, but these are prohibitively expensive for some research applications. Here, we report a relatively simple method for the bacterial production of soluble, active potato apyrase. Apyrase has several disulfide bonds, so we co-expressed the enzyme bearing a C-terminal (His)6 tag with the E. coli disulfide isomerase DsbC at low temperature (18 °C) in the oxidizing cytoplasm of E. coli Origami B (DE3). This allowed low level production of soluble apyrase. A two-step purification procedure involving Ni-affinity followed by Cibacron Blue-affinity chromatography yielded highly purified apyrase at a level of ∼0.5 mg per L of bacterial culture. The purified enzyme was functional for ATP hydrolysis in an ATPase assay and for GTP/GDP hydrolysis in a GPCR-G protein coupling assay. This methodology enables the time- and cost-efficient production of recombinant apyrase for various research applications.


Subject(s)
Apyrase , Solanum tuberosum , Apyrase/genetics , Apyrase/chemistry , Escherichia coli/metabolism , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , Recombinant Proteins/chemistry , Solanum tuberosum/genetics , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism
7.
Proc Natl Acad Sci U S A ; 117(48): 30755-30762, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33199589

ABSTRACT

Agonist binding promotes activation of G protein-coupled receptors (GPCRs) and association of active receptors with G protein heterotrimers. The resulting active-state ternary complex is the basis for conventional stimulus-response coupling. Although GPCRs can also associate with G proteins before agonist binding, the impact of such preassociated complexes on agonist-induced signaling is poorly understood. Here we show that preassociation of 5-HT7 serotonin receptors with Gs heterotrimers is necessary for agonist-induced signaling. 5-HT7 receptors in their inactive state associate with Gs, as these complexes are stabilized by inverse agonists and receptor mutations that favor the inactive state. Inactive-state 5-HT7-Gs complexes dissociate in response to agonists, allowing the formation of conventional agonist-5-HT7-Gs ternary complexes and subsequent Gs activation. Inactive-state 5-HT7-Gs complexes are required for the full dynamic range of agonist-induced signaling, as 5-HT7 receptors spontaneously activate Gs variants that cannot form inactive-state complexes. Therefore, agonist-induced signaling in this system involves two distinct receptor-G protein complexes, a conventional ternary complex that activates G proteins and an inverse-coupled binary complex that maintains the inactive state when agonist is not present.


Subject(s)
GTP-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Dose-Response Relationship, Drug , GTP-Binding Proteins/chemistry , Kinetics , Ligands , Models, Biological , Multiprotein Complexes/chemistry , Protein Binding , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Receptors, Serotonin/chemistry , Receptors, Serotonin/metabolism , Serotonin Antagonists , Serotonin Receptor Agonists , Signal Transduction/drug effects
8.
Proc Natl Acad Sci U S A ; 117(35): 21723-21730, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817560

ABSTRACT

G proteins are activated when they associate with G protein-coupled receptors (GPCRs), often in response to agonist-mediated receptor activation. It is generally thought that agonist-induced receptor-G protein association necessarily promotes G protein activation and, conversely, that activated GPCRs do not interact with G proteins that they do not activate. Here we show that GPCRs can form agonist-dependent complexes with G proteins that they do not activate. Using cell-based bioluminescence resonance energy transfer (BRET) and luminescence assays we find that vasopressin V2 receptors (V2R) associate with both Gs and G12 heterotrimers when stimulated with the agonist arginine vasopressin (AVP). However, unlike V2R-Gs complexes, V2R-G12 complexes are not destabilized by guanine nucleotides and do not promote G12 activation. Activating V2R does not lead to signaling responses downstream of G12 activation, but instead inhibits basal G12-mediated signaling, presumably by sequestering G12 heterotrimers. Overexpressing G12 inhibits G protein receptor kinase (GRK) and arrestin recruitment to V2R and receptor internalization. Formyl peptide (FPR1 and FPR2) and Smoothened (Smo) receptors also form complexes with G12 that are insensitive to nucleotides, suggesting that unproductive GPCR-G12 complexes are not unique to V2R. These results indicate that agonist-dependent receptor-G protein association does not always lead to G protein activation and may in fact inhibit G protein activation.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Bioluminescence Resonance Energy Transfer Techniques/methods , Cyclic AMP/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/physiology , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein alpha Subunits, Gs/physiology , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Ligands , Protein Binding/physiology , Receptors, Vasopressin/metabolism , Signal Transduction/physiology , Vasopressins/metabolism , beta-Arrestins/metabolism
9.
Int J Mol Sci ; 24(11)2023 May 28.
Article in English | MEDLINE | ID: mdl-37298349

ABSTRACT

Phosphodiesterase-5 inhibitors (PDE5i) are under investigation for repurposing for colon cancer prevention. A drawback to conventional PDE5i are their side-effects and drug-drug interactions. We designed an analog of the prototypical PDE5i sildenafil by replacing the methyl group on the piperazine ring with malonic acid to reduce lipophilicity, and measured its entry into the circulation and effects on colon epithelium. This modification did not affect pharmacology as malonyl-sildenafil had a similar IC50 to sildenafil but exhibited an almost 20-fold reduced EC50 for increasing cellular cGMP. Using an LC-MS/MS approach, malonyl-sildenafil was negligible in mouse plasma after oral administration but was detected at high levels in the feces. No bioactive metabolites of malonyl-sildenafil were detected in the circulation by measuring interactions with isosorbide mononitrate. The treatment of mice with malonyl-sildenafil in the drinking water resulted in a suppression of proliferation in the colon epithelium that is consistent with results previously published for mice treated with PDE5i. A carboxylic-acid-containing analog of sildenafil prohibits the systemic delivery of the compound but maintains sufficient penetration into the colon epithelium to suppress proliferation. This highlights a novel approach to generating a first-in-class drug for colon cancer chemoprevention.


Subject(s)
Colonic Neoplasms , Phosphodiesterase 5 Inhibitors , Mice , Animals , Phosphodiesterase 5 Inhibitors/pharmacology , Sildenafil Citrate/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 5 , Chromatography, Liquid , Tandem Mass Spectrometry , Colonic Neoplasms/drug therapy , Colonic Neoplasms/prevention & control , Cell Proliferation , Cyclic GMP/metabolism
10.
Traffic ; 21(4): 324-332, 2020 04.
Article in English | MEDLINE | ID: mdl-32096320

ABSTRACT

The G protein signaling cascade is a key player in cell signaling. Cascade activation leads to a redistribution of its members in various cellular compartments. These changes are likely related to the "second wave" of signaling from endosomes. Here, we set out to determine whether Gs signaling cascade members expressed at very low levels exhibit altered mobility and localize in clathrin-coated structures (CCSs) or caveolae upon activation by ß2 -adrenergic receptors (ß2 AR). Activated ß2 AR showed decreased mobility and sustained accumulation in CCSs but not in caveolae. Arrestin 3 translocated to the plasma membrane after ß2 AR activation and showed very low mobility and pronounced accumulation in CCSs. In contrast, Gαs and Gγ2 exhibited a modest reduction in mobility but no detectable accumulation in or exclusion from CCSs or caveolae. The effector adenylyl cyclase 5 (AC5) showed a slight mobility increase upon ß2 AR stimulation, no redistribution to CCSs, and weak activation-independent accumulation in caveolae. Our findings show an overall decrease in the mobility of most activated Gs signaling cascade members and confirm that ß2 AR and arrestin 3 accumulate in CCSs, while Gαs , Gγ2 and AC5 can transiently enter CCSs and caveolae but do not accumulate in and are not excluded from these domains.


Subject(s)
Caveolae/metabolism , Cell Membrane/metabolism , Signal Transduction , GTP-Binding Proteins/metabolism , Receptors, Adrenergic, beta-2/metabolism
11.
J Biol Chem ; 296: 100325, 2021.
Article in English | MEDLINE | ID: mdl-33493514

ABSTRACT

The Golgi apparatus (GA) is a cellular organelle that plays a critical role in the processing of proteins for secretion. Activation of G protein-coupled receptors at the plasma membrane (PM) induces the translocation of G protein ßγ dimers to the GA. However, the functional significance of this translocation is largely unknown. Here, we study PM-GA translocation of all 12 Gγ subunits in response to chemokine receptor CXCR4 activation and demonstrate that Gγ9 is a unique Golgi-translocating Gγ subunit. CRISPR-Cas9-mediated knockout of Gγ9 abolishes activation of extracellular signal-regulated kinase 1 and 2 (ERK1/2), two members of the mitogen-activated protein kinase family, by CXCR4. We show that chemically induced recruitment to the GA of Gßγ dimers containing different Gγ subunits activates ERK1/2, whereas recruitment to the PM is ineffective. We also demonstrate that pharmacological inhibition of phosphoinositide 3-kinase γ (PI3Kγ) and depletion of its subunits p110γ and p101 abrogate ERK1/2 activation by CXCR4 and Gßγ recruitment to the GA. Knockout of either Gγ9 or PI3Kγ significantly suppresses prostate cancer PC3 cell migration, invasion, and metastasis. Collectively, our data demonstrate a novel function for Gßγ translocation to the GA, via activating PI3Kγ heterodimers p110γ-p101, to spatiotemporally regulate mitogen-activated protein kinase activation by G protein-coupled receptors and ultimately control tumor progression.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/genetics , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein gamma Subunits/genetics , Golgi Apparatus/genetics , Receptors, CXCR4/genetics , Cell Membrane/genetics , Dimerization , HEK293 Cells , Humans , Mitogen-Activated Protein Kinase Kinases/genetics , Phosphatidylinositol 3-Kinases/genetics , Protein Transport/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics
12.
Nat Chem Biol ; 16(12): 1440, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32807971

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

13.
Nat Chem Biol ; 16(12): 1343-1350, 2020 12.
Article in English | MEDLINE | ID: mdl-32778842

ABSTRACT

The adhesion G-protein-coupled receptor (GPCR) latrophilin 3 (ADGRL3) has been associated with increased risk of attention deficit hyperactivity disorder (ADHD) and substance use in human genetic studies. Knockdown in multiple species leads to hyperlocomotion and altered dopamine signaling. Thus, ADGRL3 is a potential target for treatment of neuropsychiatric disorders that involve dopamine dysfunction, but its basic signaling properties are poorly understood. Identification of adhesion GPCR signaling partners has been limited by a lack of tools to acutely activate these receptors in living cells. Here, we design a novel acute activation strategy to characterize ADGRL3 signaling by engineering a receptor construct in which we could trigger acute activation enzymatically. Using this assay, we found that ADGRL3 signals through G12/G13 and Gq, with G12/13 the most robustly activated. Gα12/13 is a new player in ADGRL3 biology, opening up unexplored roles for ADGRL3 in the brain. Our methodological advancements should be broadly useful in adhesion GPCR research.


Subject(s)
Activating Transcription Factor 6/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Peptides/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Activating Transcription Factor 6/agonists , Activating Transcription Factor 6/chemistry , Activating Transcription Factor 6/genetics , Animals , Arrestin/chemistry , Arrestin/genetics , Arrestin/metabolism , CRISPR-Cas Systems , Cell Engineering , GTP-Binding Protein alpha Subunits, G12-G13/chemistry , GTP-Binding Protein alpha Subunits, G12-G13/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression , HEK293 Cells , Humans , Kinetics , Mice , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/chemistry , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Peptides/chemistry , Peptides/pharmacology , Protein Binding , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/chemistry , Receptors, Peptide/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
14.
Proc Natl Acad Sci U S A ; 116(24): 12054-12059, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31142646

ABSTRACT

G protein-coupled receptors (GPCRs) activate four families of heterotrimeric G proteins, and individual receptors must select a subset of G proteins to produce appropriate cellular responses. Although the precise mechanisms of coupling selectivity are uncertain, the Gα subunit C terminus is widely believed to be the primary determinant recognized by cognate receptors. Here, we directly assess coupling between 14 representative GPCRs and 16 Gα subunits, including one wild-type Gα subunit from each of the four families and 12 chimeras with exchanged C termini. We use a sensitive bioluminescence resonance energy transfer (BRET) assay that provides control over both ligand and nucleotide binding, and allows direct comparison across G protein families. We find that the Gs- and Gq-coupled receptors we studied are relatively promiscuous and always couple to some extent to Gi1 heterotrimers. In contrast, Gi-coupled receptors are more selective. Our results with Gα subunit chimeras show that the Gα C terminus is important for coupling selectivity, but no more so than the Gα subunit core. The relative importance of the Gα subunit core and C terminus is highly variable and, for some receptors, the Gα core is more important for selective coupling than the C terminus. Our results suggest general rules for GPCR-G protein coupling and demonstrate that the critical G protein determinants of selectivity vary widely, even for different receptors that couple to the same G protein.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, G-Protein-Coupled/metabolism , Cell Line , HEK293 Cells , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Ligands , Protein Subunits/metabolism , Signal Transduction/physiology
15.
J Biol Chem ; 295(42): 14250-14259, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32683324

ABSTRACT

Motile cilia are hairlike structures that line the respiratory and reproductive tracts and the middle ear and generate fluid flow in these organs via synchronized beating. Cilium growth is a highly regulated process that is assumed to be important for flow generation. Recently, Kif19a, a kinesin residing at the cilia tip, was identified to be essential for ciliary length control through its microtubule depolymerization function. However, there is a lack of information on the nature of proteins and the integrated signaling mechanism regulating growth of motile cilia. Here, we report that adenylate cyclase 6 (AC6), a highly abundant AC isoform in airway epithelial cells, inhibits degradation of Kif19a by inhibiting autophagy, a cellular recycling mechanism for damaged proteins and organelles. Using epithelium-specific knockout mice of AC6, we demonstrated that AC6 knockout airway epithelial cells have longer cilia compared with the WT cells because of decreased Kif19a protein levels in the cilia. We demonstrated in vitro that AC6 inhibits AMP-activated kinase (AMPK), an important modulator of cellular energy-conserving mechanisms, and uncouples its binding with ciliary kinesin Kif19a. In the absence of AC6, activation of AMPK mobilizes Kif19a into autophagosomes for degradation in airway epithelial cells. Lower Kif19a levels upon pharmacological activation of AMPK in airway epithelial cells correlated with elongated cilia and vice versa. In all, the AC6-AMPK pathway, which is tunable to cellular cues, could potentially serve as one of the crucial ciliary growth checkpoints and could be channeled to develop therapeutic interventions for cilia-associated disorders.


Subject(s)
Adenylyl Cyclases/metabolism , Cilia/physiology , Kinesins/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Adenylyl Cyclases/chemistry , Adenylyl Cyclases/deficiency , Adenylyl Cyclases/genetics , Animals , Autophagosomes/metabolism , Autophagy/drug effects , Autophagy-Related Protein 5/antagonists & inhibitors , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Chloroquine/pharmacology , Cilia/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Kinesins/antagonists & inhibitors , Kinesins/genetics , Male , Mice , Mice, Knockout , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Trachea/cytology , Trachea/metabolism
16.
J Am Chem Soc ; 142(19): 8962-8971, 2020 05 13.
Article in English | MEDLINE | ID: mdl-32307996

ABSTRACT

Site-selective C-C bond formation through the direct coupling of C(sp3)-H bonds with unsaturated hydrocarbons represents an atom-economical and redox-neutral way to functionalize chemically inert positions, such as those ß to a carbonyl group. While most existing ß-functionalization methods utilize a directing group (DG) strategy, here we report a Pd-catalyzed intramolecular ß-alkenylation of ketones using alkynes as the coupling partner without the aid of DGs. Mediated by a ketone desaturation process, the reaction is redox-neutral and avoids using strong acids or bases. The resulting cis-5,6-fused bicycles can be diversely derivatized with excellent selectivity. Mechanistic studies imply an unusual "hydride-transfer" chain-like pathway, which involves the cyclometalation of an enyne intermediate and protonation of the resulting Pd enolate followed by an intermolecular hydride transfer through the desaturation of another substrate.

17.
Am J Med Genet A ; 182(5): 1104-1116, 2020 05.
Article in English | MEDLINE | ID: mdl-32133772

ABSTRACT

Craniofacial morphogenesis is regulated in part by signaling from the Endothelin receptor type A (EDNRA). Pathogenic variants in EDNRA signaling pathway components EDNRA, GNAI3, PCLB4, and EDN1 cause Mandibulofacial Dysostosis with Alopecia (MFDA), Auriculocondylar syndrome (ARCND) 1, 2, and 3, respectively. However, cardiovascular development is normal in MFDA and ARCND individuals, unlike Ednra knockout mice. One explanation may be that partial EDNRA signaling remains in MFDA and ARCND, as mice with reduced, but not absent, EDNRA signaling also lack a cardiovascular phenotype. Here we report an individual with craniofacial and cardiovascular malformations mimicking the Ednra -/- mouse phenotype, including a distinctive micrognathia with microstomia and a hypoplastic aortic arch. Exome sequencing found a novel homozygous missense variant in EDNRA (c.1142A>C; p.Q381P). Bioluminescence resonance energy transfer assays revealed that this amino acid substitution in helix 8 of EDNRA prevents recruitment of G proteins to the receptor, abrogating subsequent receptor activation by its ligand, Endothelin-1. This homozygous variant is thus the first reported loss-of-function EDNRA allele, resulting in a syndrome we have named Oro-Oto-Cardiac Syndrome. Further, our results illustrate that EDNRA signaling is required for both normal human craniofacial and cardiovascular development, and that limited EDNRA signaling is likely retained in ARCND and MFDA individuals. This work illustrates a straightforward approach to identifying the functional consequence of novel genetic variants in signaling molecules associated with malformation syndromes.


Subject(s)
Craniofacial Abnormalities/genetics , Ear Diseases/genetics , Ear/abnormalities , Genetic Predisposition to Disease , Mandibulofacial Dysostosis/genetics , Receptor, Endothelin A/genetics , Animals , Craniofacial Abnormalities/physiopathology , Ear/physiopathology , Ear Diseases/physiopathology , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Loss of Function Mutation/genetics , Mandibulofacial Dysostosis/physiopathology , Mice , Mice, Knockout , Morphogenesis/genetics , Neural Crest/growth & development , Neural Crest/pathology , Phenotype , Signal Transduction/genetics
18.
J Biol Chem ; 293(19): 7466-7473, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29523687

ABSTRACT

G protein-coupled receptors (GPCRs) are key signaling proteins that regulate nearly every aspect of cell function. Studies of GPCRs have benefited greatly from the development of molecular tools to monitor receptor activation and downstream signaling. Here, we show that mini G proteins are robust probes that can be used in a variety of assay formats to report GPCR activity in living cells. Mini G (mG) proteins are engineered GTPase domains of Gα subunits that were developed for structural studies of active-state GPCRs. Confocal imaging revealed that mG proteins fused to fluorescent proteins were located diffusely in the cytoplasm and translocated to sites of receptor activation at the cell surface and at intracellular organelles. Bioluminescence resonance energy transfer (BRET) assays with mG proteins fused to either a fluorescent protein or luciferase reported agonist, superagonist, and inverse agonist activities. Variants of mG proteins (mGs, mGsi, mGsq, and mG12) corresponding to the four families of Gα subunits displayed appropriate coupling to their cognate GPCRs, allowing quantitative profiling of subtype-specific coupling to individual receptors. BRET between luciferase-mG fusion proteins and fluorescent markers indicated the presence of active GPCRs at the plasma membrane, Golgi apparatus, and endosomes. Complementation assays with fragments of NanoLuc luciferase fused to GPCRs and mG proteins reported constitutive receptor activity and agonist-induced activation with up to 20-fold increases in luminescence. We conclude that mG proteins are versatile tools for studying GPCR activation and coupling specificity in cells and should be useful for discovering and characterizing G protein subtype-biased ligands.


Subject(s)
GTP-Binding Proteins/metabolism , Molecular Probes/metabolism , Receptors, G-Protein-Coupled/metabolism , Binding Sites , Cell Compartmentation , Energy Transfer , HEK293 Cells , Humans , Luciferases/metabolism , Microscopy, Confocal , Mutation , Protein Binding , Receptors, G-Protein-Coupled/genetics
19.
J Biol Chem ; 291(39): 20295-20302, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27528603

ABSTRACT

Heterotrimeric G proteins are localized to the plasma membrane where they transduce extracellular signals to intracellular effectors. G proteins also act at intracellular locations, and can translocate between cellular compartments. For example, Gαs can leave the plasma membrane and move to the cell interior after activation. However, the mechanism of Gαs translocation and its intracellular destination are not known. Here we use bioluminescence resonance energy transfer (BRET) to show that after activation, Gαs rapidly associates with the endoplasmic reticulum, mitochondria, and endosomes, consistent with indiscriminate sampling of intracellular membranes from the cytosol rather than transport via a specific vesicular pathway. The primary source of Gαs for endosomal compartments is constitutive endocytosis rather than activity-dependent internalization. Recycling of Gαs to the plasma membrane is complete 25 min after stimulation is discontinued. We also show that an acylation-deacylation cycle is important for the steady-state localization of Gαs at the plasma membrane, but our results do not support a role for deacylation in activity-dependent Gαs internalization.


Subject(s)
Chromogranins/metabolism , Endocytosis/physiology , Endoplasmic Reticulum/enzymology , GTP-Binding Protein alpha Subunits, Gs/metabolism , Intracellular Membranes/enzymology , Acylation , Bioluminescence Resonance Energy Transfer Techniques/methods , Chromogranins/genetics , Endoplasmic Reticulum/genetics , Enzyme Activation/physiology , GTP-Binding Protein alpha Subunits, Gs/genetics , HEK293 Cells , Humans , Protein Transport/physiology
20.
Spinal Cord ; 54(4): 248-53, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26690858

ABSTRACT

BACKGROUND: Accurate estimation of energy needs is vital for effective nutritional management of individuals with spinal cord injury (SCI). Inappropriate energy prescription after SCI can compound the rates of malnutrition or obesity, increase the risk of complications and negatively influence outcomes. Energy requirements following SCI are not well understood, and there is currently no universally accepted method of estimating energy needs in clinical practice. STUDY DESIGN: This is a systematic literature review. OBJECTIVES: The objectives of this study were to investigate and compare the measured resting energy needs of adults with SCI across different phases of rehabilitation, and to identify appropriate energy prediction equations for use in SCI. SETTING: This study was conducted in Australia. METHODS: MEDLINE, EMBASE and CENTRAL databases were searched for studies published between 1975 and April 2015, identifying 298 articles. Full articles in English language of adults with SCI who were fasted for a minimum of 8 hours before undergoing indirect calorimetry to measure resting energy expenditure (REE) for at least 20 min were selected. On the basis of the inclusion criteria, 18 articles remained for data extraction. One author extracted information from all articles, and inter-rater reliability was tested in five articles. RESULTS: REE across three phases of injury was assessed: acute, sub-acute and chronic. Few studies (n=2) have investigated REE in the acute and sub-acute injury stages of SCI recovery. The factors influencing chronic energy needs in SCI patient populations are many and varied, and a valid predictive equation for use in SCI remains elusive. CONCLUSION: Indirect calorimetry remains the only accurate assessment of REE for health practitioners working with patients after SCI.


Subject(s)
Energy Metabolism/physiology , Rest/physiology , Spinal Cord Injuries/diagnosis , Spinal Cord Injuries/physiopathology , Adult , Animals , Australia , Databases, Bibliographic/statistics & numerical data , Humans , Predictive Value of Tests , Spinal Cord Injuries/rehabilitation
SELECTION OF CITATIONS
SEARCH DETAIL