Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.265
Filter
Add more filters

Publication year range
1.
Cell ; 182(6): 1531-1544.e15, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32846158

ABSTRACT

The fidelity of intracellular signaling hinges on the organization of dynamic activity architectures. Spatial compartmentation was first proposed over 30 years ago to explain how diverse G protein-coupled receptors achieve specificity despite converging on a ubiquitous messenger, cyclic adenosine monophosphate (cAMP). However, the mechanisms responsible for spatially constraining this diffusible messenger remain elusive. Here, we reveal that the type I regulatory subunit of cAMP-dependent protein kinase (PKA), RIα, undergoes liquid-liquid phase separation (LLPS) as a function of cAMP signaling to form biomolecular condensates enriched in cAMP and PKA activity, critical for effective cAMP compartmentation. We further show that a PKA fusion oncoprotein associated with an atypical liver cancer potently blocks RIα LLPS and induces aberrant cAMP signaling. Loss of RIα LLPS in normal cells increases cell proliferation and induces cell transformation. Our work reveals LLPS as a principal organizer of signaling compartments and highlights the pathological consequences of dysregulating this activity architecture.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/genetics , Cell Compartmentation/genetics , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Cyclic AMP/metabolism , HSP40 Heat-Shock Proteins/genetics , Liver Neoplasms/genetics , Signal Transduction , Animals , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinoma, Hepatocellular/metabolism , Cell Compartmentation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoplasm/metabolism , Humans , Liver Neoplasms/metabolism , Mice , Oncogenes/genetics , Protein Domains , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins , Spectroscopy, Fourier Transform Infrared , Time-Lapse Imaging/methods
2.
Cell ; 176(6): 1461-1476.e23, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30849374

ABSTRACT

Maintaining the optimal performance of cell processes and organelles is the task of auto-regulatory systems. Here we describe an auto-regulatory device that helps to maintain homeostasis of the endoplasmic reticulum (ER) by adjusting the secretory flux to the cargo load. The cargo-recruiting subunit of the coatomer protein II (COPII) coat, Sec24, doubles as a sensor of folded cargo and, upon cargo binding, acts as a guanine nucleotide exchange factor to activate the signaling protein Gα12 at the ER exit sites (ERESs). This step, in turn, activates a complex signaling network that activates and coordinates the ER export machinery and attenuates proteins synthesis, thus preventing large fluctuations of folded and potentially active cargo that could be harmful to the cell or the organism. We call this mechanism AREX (autoregulation of ER export) and expect that its identification will aid our understanding of human physiology and diseases that develop from secretory dysfunction.


Subject(s)
Endoplasmic Reticulum/metabolism , Vesicular Transport Proteins/metabolism , Biological Transport , COP-Coated Vesicles/metabolism , COP-Coated Vesicles/physiology , Cell Line , Coatomer Protein/metabolism , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Stress/physiology , Female , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Golgi Apparatus/metabolism , Guanine Nucleotide Exchange Factors/physiology , HeLa Cells , Humans , Male , Protein Folding , Protein Transport , Proteostasis/physiology , Signal Transduction
3.
Cell ; 171(4): 836-848.e13, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28988768

ABSTRACT

Adrenergic stimulation promotes lipid mobilization and oxidation in brown and beige adipocytes, where the harnessed energy is dissipated as heat in a process known as adaptive thermogenesis. The signaling cascades and energy-dissipating pathways that facilitate thermogenesis have been extensively described, yet little is known about the counterbalancing negative regulatory mechanisms. Here, we identify a two-pore-domain potassium channel, KCNK3, as a built-in rheostat negatively regulating thermogenesis. Kcnk3 is transcriptionally wired into the thermogenic program by PRDM16, a master regulator of thermogenesis. KCNK3 antagonizes norepinephrine-induced membrane depolarization by promoting potassium efflux in brown adipocytes. This limits calcium influx through voltage-dependent calcium channels and dampens adrenergic signaling, thereby attenuating lipolysis and thermogenic respiration. Adipose-specific Kcnk3 knockout mice display increased energy expenditure and are resistant to hypothermia and obesity. These findings uncover a critical K+-Ca2+-adrenergic signaling axis that acts to dampen thermogenesis, maintain tissue homeostasis, and reveal an electrophysiological regulatory mechanism of adipocyte function.


Subject(s)
Adipose Tissue/metabolism , Nerve Tissue Proteins/metabolism , Obesity/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Receptors, Adrenergic/metabolism , Signal Transduction , Thermogenesis , Adipocytes, Brown/metabolism , Adipose Tissue/pathology , Animals , Cell Separation , Cells, Cultured , Electrophysiological Phenomena , Female , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Obesity/pathology , Potassium Channels, Tandem Pore Domain/genetics
4.
Mol Cell ; 83(7): 1093-1108.e8, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36863348

ABSTRACT

The glucagon-PKA signal is generally believed to control hepatic gluconeogenesis via the CREB transcription factor. Here we uncovered a distinct function of this signal in directly stimulating histone phosphorylation for gluconeogenic gene regulation in mice. In the fasting state, CREB recruited activated PKA to regions near gluconeogenic genes, where PKA phosphorylated histone H3 serine 28 (H3S28ph). H3S28ph, recognized by 14-3-3ζ, promoted recruitment of RNA polymerase II and transcriptional stimulation of gluconeogenic genes. In contrast, in the fed state, more PP2A was found near gluconeogenic genes, which counteracted PKA by dephosphorylating H3S28ph and repressing transcription. Importantly, ectopic expression of phosphomimic H3S28 efficiently restored gluconeogenic gene expression when liver PKA or CREB was depleted. These results together highlight a different functional scheme in regulating gluconeogenesis by the glucagon-PKA-CREB-H3S28ph cascade, in which the hormone signal is transmitted to chromatin for rapid and efficient gluconeogenic gene activation.


Subject(s)
Glucagon , Gluconeogenesis , Animals , Mice , Gluconeogenesis/genetics , Glucagon/metabolism , Histones/metabolism , Phosphorylation , 14-3-3 Proteins/metabolism , Liver/metabolism , Fasting/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism
5.
Annu Rev Genet ; 56: 253-278, 2022 11 30.
Article in English | MEDLINE | ID: mdl-36449357

ABSTRACT

Most cells live in environments that are permissive for proliferation only a small fraction of the time. Entering quiescence enables cells to survive long periods of nondivision and reenter the cell cycle when signaled to do so. Here, we describe what is known about the molecular basis for quiescence in Saccharomyces cerevisiae, with emphasis on the progress made in the last decade. Quiescence is triggered by depletion of an essential nutrient. It begins well before nutrient exhaustion, and there is extensive crosstalk between signaling pathways to ensure that all proliferation-specific activities are stopped when any one essential nutrient is limiting. Every aspect of gene expression is modified to redirect and conserve resources. Chromatin structure and composition change on a global scale, from histone modifications to three-dimensional chromatin structure. Thousands of proteins and RNAs aggregate, forming unique structures with unique fates, and the cytoplasm transitions to a glass-like state.


Subject(s)
Protein Processing, Post-Translational , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Cell Cycle/genetics , Cytoplasm , Chromatin/genetics
6.
Mol Cell ; 81(22): 4622-4634.e8, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34551282

ABSTRACT

AKT is a serine/threonine kinase that plays an important role in metabolism, cell growth, and cytoskeletal dynamics. AKT is activated by two kinases, PDK1 and mTORC2. Although the regulation of PDK1 is well understood, the mechanism that controls mTORC2 is unknown. Here, by investigating insulin receptor signaling in human cells and biochemical reconstitution, we found that insulin induces the activation of mTORC2 toward AKT by assembling a supercomplex with KRAS4B and RHOA GTPases, termed KARATE (KRAS4B-RHOA-mTORC2 Ensemble). Insulin-induced KARATE assembly is controlled via phosphorylation of GTP-bound KRAS4B at S181 and GDP-bound RHOA at S188 by protein kinase A. By developing a KARATE inhibitor, we demonstrate that KRAS4B-RHOA interaction drives KARATE formation. In adipocytes, KARATE controls insulin-dependent translocation of the glucose transporter GLUT4 to the plasma membrane for glucose uptake. Thus, our work reveals a fundamental mechanism that activates mTORC2 toward AKT in insulin-regulated glucose homeostasis.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , Glucose/metabolism , Insulin/metabolism , Mechanistic Target of Rapamycin Complex 2/chemistry , rhoA GTP-Binding Protein/chemistry , 3T3-L1 Cells , Adipocytes/cytology , Animals , Cell Membrane/metabolism , Cytoskeleton/metabolism , Dictyostelium , Glucose Transporter Type 4/metabolism , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , HEK293 Cells , Humans , Mice , Phosphorylation , Protein Transport , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/metabolism
7.
Mol Cell ; 81(4): 675-690.e8, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33453167

ABSTRACT

Neural network computations are usually assumed to emerge from patterns of fast electrical activity. Challenging this view, we show that a male fly's decision to persist in mating hinges on a biochemical computation that enables processing over minutes to hours. Each neuron in a recurrent network contains slightly different internal molecular estimates of mating progress. Protein kinase A (PKA) activity contrasts this internal measurement with input from the other neurons to represent accumulated evidence that the goal of the network has been achieved. When consensus is reached, PKA pushes the network toward a large-scale and synchronized burst of calcium influx that we call an eruption. Eruptions transform continuous deliberation within the network into an all-or-nothing output, after which the male will no longer sacrifice his life to continue mating. Here, biochemical activity, invisible to most large-scale recording techniques, is the key computational currency directing behavior and motivational state.


Subject(s)
Calcium Signaling , Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Drosophila Proteins/metabolism , Nerve Net/metabolism , Neurons/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , Drosophila Proteins/genetics , Drosophila melanogaster
8.
Genes Dev ; 34(7-8): 495-510, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32139423

ABSTRACT

Obesity-induced diabetes affects >400 million people worldwide. Uncontrolled lipolysis (free fatty acid release from adipocytes) can contribute to diabetes and obesity. To identify future therapeutic avenues targeting this pathway, we performed a high-throughput screen and identified the extracellular-regulated kinase 3 (ERK3) as a hit. We demonstrated that ß-adrenergic stimulation stabilizes ERK3, leading to the formation of a complex with the cofactor MAP kinase-activated protein kinase 5 (MK5), thereby driving lipolysis. Mechanistically, we identified a downstream target of the ERK3/MK5 pathway, the transcription factor FOXO1, which promotes the expression of the major lipolytic enzyme ATGL. Finally, we provide evidence that targeted deletion of ERK3 in mouse adipocytes inhibits lipolysis, but elevates energy dissipation, promoting lean phenotype and ameliorating diabetes. Thus, ERK3/MK5 represents a previously unrecognized signaling axis in adipose tissue and an attractive target for future therapies aiming to combat obesity-induced diabetes.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Energy Metabolism/genetics , Lipolysis/genetics , Mitogen-Activated Protein Kinase 6/genetics , Mitogen-Activated Protein Kinase 6/metabolism , Obesity/complications , 3T3 Cells , Adipose Tissue/enzymology , Animals , Diabetes Mellitus, Type 2/drug therapy , Drug Evaluation, Preclinical , Forkhead Box Protein O1/metabolism , Gene Deletion , HEK293 Cells , Humans , Hypoglycemic Agents/therapeutic use , Intracellular Signaling Peptides and Proteins/metabolism , Lipase/genetics , Lipase/metabolism , Mice , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics
9.
Proc Natl Acad Sci U S A ; 121(26): e2405553121, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38889144

ABSTRACT

The cytoplasm is a complex, crowded environment that influences myriad cellular processes including protein folding and metabolic reactions. Recent studies have suggested that changes in the biophysical properties of the cytoplasm play a key role in cellular homeostasis and adaptation. However, it still remains unclear how cells control their cytoplasmic properties in response to environmental cues. Here, we used fission yeast spores as a model system of dormant cells to elucidate the mechanisms underlying regulation of the cytoplasmic properties. By tracking fluorescent tracer particles, we found that particle mobility decreased in spores compared to vegetative cells and rapidly increased at the onset of dormancy breaking upon glucose addition. This cytoplasmic fluidization depended on glucose-sensing via the cyclic adenosine monophosphate-protein kinase A pathway. PKA activation led to trehalose degradation through trehalase Ntp1, thereby increasing particle mobility as the amount of trehalose decreased. In contrast, the rapid cytoplasmic fluidization did not require de novo protein synthesis, cytoskeletal dynamics, or cell volume increase. Furthermore, the measurement of diffusion coefficients with tracer particles of different sizes suggests that the spore cytoplasm impedes the movement of larger protein complexes (40 to 150 nm) such as ribosomes, while allowing free diffusion of smaller molecules (~3 nm) such as second messengers and signaling proteins. Our experiments have thus uncovered a series of signaling events that enable cells to quickly fluidize the cytoplasm at the onset of dormancy breaking.


Subject(s)
Cytoplasm , Schizosaccharomyces , Spores, Fungal , Trehalose , Spores, Fungal/metabolism , Spores, Fungal/physiology , Schizosaccharomyces/metabolism , Schizosaccharomyces/physiology , Cytoplasm/metabolism , Trehalose/metabolism , Glucose/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/genetics , Signal Transduction
10.
Proc Natl Acad Sci U S A ; 121(24): e2400732121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38838021

ABSTRACT

Cytoplasmic mislocalization and aggregation of TDP-43 protein are hallmarks of amyotrophic lateral sclerosis (ALS) and are observed in the vast majority of both familial and sporadic cases. How these two interconnected processes are regulated on a molecular level, however, remains enigmatic. Genome-wide screens for modifiers of the ALS-associated genes TDP-43 and FUS have identified the phospholipase D (Pld) pathway as a key regulator of ALS-related phenotypes in the fruit fly Drosophila melanogaster [M. W. Kankel et al., Genetics 215, 747-766 (2020)]. Here, we report the results of our search for downstream targets of the enzymatic product of Pld, phosphatidic acid. We identify two conserved negative regulators of the cAMP/PKA signaling pathway, the phosphodiesterase dunce and the inhibitory subunit PKA-R2, as modifiers of pathogenic phenotypes resulting from overexpression of the Drosophila TDP-43 ortholog TBPH. We show that knockdown of either of these genes results in a mitigation of both TBPH aggregation and mislocalization in larval motor neuron cell bodies, as well as an amelioration of adult-onset motor defects and shortened lifespan induced by TBPH. We determine that PKA kinase activity is downstream of both TBPH and Pld and that overexpression of the PKA target CrebA can rescue TBPH mislocalization. These findings suggest a model whereby increasing cAMP/PKA signaling can ameliorate the molecular and functional effects of pathological TDP-43.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Cyclic AMP , DNA-Binding Proteins , Drosophila Proteins , Drosophila melanogaster , Signal Transduction , Animals , Cyclic AMP/metabolism , Drosophila melanogaster/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Humans , Motor Neurons/metabolism
11.
EMBO J ; 41(22): e110963, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36217825

ABSTRACT

Autophagy provides nutrients during starvation and eliminates detrimental cellular components. However, accumulating evidence indicates that autophagy is not merely a housekeeping process. Here, by combining mouse models of neuron-specific ATG5 deficiency in either excitatory or inhibitory neurons with quantitative proteomics, high-content microscopy, and live-imaging approaches, we show that autophagy protein ATG5 functions in neurons to regulate cAMP-dependent protein kinase A (PKA)-mediated phosphorylation of a synapse-confined proteome. This function of ATG5 is independent of bulk turnover of synaptic proteins and requires the targeting of PKA inhibitory R1 subunits to autophagosomes. Neuronal loss of ATG5 causes synaptic accumulation of PKA-R1, which sequesters the PKA catalytic subunit and diminishes cAMP/PKA-dependent phosphorylation of postsynaptic cytoskeletal proteins that mediate AMPAR trafficking. Furthermore, ATG5 deletion in glutamatergic neurons augments AMPAR-dependent excitatory neurotransmission and causes the appearance of spontaneous recurrent seizures in mice. Our findings identify a novel role of autophagy in regulating PKA signaling at glutamatergic synapses and suggest the PKA as a target for restoration of synaptic function in neurodegenerative conditions with autophagy dysfunction.


Subject(s)
Neurons , Synapses , Mice , Animals , Synapses/metabolism , Neurons/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Signal Transduction , Autophagy
12.
EMBO J ; 41(1): e105026, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34791698

ABSTRACT

Intronic GGGGCC (G4C2) hexanucleotide repeat expansion within the human C9orf72 gene represents the most common cause of familial forms of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) (C9ALS/FTD). Repeat-associated non-AUG (RAN) translation of repeat-containing C9orf72 RNA results in the production of neurotoxic dipeptide-repeat proteins (DPRs). Here, we developed a high-throughput drug screen for the identification of positive and negative modulators of DPR levels. We found that HSP90 inhibitor geldanamycin and aldosterone antagonist spironolactone reduced DPR levels by promoting protein degradation via the proteasome and autophagy pathways respectively. Surprisingly, cAMP-elevating compounds boosting protein kinase A (PKA) activity increased DPR levels. Inhibition of PKA activity, by both pharmacological and genetic approaches, reduced DPR levels in cells and rescued pathological phenotypes in a Drosophila model of C9ALS/FTD. Moreover, knockdown of PKA-catalytic subunits correlated with reduced translation efficiency of DPRs, while the PKA inhibitor H89 reduced endogenous DPR levels in C9ALS/FTD patient-derived iPSC motor neurons. Together, our results suggest new and druggable pathways modulating DPR levels in C9ALS/FTD.


Subject(s)
C9orf72 Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dipeptides/metabolism , Proteolysis , Small Molecule Libraries/pharmacology , Animals , Cell Line , Codon, Initiator/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Repeat Expansion/genetics , Disease Models, Animal , Drosophila/drug effects , Frontotemporal Dementia/pathology , HEK293 Cells , High-Throughput Screening Assays , Humans , Induced Pluripotent Stem Cells/pathology , Isoquinolines/pharmacology , Longevity/drug effects , Motor Neurons/drug effects , Motor Neurons/pathology , Protein Biosynthesis/drug effects , Proteolysis/drug effects , RNA Interference , Sulfonamides/pharmacology
13.
Brief Bioinform ; 25(5)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39171986

ABSTRACT

During the drug discovery and design process, the acid-base dissociation constant (pKa) of a molecule is critically emphasized due to its crucial role in influencing the ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties and biological activity. However, the experimental determination of pKa values is often laborious and complex. Moreover, existing prediction methods exhibit limitations in both the quantity and quality of the training data, as well as in their capacity to handle the complex structural and physicochemical properties of compounds, consequently impeding accuracy and generalization. Therefore, developing a method that can quickly and accurately predict molecular pKa values will to some extent help the structural modification of molecules, and thus assist the development process of new drugs. In this study, we developed a cutting-edge pKa prediction model named GR-pKa (Graph Retention pKa), leveraging a message-passing neural network and employing a multi-fidelity learning strategy to accurately predict molecular pKa values. The GR-pKa model incorporates five quantum mechanical properties related to molecular thermodynamics and dynamics as key features to characterize molecules. Notably, we originally introduced the novel retention mechanism into the message-passing phase, which significantly improves the model's ability to capture and update molecular information. Our GR-pKa model outperforms several state-of-the-art models in predicting macro-pKa values, achieving impressive results with a low mean absolute error of 0.490 and root mean square error of 0.588, and a high R2 of 0.937 on the SAMPL7 dataset.


Subject(s)
Neural Networks, Computer , Thermodynamics , Drug Discovery/methods
14.
Proc Natl Acad Sci U S A ; 120(26): e2220343120, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37339196

ABSTRACT

In bacterial voltage-gated sodium channels, the passage of ions through the pore is controlled by a selectivity filter (SF) composed of four glutamate residues. The mechanism of selectivity has been the subject of intense research, with suggested mechanisms based on steric effects, and ion-triggered conformational change. Here, we propose an alternative mechanism based on ion-triggered shifts in pKa values of SF glutamates. We study the NavMs channel for which the open channel structure is available. Our free-energy calculations based on molecular dynamics simulations suggest that pKa values of the four glutamates are higher in solution of K+ ions than in solution of Na+ ions. Higher pKa in the presence of K+ stems primarily from the higher population of dunked conformations of the protonated Glu sidechain, which exhibit a higher pKa shift. Since pKa values are close to the physiological pH, this results in predominant population of the fully deprotonated state of glutamates in Na+ solution, while protonated states are predominantly populated in K+ solution. Through molecular dynamics simulations we calculate that the deprotonated state is the most conductive, the singly protonated state is less conductive, and the doubly protonated state has significantly reduced conductance. Thus, we propose that a significant component of selectivity is achieved through ion-triggered shifts in the protonation state, which favors more conductive states for Na+ ions and less conductive states for K+ ions. This mechanism also suggests a strong pH dependence of selectivity, which has been experimentally observed in structurally similar NaChBac channels.


Subject(s)
Bacteria , Voltage-Gated Sodium Channels , Ions , Bacteria/metabolism , Molecular Dynamics Simulation , Glutamates , Potassium/metabolism
15.
Proc Natl Acad Sci U S A ; 120(4): e2208749120, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36656863

ABSTRACT

cAMP, a key player in many physiological processes, was classically considered to originate solely from the plasma membrane (PM). This view was recently challenged by observations showing that upon internalization GsPCRs can sustain signaling from endosomes and/or the trans-Golgi network (TGN). In this new view, after the first PM-generated cAMP wave, the internalization of GsPCRs and ACs generates a second wave that was strictly associated with nuclear transcriptional events responsible for triggering specific biological responses. Here, we report that the endogenously expressed TSHR, a canonical GsPCR, triggers an internalization-dependent, calcium-mediated nuclear sAC activation that drives PKA activation and CREB phosphorylation. Both pharmacological and genetic sAC inhibition, which did not affect the cytosolic cAMP levels, blunted nuclear cAMP accumulation, PKA activation, and cell proliferation, while an increase in nuclear sAC expression significantly enhanced cell proliferation. Furthermore, using novel nuclear-targeted optogenetic actuators, we show that light-stimulated nuclear cAMP synthesis can mimic the proliferative action of TSH by activating PKA and CREB. Therefore, based on our results, we propose a novel three-wave model in which the "third" wave of cAMP is generated by nuclear sAC. Despite being downstream of events occurring at the PM (first wave) and endosomes/TGN (second wave), the nuclear sAC-generated cAMP (third wave) is sufficient and rate-limiting for thyroid cell proliferation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Cyclic AMP , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Signal Transduction , Cell Nucleus/metabolism , Cell Proliferation , Phosphorylation
16.
J Neurosci ; 44(12)2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38267256

ABSTRACT

Imaging brain learning and memory circuit kinase signaling is a monumental challenge. The separation of phases-based activity reporter of kinase (SPARK) biosensors allow circuit-localized studies of multiple interactive kinases in vivo, including protein kinase A (PKA) and extracellular signal-regulated kinase (ERK) signaling. In the precisely-mapped Drosophila brain learning/memory circuit, we find PKA and ERK signaling differentially enriched in distinct Kenyon cell connectivity nodes. We discover that potentiating normal circuit activity induces circuit-localized PKA and ERK signaling, expanding kinase function within new presynaptic and postsynaptic domains. Activity-induced PKA signaling shows extensive overlap with previously selective ERK signaling nodes, while activity-induced ERK signaling arises in new connectivity nodes. We find targeted synaptic transmission blockade in Kenyon cells elevates circuit-localized ERK induction in Kenyon cells with normally high baseline ERK signaling, suggesting lateral and feedback inhibition. We discover overexpression of the pathway-linking Meng-Po (human SBK1) serine/threonine kinase to improve learning acquisition and memory consolidation results in dramatically heightened PKA and ERK signaling in separable Kenyon cell circuit connectivity nodes, revealing both synchronized and untapped signaling potential. Finally, we find that a mechanically-induced epileptic seizure model (easily shocked "bang-sensitive" mutants) has strongly elevated, circuit-localized PKA and ERK signaling. Both sexes were used in all experiments, except for the hemizygous male-only seizure model. Hyperexcitable, learning-enhanced, and epileptic seizure models have comparably elevated interactive kinase signaling, suggesting a common basis of use-dependent induction. We conclude that PKA and ERK signaling modulation is locally coordinated in use-dependent spatial circuit dynamics underlying seizure susceptibility linked to learning/memory potential.


Subject(s)
Learning , Signal Transduction , Animals , Female , Male , Humans , Learning/physiology , Signal Transduction/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Brain/metabolism , Drosophila/metabolism , Seizures
17.
J Biol Chem ; : 107656, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39128715

ABSTRACT

Adrenergic modulation of voltage gated Ca2+ currents is a context specific process. In the heart Cav1.2 channels initiate excitation-contraction coupling. This requires protein kinase A (PKA) phosphorylation of the small GTPase Rad (Ras associated with diabetes) and involves direct phosphorylation of a1 subunit of the Cav1.2 at Ser1700. A contributing factor is the proximity of PKA to the channel through association with A-kinase anchoring proteins (AKAPs). Disruption of PKA anchoring by the disruptor peptide AKAP-IS prevents up-regulation of Cav1.2 currents in tsA-201 cells. Biochemical analyses demonstrate that Rad does not function as an A-kinase anchoring protein. Electrophysiological recording shows that channel mutants lacking phosphorylation sites (Cav1.2 STAA) lose responsivity to the second messenger cAMP. Measurements in cardiomyocytes isolated from Rad-/- mice show that adrenergic activation of Cav1.2 is attenuated but not completely abolished. Whole animal electrocardiography studies reveal that cardiac selective Rad knockout mice exhibited higher baseline left-ventricular ejection fraction (EF), greater fractional shortening (FS), and increased heart rate as compared to control animals. Yet, each parameter of cardiac function was slightly elevated when Rad-/- mice were treated with the adrenergic agonist isoproterenol. Thus, phosphorylation of Cav1.2 and dissociation of phospho-Rad from the channel are local cAMP responsive events that act in concert to enhance L-type calcium currents. This convergence of local PKA regulatory events at the cardiac L-type calcium channel may permit maximal ß-adrenergic influence on the fight-or-flight response.

18.
J Biol Chem ; 300(5): 107149, 2024 May.
Article in English | MEDLINE | ID: mdl-38479599

ABSTRACT

Persulfides (RSSH/RSS-) participate in sulfur metabolism and are proposed to transduce hydrogen sulfide (H2S) signaling. Their biochemical properties are poorly understood. Herein, we studied the acidity and nucleophilicity of several low molecular weight persulfides using the alkylating agent, monobromobimane. The different persulfides presented similar pKa values (4.6-6.3) and pH-independent rate constants (3.2-9.0 × 103 M-1 s-1), indicating that the substituents in persulfides affect properties to a lesser extent than in thiols because of the larger distance to the outer sulfur. The persulfides had higher reactivity with monobromobimane than analogous thiols and putative thiols with the same pKa, providing evidence for the alpha effect (enhanced nucleophilicity by the presence of a contiguous atom with high electron density). Additionally, we investigated two enzymes from the human mitochondrial H2S oxidation pathway that form catalytic persulfide intermediates, sulfide quinone oxidoreductase and thiosulfate sulfurtransferase (TST, rhodanese). The pH dependence of the activities of both enzymes was measured using sulfite and/or cyanide as sulfur acceptors. The TST half-reactions were also studied by stopped-flow fluorescence spectroscopy. Both persulfidated enzymes relied on protonated groups for reaction with the acceptors. Persulfidated sulfide quinone oxidoreductase appeared to have a pKa of 7.8 ± 0.2. Persulfidated TST presented a pKa of 9.38 ± 0.04, probably due to a critical active site residue rather than the persulfide itself. The TST thiol reacted in the anionic state with thiosulfate, with an apparent pKa of 6.5 ± 0.1. Overall, our study contributes to a fundamental understanding of persulfide properties and their modulation by protein environments.


Subject(s)
Sulfides , Thiosulfate Sulfurtransferase , Humans , Bridged Bicyclo Compounds , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/chemistry , Hydrogen-Ion Concentration , Oxidation-Reduction , Quinone Reductases/metabolism , Quinone Reductases/chemistry , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism , Sulfides/chemistry , Sulfides/metabolism , Thiosulfate Sulfurtransferase/metabolism , Thiosulfate Sulfurtransferase/chemistry , Quinones/chemistry , Quinones/metabolism , Substrate Specificity
19.
J Biol Chem ; 300(7): 107444, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38838772

ABSTRACT

Candida albicans is an opportunistic fungal pathogen that can switch between yeast and hyphal morphologies depending on the environmental cues it receives. The switch to hyphal form is crucial for the establishment of invasive infections. The hyphal form is also characterized by the cell surface expression of hyphae-specific proteins, many of which are GPI-anchored and important determinants of its virulence. The coordination between hyphal morphogenesis and the expression of GPI-anchored proteins is made possible by an interesting cross-talk between GPI biosynthesis and the cAMP-PKA signaling cascade in the fungus; a parallel interaction is not found in its human host. On the other hand, in the nonpathogenic yeast, Saccharomyces cerevisiae, GPI biosynthesis is shut down when filamentation is activated and vice versa. This too is achieved by a cross-talk between GPI biosynthesis and cAMP-PKA signaling. How are diametrically opposite effects obtained from the cross-talk between two reasonably well-conserved pathways present ubiquitously across eukarya? This Review attempts to provide a model to explain these differences. In order to do so, it first provides an overview of the two pathways for the interested reader, highlighting the similarities and differences that are observed in C. albicans versus the well-studied S. cerevisiae model, before going on to explain how the different mechanisms of regulation are effected. While commonalities enable the development of generalized theories, it is hoped that a more nuanced approach, that takes into consideration species-specific differences, will enable organism-specific understanding of these processes and contribute to the development of targeted therapies.


Subject(s)
Candida albicans , Cyclic AMP-Dependent Protein Kinases , Cyclic AMP , Hyphae , Saccharomyces cerevisiae , Signal Transduction , Candida albicans/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Hyphae/metabolism , Fungal Proteins/metabolism , Fungal Proteins/genetics , Glycosylphosphatidylinositols/metabolism , Glycosylphosphatidylinositols/biosynthesis , Humans , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics
20.
J Biol Chem ; 300(5): 107234, 2024 May.
Article in English | MEDLINE | ID: mdl-38552737

ABSTRACT

Focal adhesions (FAs) form the junction between extracellular matrix (ECM)-bound integrins and the actin cytoskeleton and also transmit signals that regulate cell adhesion, cytoskeletal dynamics, and cell migration. While many of these signals are rooted in reversible tyrosine phosphorylation, phosphorylation of FA proteins on Ser/Thr residues is far more abundant yet its mechanisms and consequences are far less understood. The cAMP-dependent protein kinase (protein kinase A; PKA) has important roles in cell adhesion and cell migration and is both an effector and regulator of integrin-mediated adhesion to the ECM. Importantly, subcellular localization plays a critically important role in specifying PKA function. Here, we show that PKA is present in isolated FA-cytoskeleton complexes and active within FAs in live cells. Furthermore, using kinase-catalyzed biotinylation of isolated FA-cytoskeleton complexes, we identify 53 high-stringency candidate PKA substrates within FAs. From this list, we validate tensin-3 (Tns3)-a well-established molecular scaffold, regulator of cell migration, and a component of focal and fibrillar adhesions-as a novel direct substrate for PKA. These observations identify a new pathway for phospho-regulation of Tns3 and, importantly, establish a new and important niche for localized PKA signaling and thus provide a foundation for further investigation of the role of PKA in the regulation of FA dynamics and signaling.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Focal Adhesions , Tensins , Animals , Humans , Cell Adhesion , Cell Movement , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeleton/metabolism , Focal Adhesions/enzymology , Phosphorylation , Tensins/metabolism , Mice , Rats , Cell Line , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL