Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
J Phys Chem B ; 128(15): 3554-3562, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38580321

ABSTRACT

Understanding how signaling proteins like G proteins are allosterically activated is a long-standing challenge with significant biological and medical implications. Because it is difficult to directly observe such dynamic processes, much of our understanding is based on inferences from a limited number of static snapshots of relevant protein structures, mutagenesis data, and patterns of sequence conservation. Here, we use computer simulations to directly interrogate allosteric coupling in six G protein α-subunit isoforms covering all four G protein families. To analyze this data, we introduce automated methods for inferring allosteric networks from simulation data and assessing how allostery is conserved or diverged among related protein isoforms. We find that the allosteric networks in these six G protein α subunits are largely conserved and consist of two pathways, which we call pathway-I and pathway-II. This analysis predicts that pathway-I is generally dominant over pathway-II, which we experimentally corroborate by showing that mutations to pathway-I perturb nucleotide exchange more than mutations to pathway-II. In the future, insights into unique elements of each G protein family could inform the design of isoform-specific drugs. More broadly, our tools should also be useful for studying allostery in other proteins and assessing the extent to which this allostery is conserved in related proteins.


Subject(s)
GTP-Binding Protein alpha Subunits , Proteins , Allosteric Regulation , Proteins/chemistry , Computer Simulation , GTP-Binding Protein alpha Subunits/genetics
2.
Mol Cell Proteomics ; 22(11): 100649, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37730182

ABSTRACT

Metastatic uveal melanoma (UM) patients typically survive only 2 to 3 years because effective therapy does not yet exist. Here, to facilitate the discovery of therapeutic targets in UM, we have identified protein kinase signaling mechanisms elicited by the drivers in 90% of UM tumors: mutant constitutively active G protein α-subunits encoded by GNAQ (Gq) or GNA11 (G11). We used the highly specific Gq/11 inhibitor FR900359 (FR) to elucidate signaling networks that drive proliferation, metabolic reprogramming, and dedifferentiation of UM cells. We determined the effects of FR on the proteome and phosphoproteome of UM cells as indicated by bioinformatic analyses with CausalPath and site-specific gene set enrichment analysis. We found that inhibition of oncogenic Gq/11 caused deactivation of PKC, Erk, and the cyclin-dependent kinases CDK1 and CDK2 that drive proliferation. Inhibition of oncogenic Gq/11 in UM cells with low metastatic risk relieved inhibitory phosphorylation of polycomb-repressive complex subunits that regulate melanocytic redifferentiation. Site-specific gene set enrichment analysis, unsupervised analysis, and functional studies indicated that mTORC1 and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 2 drive metabolic reprogramming in UM cells. Together, these results identified protein kinase signaling networks driven by oncogenic Gq/11 that regulate critical aspects of UM cell biology and provide targets for therapeutic investigation.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11 , Uveal Neoplasms , Humans , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/pharmacology , Cell Proliferation , Uveal Neoplasms/genetics , Uveal Neoplasms/metabolism , Uveal Neoplasms/pathology , Protein Kinase C/metabolism , Computational Biology , Mutation
3.
Synthesis (Stuttg) ; 55(1): 90-106, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36644007

ABSTRACT

The biological activity of natural products YM-254890 (YM) and FR900359 (FR) has led to significant interest in both their synthesis and the construction of more simplified analogs. While the simplified analogs lose much of the potency of the natural products, they are of interest in their own right, and their synthesis has revealed synthetic barriers to the family of molecules that need to be addressed if a scalable synthesis of YM and FR analogs is to be constructed. In the work described here, a synthetic route to simplified analogs of YM is examined and strategies for circumventing some of the challenges inherent to constructing the molecules are forwarded.

4.
European J Org Chem ; 26(20)2023 May 22.
Article in English | MEDLINE | ID: mdl-38188369

ABSTRACT

YM-254890 and FR900359 are potent and selective inhibitors of the Gq/11-signaling pathway. As such, they have been attractive targets for both synthesis and biological studies. Yet in spite of this effort, a versatile synthetic approach to the molecules that allows for the rapid construction of a variety of non-natural and labelled analogs and an increase in the amount of those analogs available remains elusive. We report here a convergent building block approach to the molecules that can solve this challenge.

5.
J Biol Chem ; 298(1): 101495, 2022 01.
Article in English | MEDLINE | ID: mdl-34919964

ABSTRACT

Metabolic reprogramming has been shown to occur in uveal melanoma (UM), the most common intraocular tumor in adults. Mechanisms driving metabolic reprogramming in UM are poorly understood. Elucidation of these mechanisms could inform development of new therapeutic strategies for metastatic UM, which has poor prognosis because existing therapies are ineffective. Here, we determined whether metabolic reprogramming is driven by constitutively active mutant α-subunits of the heterotrimeric G proteins Gq or G11 (Gq/11), the oncogenic drivers in ∼90% of UM patients. Using PET-computed tomography imaging, microphysiometry, and GC/MS, we found that inhibition of oncogenic Gq/11 with the small molecule FR900359 (FR) attenuated glucose uptake by UM cells in vivo and in vitro, blunted glycolysis and mitochondrial respiration in UM cell lines and tumor cells isolated from patients, and reduced levels of several glycolytic and tricarboxylic acid cycle intermediates. FR acutely inhibited glycolysis and respiration and chronically attenuated expression of genes in both metabolic processes. UM therefore differs from other melanomas that exhibit a classic Warburg effect. Metabolic reprogramming in UM cell lines and patient samples involved protein kinase C and extracellular signal-regulated protein kinase 1/2 signaling downstream of oncogenic Gq/11. Chronic administration of FR upregulated expression of genes involved in metabolite scavenging and redox homeostasis, potentially as an adaptive mechanism explaining why FR does not efficiently kill UM tumor cells or regress UM tumor xenografts. These results establish that oncogenic Gq/11 signaling is a crucial driver of metabolic reprogramming in UM and lay a foundation for studies aimed at targeting metabolic reprogramming for therapeutic development.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11 , GTP-Binding Protein alpha Subunits , Melanoma , Uveal Neoplasms , Carcinogenesis , Cell Line, Tumor , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Melanoma/metabolism , Melanoma/pathology , Uveal Neoplasms/metabolism
6.
J Biol Chem ; 296: 100403, 2021.
Article in English | MEDLINE | ID: mdl-33577798

ABSTRACT

Uveal melanoma (UM) is the most common intraocular tumor in adults. Nearly half of UM patients develop metastatic disease and often succumb within months because effective therapy is lacking. A novel therapeutic approach has been suggested by the discovery that UM cell lines driven by mutant constitutively active Gq or G11 can be targeted by FR900359 (FR) or YM-254890, which are bioavailable, selective inhibitors of the Gq/11/14 subfamily of heterotrimeric G proteins. Here, we have addressed the therapeutic potential of FR for UM. We found that FR inhibited all oncogenic Gq/11 mutants reported in UM. FR arrested growth of all Gq/11-driven UM cell lines tested, but induced apoptosis only in a few. Similarly, FR inhibited growth of, but did not efficiently kill, UM tumor cells from biopsies of primary or metastatic tumors. FR evoked melanocytic redifferentiation of UM tumor cells with low (class 1), but not high (class 2), metastatic potential. FR administered systemically below its LD50 strongly inhibited growth of PDX-derived class 1 and class 2 UM tumors in mouse xenograft models and reduced blood pressure transiently. FR did not regress xenografted UM tumors or significantly affect heart rate, liver function, hematopoiesis, or behavior. These results indicated the existence of a therapeutic window in which FR can be explored for treating UM and potentially other diseases caused by constitutively active Gq/11.


Subject(s)
Depsipeptides/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , Liver Neoplasms/drug therapy , Melanoma/drug therapy , Peptides, Cyclic/pharmacology , Uveal Neoplasms/drug therapy , Animals , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Male , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Neoplasm Metastasis , Uveal Neoplasms/genetics , Uveal Neoplasms/metabolism , Uveal Neoplasms/pathology , Xenograft Model Antitumor Assays
7.
Mol Biol Cell ; 32(5): 413-421, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33405963

ABSTRACT

Uveal melanomas (UMs) are malignant cancers arising from the pigmented layers of the eye. UM cells spread through the bloodstream, and circulating UM cells are detectable in patients before metastases appear. Extravasation of UM cells is necessary for formation of metastases, and transendothelial migration (TEM) is a key step in extravasation. UM cells execute TEM via a stepwise process involving the actin-based processes of ameboid blebbing and mesenchymal lamellipodial protrusion. UM cancers are driven by oncogenic mutations that activate Gαq/11, and this activates TRIO, a guanine nucleotide exchange factor for RhoA and Rac1. We found that pharmacologic inhibition of Gαq/11 in UM cells reduced TEM. Inhibition of the RhoA pathway blocked amoeboid motility but led to enhanced TEM; in contrast, inhibition of the Rac1 pathway decreased mesenchymal motility and reduced TEM. Inhibition of Arp2/3 complex allowed cells to transmigrate without intercalation, a direct mechanism similar to the one often displayed by immune cells. BAP1-deficient (+/-) UM subclones displayed motility behavior and increased levels of TEM, similar to the effects of RhoA inhibitors. We conclude that RhoA and Rac1 signaling pathways, downstream of oncogenic Gαq/11, combine with pathways regulated by BAP1 to control the motility and transmigration of UM cells.


Subject(s)
Cell Movement/physiology , Melanoma/metabolism , Transendothelial and Transepithelial Migration/physiology , Uveal Neoplasms/metabolism , Blister/metabolism , Cell Line, Tumor , Cytoplasmic Streaming/physiology , Endothelium/metabolism , Endothelium/pathology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Melanoma/pathology , Pseudopodia/metabolism , Signal Transduction/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Uveal Neoplasms/pathology , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
8.
Clin Cancer Res ; 26(23): 6374-6386, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32933997

ABSTRACT

PURPOSE: Mutational activation of GNAQ or GNA11 (GNAQ/11), detected in >90% of uveal melanomas, leads to constitutive activation of oncogenic pathways, including MAPK and YAP. To date, chemo- or pathway-targeted therapies, either alone or in combination, have proven ineffective in the treatment of patients with metastatic uveal melanoma. EXPERIMENTAL DESIGN: We tested the efficacy of chloroquine or hydroxychloroquine, in combination with MAPK pathway inhibition in GNAQ/11-mutated cells in vitro and in vivo and identified mechanisms of MEK1/2 inhibitor plus chloroquine-induced cytotoxicity. RESULTS: Inhibition of GNAQ/11-mediated activation of MAPK signaling resulted in the induction of autophagy. Combined inhibition of Gα and autophagy or lysosome function resulted in enhanced cell death. Moreover, the combination of MEK1/2 inhibition, using trametinib, with the lysosome inhibitor, chloroquine, also increased cytotoxicity. Treatment of mice bearing GNAQ/11-driven melanomas with trametinib plus hydroxychloroquine resulted in inhibition of tumor growth and significantly prolonged survival. Interestingly, lysosomal- and autophagy-specific inhibition with bafilomycin A1 was not sufficient to promote cytotoxicity in combination with trametinib. However, the addition of YAP inhibition with trametinib plus bafilomycin A1 resulted in cell death at comparable levels to trametinib plus chloroquine (T/CQ) treatment. Furthermore, T/CQ-treated cells displayed decreased YAP nuclear localization and decreased YAP transcriptional activity. Expression of a constitutively active YAP5SA mutant conferred resistance to T/CQ-induced cell death. CONCLUSIONS: These results suggest that YAP, MEK1/2, and lysosome function are necessary and critical targets for the therapy of GNAQ/11-driven melanoma, and identify trametinib plus hydroxychloroquine as a potential treatment strategy for metastatic uveal melanoma.


Subject(s)
Chloroquine/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits/genetics , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Melanoma/drug therapy , Mutation , Pyridones/pharmacology , Pyrimidinones/pharmacology , Uveal Neoplasms/drug therapy , Animals , Antimalarials/pharmacology , Apoptosis , Cell Proliferation , Drug Resistance, Neoplasm , Drug Therapy, Combination , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Protein Kinase Inhibitors/pharmacology , Tumor Cells, Cultured , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , Xenograft Model Antitumor Assays
9.
Elife ; 72018 10 05.
Article in English | MEDLINE | ID: mdl-30289386

ABSTRACT

Activation of heterotrimeric G proteins is a key step in many signaling cascades. However, a complete mechanism for this process, which requires allosteric communication between binding sites that are ~30 Å apart, remains elusive. We construct an atomically detailed model of G protein activation by combining three powerful computational methods: metadynamics, Markov state models (MSMs), and CARDS analysis of correlated motions. We uncover a mechanism that is consistent with a wide variety of structural and biochemical data. Surprisingly, the rate-limiting step for GDP release correlates with tilting rather than translation of the GPCR-binding helix 5. ß-Strands 1 - 3 and helix 1 emerge as hubs in the allosteric network that links conformational changes in the GPCR-binding site to disordering of the distal nucleotide-binding site and consequent GDP release. Our approach and insights provide foundations for understanding disease-implicated G protein mutants, illuminating slow events in allosteric networks, and examining unbinding processes with slow off-rates.


Subject(s)
GTP-Binding Proteins/metabolism , Guanosine Diphosphate/metabolism , Molecular Dynamics Simulation , Allosteric Regulation , Binding Sites , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Proteins/chemistry , Guanosine Diphosphate/chemistry , Markov Chains , Probability , Protein Structure, Secondary , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Thermodynamics
10.
Sci Signal ; 11(546)2018 09 04.
Article in English | MEDLINE | ID: mdl-30181242

ABSTRACT

Constitutively active G protein α subunits cause cancer, cholera, Sturge-Weber syndrome, and other disorders. Therapeutic intervention by targeted inhibition of constitutively active Gα subunits in these disorders has yet to be achieved. We found that constitutively active Gαq in uveal melanoma (UM) cells was inhibited by the cyclic depsipeptide FR900359 (FR). FR allosterically inhibited guanosine diphosphate-for-guanosine triphosphate (GDP/GTP) exchange to trap constitutively active Gαq in inactive, GDP-bound Gαßγ heterotrimers. Allosteric inhibition of other Gα subunits was achieved by the introduction of an FR-binding site. In UM cells driven by constitutively active Gαq, FR inhibited second messenger signaling, arrested cell proliferation, reinstated melanocytic differentiation, and stimulated apoptosis. In contrast, FR had no effect on BRAF-driven UM cells. FR promoted UM cell differentiation by reactivating polycomb repressive complex 2 (PRC2)-mediated gene silencing, a heretofore unrecognized effector system of constitutively active Gαq in UM. Constitutively active Gαq and PRC2 therefore provide therapeutic targets for UM. The development of FR analogs specific for other Gα subunit subtypes may provide novel therapeutic approaches for diseases driven by constitutively active Gα subunits or multiple G protein-coupled receptors (GPCRs) where targeting a single receptor is ineffective.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Neoplasms/metabolism , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Depsipeptides/pharmacology , GTP-Binding Protein alpha Subunits/antagonists & inhibitors , HEK293 Cells , Humans , Mice , Neoplasms/pathology , Signal Transduction/drug effects
11.
J Biol Chem ; 292(47): 19266-19278, 2017 11 24.
Article in English | MEDLINE | ID: mdl-28974581

ABSTRACT

Regulator of G protein signaling 2 (RGS2) controls signaling by receptors coupled to the Gq/11 class heterotrimeric G proteins. RGS2 deficiency causes several phenotypes in mice and occurs in several diseases, including hypertension in which a proteolytically unstable RGS2 mutant has been reported. However, the mechanisms and functions of RGS2 proteolysis remain poorly understood. Here we addressed these questions by identifying degradation signals in RGS2, and studying dynamic regulation of Gq/11-evoked Ca2+ signaling and vascular contraction. We identified a novel bipartite degradation signal in the N-terminal domain of RGS2. Mutations disrupting this signal blunted proteolytic degradation downstream of E3 ubiquitin ligase binding to RGS2. Analysis of RGS2 mutants proteolyzed at various rates and the effects of proteasome inhibition indicated that proteolytic degradation controls agonist efficacy by setting RGS2 protein expression levels, and affecting the rate at which cells regain agonist responsiveness as synthesis of RGS2 stops. Analyzing contraction of mesenteric resistance arteries supported the biological relevance of this mechanism. Because RGS2 mRNA expression often is strikingly and transiently up-regulated and then down-regulated upon cell stimulation, our findings indicate that proteolytic degradation tightly couples RGS2 transcription, protein levels, and function. Together these mechanisms provide tight temporal control of Gq/11-coupled receptor signaling in the cardiovascular, immune, and nervous systems.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Mesenteric Arteries/physiology , Muscle Contraction/physiology , RGS Proteins/physiology , Animals , Cells, Cultured , Male , Mesenteric Arteries/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Proteolysis , Signal Transduction
12.
J Biol Chem ; 292(24): 9906-9918, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28432124

ABSTRACT

The R7 regulator of G protein signaling family (R7-RGS) critically regulates nervous system development and function. Mice lacking all R7-RGS subtypes exhibit diverse neurological phenotypes, and humans bearing mutations in the retinal R7-RGS isoform RGS9-1 have vision deficits. Although each R7-RGS subtype forms heterotrimeric complexes with Gß5 and R7-RGS-binding protein (R7BP) that regulate G protein-coupled receptor signaling by accelerating deactivation of Gi/o α-subunits, several neurological phenotypes of R7-RGS knock-out mice are not readily explained by dysregulated Gi/o signaling. Accordingly, we used tandem affinity purification and LC-MS/MS to search for novel proteins that interact with R7-RGS heterotrimers in the mouse brain. Among several proteins detected, we focused on Gα13 because it had not been linked to R7-RGS complexes before. Split-luciferase complementation assays indicated that Gα13 in its active or inactive state interacts with R7-RGS heterotrimers containing any R7-RGS isoform. LARG (leukemia-associated Rho guanine nucleotide exchange factor (GEF)), PDZ-RhoGEF, and p115RhoGEF augmented interaction between activated Gα13 and R7-RGS heterotrimers, indicating that these effector RhoGEFs can engage Gα13·R7-RGS complexes. Because Gα13/R7-RGS interaction required R7BP, we analyzed phenotypes of neuronal cell lines expressing RGS7 and Gß5 with or without R7BP. We found that neurite retraction evoked by Gα12/13-dependent lysophosphatidic acid receptors was augmented in R7BP-expressing cells. R7BP expression blunted neurite formation evoked by serum starvation by signaling mechanisms involving Gα12/13 but not Gαi/o These findings provide the first evidence that R7-RGS heterotrimers interact with Gα13 to augment signaling pathways that regulate neurite morphogenesis. This mechanism expands the diversity of functions whereby R7-RGS complexes regulate critical aspects of nervous system development and function.


Subject(s)
Brain/metabolism , Carrier Proteins/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Neurons/metabolism , RGS Proteins/metabolism , Amino Acid Substitution , Animals , Brain/cytology , Brain/enzymology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , GTP-Binding Protein alpha Subunits, G12-G13/chemistry , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurites/enzymology , Neurons/cytology , Neurons/enzymology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Multimerization , RGS Proteins/chemistry , RGS Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction
13.
Prog Mol Biol Transl Sci ; 133: 77-92, 2015.
Article in English | MEDLINE | ID: mdl-26123303

ABSTRACT

Regulators of G protein signaling (RGS) proteins of the B/R4 family are widely expressed in the cardiovascular system where their role in fine-tuning G protein signaling is critical to maintaining homeostasis. Among members of this family, RGS2 and RGS5 have been shown to play key roles in cardiac and smooth muscle function by tightly regulating signaling pathways that are activated through Gq/11 and Gi/o classes of heterotrimeric G proteins. This chapter reviews accumulating evidence supporting a key role for RGS2 in vascular function and the implication of changes in RGS2 function and/or expression in the pathogenesis of blood pressure disorders, particularly hypertension. With such understanding, RGS2 and the signaling pathways it controls may emerge as novel targets for developing next-generation antihypertensive drugs/agents.


Subject(s)
Blood Vessels/physiology , RGS Proteins/metabolism , Signal Transduction , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular System/metabolism , Cardiovascular System/pathology , Humans , Myocytes, Smooth Muscle/metabolism , RGS Proteins/chemistry , RGS Proteins/genetics
14.
PLoS One ; 10(7): e0132594, 2015.
Article in English | MEDLINE | ID: mdl-26193676

ABSTRACT

Regulator of G protein signaling 2 (RGS2) controls G protein coupled receptor (GPCR) signaling by acting as a GTPase-activating protein for heterotrimeric G proteins. Certain Rgs2 gene mutations have been linked to human hypertension. Renal RGS2 deficiency is sufficient to cause hypertension in mice; however, the pathological mechanisms are unknown. Here we determined how the loss of RGS2 affects renal function. We examined renal hemodynamics and tubular function by monitoring renal blood flow (RBF), glomerular filtration rate (GFR), epithelial sodium channel (ENaC) expression and localization, and pressure natriuresis in wild type (WT) and RGS2 null (RGS2-/-) mice. Pressure natriuresis was determined by stepwise increases in renal perfusion pressure (RPP) and blood flow, or by systemic blockade of nitric oxide synthase with L-NG-Nitroarginine methyl ester (L-NAME). Baseline GFR was markedly decreased in RGS2-/- mice compared to WT controls (5.0 ± 0.8 vs. 2.5 ± 0.1 µl/min/g body weight, p<0.01). RBF was reduced (35.4 ± 3.6 vs. 29.1 ± 2.1 µl/min/g body weight, p=0.08) while renal vascular resistance (RVR; 2.1 ± 0.2 vs. 3.0 ± 0.2 mmHg/µl/min/g body weight, p<0.01) was elevated in RGS2-/- compared to WT mice. RGS2 deficiency caused decreased sensitivity and magnitude of changes in RVR and RBF after a step increase in RPP. The acute pressure-natriuresis curve was shifted rightward in RGS2-/- relative to WT mice. Sodium excretion rate following increased RPP by L-NAME was markedly decreased in RGS2-/- mice and accompanied by increased translocation of ENaC to the luminal wall. We conclude that RGS2 deficiency impairs renal function and autoregulation by increasing renal vascular resistance and reducing renal blood flow. These changes impair renal sodium handling by favoring sodium retention. The findings provide a new line of evidence for renal dysfunction as a primary cause of hypertension.


Subject(s)
Hemodynamics/physiology , Kidney/blood supply , RGS Proteins/metabolism , Renal Circulation/physiology , Animals , Epithelial Sodium Channels/metabolism , Glomerular Filtration Rate/physiology , Kidney/metabolism , Mice , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Natriuresis/physiology , Nitric Oxide Synthase/metabolism , RGS Proteins/genetics
15.
Org Lett ; 17(9): 2270-3, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25875152

ABSTRACT

A simplified analog (WU-07047) of the selective Gαq/11 inhibitor YM-254890 has been synthesized, and an initial probe of its activity conducted. In the analog, the two peptide-based linkers in the cyclic YM-254890 have been replaced with hydrocarbon chains. This enables a convergent approach to the synthesis of the analog. Biochemical assays showed that while the simplified analog is not as potent as YM-254890, it does still inhibit Gq.


Subject(s)
GTP-Binding Proteins/agonists , Peptides, Cyclic/chemical synthesis , Crystallography, X-Ray , GTP-Binding Proteins/chemistry , Molecular Structure , Peptides, Cyclic/chemistry , Signal Transduction
16.
Tomography ; 1(1): 61-68, 2015 Sep.
Article in English | MEDLINE | ID: mdl-30042955

ABSTRACT

The goal of this work was to demonstrate the utility of Bayesian probability theory-based model selection for choosing the optimal mathematical model from among 4 competing models of renal dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) data. DCE-MRI data were collected on 21 mice with high (n = 7), low (n = 7), or normal (n = 7) renal blood flow (RBF). Model parameters and posterior probabilities of 4 renal DCE-MRI models were estimated using Bayesian-based methods. Models investigated included (1) an empirical model that contained a monoexponential decay (washout) term and a constant offset, (2) an empirical model with a biexponential decay term (empirical/biexponential model), (3) the Patlak-Rutland model, and (4) the 2-compartment kidney model. Joint Bayesian model selection/parameter estimation demonstrated that the empirical/biexponential model was strongly favored for all 3 cohorts, the modeled DCE signals that characterized each of the 3 cohorts were distinctly different, and individual empirical/biexponential model parameter values clearly distinguished cohorts of low and high RBF from one another. The Bayesian methods can be readily extended to a variety of model analyses, making it a versatile and valuable tool for model selection and parameter estimation.

17.
Proc Natl Acad Sci U S A ; 111(37): 13379-84, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25139991

ABSTRACT

Endothelial cells (ECs) express fibroblast growth factor receptors (FGFRs) and are exquisitely sensitive to FGF signals. However, whether the EC or another vascular cell type requires FGF signaling during development, homeostasis, and response to injury is not known. Here, we show that Flk1-Cre or Tie2-Cre mediated deletion of FGFR1 and FGFR2 (Fgfr1/2(Flk1-Cre) or Fgfr1/2(Tie2-Cre) mice), which results in deletion in endothelial and hematopoietic cells, is compatible with normal embryonic development. As adults, Fgfr1/2(Flk1-Cre) mice maintain normal blood pressure and vascular reactivity and integrity under homeostatic conditions. However, neovascularization after skin or eye injury was significantly impaired in both Fgfr1/2(Flk1-Cre) and Fgfr1/2(Tie2-Cre) mice, independent of either hematopoietic cell loss of FGFR1/2 or vascular endothelial growth factor receptor 2 (Vegfr2) haploinsufficiency. Also, impaired neovascularization was associated with delayed cutaneous wound healing. These findings reveal a key requirement for cell-autonomous EC FGFR signaling in injury-induced angiogenesis, but not for vascular homeostasis, identifying the EC FGFR signaling pathway as a target for diseases associated with aberrant vascular proliferation, such as age-related macular degeneration, and for modulating wound healing without the potential toxicity associated with direct manipulation of systemic FGF or VEGF activity.


Subject(s)
Blood Vessels/pathology , Endothelial Cells/metabolism , Fibroblast Growth Factors/metabolism , Homeostasis , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Animals , Animals, Newborn , Capillary Permeability , Enzyme Activation , Eye/pathology , Hematopoiesis , Hypoxia/metabolism , Hypoxia/pathology , Integrases/metabolism , Mice , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic , Stress, Physiological , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing
18.
Am J Physiol Heart Circ Physiol ; 306(5): H654-66, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24414067

ABSTRACT

Elastin (Eln) insufficiency in mice and humans is associated with hypertension and altered structure and mechanical properties of large arteries. However, it is not known to what extent functional or structural changes in resistance arteries contribute to the elevated blood pressure that is characteristic of Eln insufficiency. Here, we investigated how Eln insufficiency affects the structure and function of the resistance vasculature. A functional profile of resistance vasculature in Eln(+/-) mice was generated by assessing small mesenteric artery (MA) contractile and vasodilatory responses to vasoactive agents. We found that Eln haploinsufficiency had a modest effect on phenylephrine-induced vasoconstriction, whereas ANG II-evoked vasoconstriction was markedly increased. Blockade of ANG II type 2 receptors with PD-123319 or modulation of Rho kinase activity with the inhibitor Y-27632 attenuated the augmented vasoconstriction, whereas acute Y-27632 administration normalized blood pressure in Eln(+/-) mice. Sodium nitroprusside- and isoproterenol-induced vasodilatation were normal, whereas ACh-induced vasodilatation was severely impaired in Eln(+/-) MAs. Histologically, the number of smooth muscle layers did not change in Eln(+/-) MAs; however, an additional discontinuous layer of Eln appeared between the smooth muscle layers that was absent in wild-type arteries. We conclude that high blood pressure arising from Eln insufficiency is due partly to permanent changes in vascular tone as a result of increased sensitivity of the resistance vasculature to circulating ANG II and to impaired vasodilatory mechanisms arising from endothelial dysfunction characterized by impaired endothelium-dependent vasodilatation. Eln insufficiency causes augmented ANG II-induced vasoconstriction in part through a novel mechanism that facilitates contraction evoked by ANG II type 2 receptors and altered G protein signaling.


Subject(s)
Arterial Pressure , Elastin/deficiency , Hypertension/metabolism , Mesenteric Arteries/metabolism , Vascular Resistance/drug effects , Vasoconstriction , Vasodilation , Angiotensin II/metabolism , Animals , Arterial Pressure/drug effects , Calcium/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Elastin/genetics , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Genetic Predisposition to Disease , Haploinsufficiency , Hemizygote , Hypertension/drug therapy , Hypertension/genetics , Hypertension/pathology , Hypertension/physiopathology , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/pathology , Mesenteric Arteries/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Kinase Inhibitors/pharmacology , Receptor, Angiotensin, Type 2/drug effects , Receptor, Angiotensin, Type 2/metabolism , Signal Transduction/drug effects , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
19.
J Biol Chem ; 289(9): 6249-57, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24385443

ABSTRACT

Reversible attachment and removal of palmitate or other long-chain fatty acids on proteins has been hypothesized, like phosphorylation, to control diverse biological processes. Indeed, palmitate turnover regulates Ras trafficking and signaling. Beyond this example, however, the functions of palmitate turnover on specific proteins remain poorly understood. Here, we show that a mechanism regulating G protein-coupled receptor signaling in neuronal cells requires palmitate turnover. We used hexadecyl fluorophosphonate or palmostatin B to inhibit enzymes in the serine hydrolase family that depalmitoylate proteins, and we studied R7 regulator of G protein signaling (RGS)-binding protein (R7BP), a palmitoylated allosteric modulator of R7 RGS proteins that accelerate deactivation of Gi/o class G proteins. Depalmitoylation inhibition caused R7BP to redistribute from the plasma membrane to endomembrane compartments, dissociated R7BP-bound R7 RGS complexes from Gi/o-gated G protein-regulated inwardly rectifying K(+) (GIRK) channels and delayed GIRK channel closure. In contrast, targeting R7BP to the plasma membrane with a polybasic domain and an irreversibly attached lipid instead of palmitate rendered GIRK channel closure insensitive to depalmitoylation inhibitors. Palmitate turnover therefore is required for localizing R7BP to the plasma membrane and facilitating Gi/o deactivation by R7 RGS proteins on GIRK channels. Our findings broaden the scope of biological processes regulated by palmitate turnover on specific target proteins. Inhibiting R7BP depalmitoylation may provide a means of enhancing GIRK activity in neurological disorders.


Subject(s)
Carrier Proteins/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Lipoylation/physiology , Protein Processing, Post-Translational/physiology , RGS Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Carrier Proteins/genetics , Cell Line, Tumor , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Humans , Intracellular Signaling Peptides and Proteins , Lipoylation/drug effects , Mice , Propiolactone/analogs & derivatives , Propiolactone/pharmacology , Protein Processing, Post-Translational/drug effects , RGS Proteins/genetics , Receptors, G-Protein-Coupled/genetics
20.
PLoS One ; 8(12): e82276, 2013.
Article in English | MEDLINE | ID: mdl-24349243

ABSTRACT

In the outer retina, G protein-coupled receptor (GPCR) signaling mediates phototransduction and synaptic transmission between photoreceptors and ON bipolar cells. In contrast, the functions of modulatory GPCR signaling networks in the inner retina are less well understood. We addressed this question by determining the consequences of augmenting modulatory Gi/o signaling driven by endogenous transmitters. This was done by analyzing the effects of genetically ablating the R7 RGS-binding protein (R7BP), a membrane-targeting protein and positive allosteric modulator of R7-RGS (regulator of the G protein signaling 7) family that deactivates Gi/oα subunits. We found that R7BP is expressed highly in starburst amacrine cells and retinal ganglion cells (RGCs). As indicated by electroretinography and multielectrode array recordings of adult retina, ablation of R7BP preserved outer retina function, but altered the firing rate and latency of ON RGCs driven by rods and cones but not rods alone. In developing retina, R7BP ablation increased the burst duration of glutamatergic waves whereas cholinergic waves were unaffected. This effect on glutamatergic waves did not result in impaired segregation of RGC projections to eye-specific domains of the dorsal lateral geniculate nucleus. R7BP knockout mice exhibited normal spatial contrast sensitivity and visual acuity as assessed by optomotor reflexes. Taken together these findings indicate that R7BP-dependent regulation of R7-RGS proteins shapes specific aspects of light-evoked and spontaneous activity of RGCs in mature and developing retina.


Subject(s)
Glutamates/metabolism , Light , RGS Proteins/metabolism , Retina/metabolism , Retina/radiation effects , Action Potentials/radiation effects , Allosteric Regulation/radiation effects , Amacrine Cells/metabolism , Amacrine Cells/radiation effects , Animals , Animals, Newborn , Choline/metabolism , Contrast Sensitivity/radiation effects , Gene Deletion , Mice , Mice, Inbred C57BL , RGS Proteins/deficiency , Retinal Bipolar Cells/metabolism , Retinal Bipolar Cells/radiation effects , Retinal Cone Photoreceptor Cells/metabolism , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/radiation effects , Retinal Rod Photoreceptor Cells/metabolism , Transcription Factor Brn-3A/metabolism , Visual Acuity/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...