Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
J Mol Med (Berl) ; 98(1): 97-110, 2020 01.
Article in English | MEDLINE | ID: mdl-31811326

ABSTRACT

Pertussis toxin (PTX) is a potent virulence factor in patients suffering from whooping cough, but in its detoxified version, it is applied for vaccination. It is thought to contribute to the pathology of the disease including various CNS malfunctions. Based on its enzymatic activity, PTX disrupts GPCR-dependent signaling by modifying the α-subunit of heterotrimeric Gi/o-proteins. It is also extensively used as a research tool to study neuronal functions in vivo and in vitro. However, data demonstrating the penetration of PTX from the blood into the brain are missing. Here, we examined the Gαi/o-modifying activity of PTX in murine brains after its parenteral application. Ex vivo biodistribution analysis of [124I]-PTX displayed poor distribution to the brain while relatively high concentrations were visible in the pancreas. PTX affected CNS and endocrine functions of the pancreas as shown by open-field and glucose tolerance tests, respectively. However, while pancreatic islet Gαi/o-proteins were modified, their neuronal counterparts in brain tissue were resistant towards PTX as indicated by different autoradiographic and immunoblot SDS-PAGE analyses. In contrast, PTX easily modified brain Gαi/o-proteins ex vivo. An attempt to increase BBB permeability by application of hypertonic mannitol did not show PTX activity on neuronal G proteins. Consistent with these findings, in vivo MRI analysis did not point to an increased blood-brain barrier (BBB) permeability following PTX treatment. Our data demonstrate that the CNS is protected from PTX. Thus, we hypothesize that the BBB hinders PTX to penetrate into the CNS and to deliver its enzymatic activity to brain Gαi/o-proteins. KEY MESSAGES: i.p. applied PTX is poorly retained in the brain while reaches high concentration in the pancreas. Pancreatic islet Gαi/o- but not cerebral Gαi/o-proteins are modified by i.p. administered PTX. Gαi/o-proteins from isolated cerebral cell membranes were easily modified by PTX ex vivo. CNS is protected from i.p. administered PTX. PTX does not permeabilize the BBB.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Injections/methods , Neuroprotection , Pertussis Toxin/administration & dosage , Pertussis Toxin/metabolism , Signal Transduction/drug effects , Animals , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/metabolism , Capillary Permeability/drug effects , Cell Membrane/metabolism , Female , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , Iodine Radioisotopes , Islets of Langerhans/diagnostic imaging , Islets of Langerhans/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Tissue Distribution
3.
Nat Commun ; 8(1): 1706, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29167435

ABSTRACT

The ß1 adrenergic receptor (ß1AR) is recognized as a classical Gαs-coupled receptor. Agonist binding not only initiates G protein-mediated signaling but also signaling through the multifunctional adapter protein ß-arrestin. Some ßAR ligands, such as carvedilol, stimulate ßAR signaling preferentially through ß-arrestin, a concept known as ß-arrestin-biased agonism. Here, we identify a signaling mechanism, unlike that previously known for any Gαs-coupled receptor, whereby carvedilol induces the transition of the ß1AR from a classical Gαs-coupled receptor to a Gαi-coupled receptor stabilizing a distinct receptor conformation to initiate ß-arrestin-mediated signaling. Recruitment of Gαi is not induced by any other ßAR ligand screened, nor is it required for ß-arrestin-bias activated by the ß2AR subtype of the ßAR family. Our findings demonstrate a previously unrecognized role for Gαi in ß1AR signaling and suggest that the concept of ß-arrestin-bias may need to be refined to incorporate the selective bias of receptors towards distinct G protein subtypes.


Subject(s)
Carbazoles/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Propanolamines/pharmacology , Receptors, Adrenergic, beta-1/metabolism , beta-Arrestins/metabolism , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Carvedilol , Female , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Ligands , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Knockout , Protein Conformation/drug effects , Receptors, Adrenergic, beta-1/chemistry , beta-Arrestins/antagonists & inhibitors , beta-Arrestins/genetics
4.
Sci Rep ; 7(1): 4156, 2017 06 23.
Article in English | MEDLINE | ID: mdl-28646160

ABSTRACT

Thymocyte and T cell trafficking relies on signals initiated by G-protein coupled receptors. To address the importance of the G-proteins Gαi2 and Gαi3 in thymocyte and T cell function, we developed several mouse models. Gαi2 deficiency in hematopoietic progenitors led to a small thymus, a double negative (DN)1/DN2 thymocyte transition block, and an accumulation of mature single positive (SP) thymocytes. Loss at the double positive (DP) stage of thymocyte development caused an increase in mature cells within the thymus. In both models an abnormal distribution of memory and naïve CD4 T cells occurred, and peripheral CD4 and CD8 T cells had reduced chemoattractant responses. The loss of Gαi3 had no discernable impact, however the lack of both G-proteins commencing at the DP stage caused a severe T cell phenotype. These mice lacked a thymic medullary region, exhibited thymocyte retention, had a peripheral T cell deficiency, and lacked T cell chemoattractant responses. Yet a noteworthy population of CD4+PD-1+CXCR5+/- cells resided in the spleen of these mice likely due to a loss of regulatory T cell function. Our results delineate a role for Gαi2 in early thymocyte development and for Gαi2/3 in multiple aspects of T cell biology.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Cell Movement , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , Programmed Cell Death 1 Receptor/metabolism , Receptors, CXCR5/metabolism , Spleen/cytology , Thymocytes/cytology , Animals , Cell Compartmentation , Cell Proliferation , Chemokines/pharmacology , DNA-Binding Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gene Deletion , Hematopoietic Stem Cells/metabolism , Integrases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Thymocytes/metabolism
5.
J Am Heart Assoc ; 5(11)2016 10 26.
Article in English | MEDLINE | ID: mdl-27912212

ABSTRACT

BACKGROUND: We previously showed that the levels of both Giα-2 and Giα-3 proteins were augmented in spontaneously hypertensive rats (SHRs) before the onset of hypertension. In addition, intraperitoneal injection of pertussis toxin, which inactivates both Giα proteins, prevented the development of hypertension in SHRs. The aim of the present study was to determine the specific contributions of Giα-2 and Giα-3 proteins to the development of hypertension. METHODS AND RESULTS: Antisense oligodeoxynucleotide of Giα-2 and Giα-3 encapsulated in PEG/DOTAP/DOPE cationic liposomes were administrated intravenously into 3-week-old prehypertensive SHRs and Wistar Kyoto rats, whereas the control Wistar Kyoto rats and SHRs received PBS, empty liposomes, or sense. The knockdown of Giα-2 but not Giα-3 protein attenuated tachycardia and prevented the development of hypertension up to age 6 weeks; thereafter, blood pressure started increasing and reached the same level as that of untreated SHRs at 9 weeks. Furthermore, Giα-2 and Giα-3 antisense oligodeoxynucleotide treatments significantly decreased the enhanced levels of Giα-2 and Giα-3 proteins, respectively, and enhanced levels of superoxide anion and NADPH oxidase activity in heart, aorta, and kidney and hyperproliferation of vascular smooth muscle cells from SHRs aged 6 weeks. In addition, antisense oligodeoxynucleotide treatment with Giα-2 but not Giα-3 restored enhanced inhibition of adenylyl cyclase by oxotremorine to WKY levels. CONCLUSIONS: These results suggested that the enhanced expression of Giα-2 but not Giα-3 protein plays an important role in the pathogenesis of hypertension and tachycardia in SHRs.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Hypertension/prevention & control , Oligodeoxyribonucleotides, Antisense/physiology , Tachycardia/prevention & control , Adenylyl Cyclase Inhibitors/pharmacology , Animals , Aorta/metabolism , Blood Pressure/physiology , Cells, Cultured , Disease Models, Animal , GTP-Binding Protein alpha Subunit, Gi2/deficiency , GTP-Binding Protein alpha Subunit, Gi2/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , Gene Knockdown Techniques , Heart Rate/physiology , Kidney/metabolism , Liposomes/administration & dosage , Male , Muscle, Smooth, Vascular/metabolism , Myocardium/metabolism , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction/physiology , Transfection/methods
6.
Proc Natl Acad Sci U S A ; 113(39): 10974-9, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27625425

ABSTRACT

In mammals, initial detection of olfactory stimuli is mediated by sensory neurons in the main olfactory epithelium (MOE) and the vomeronasal organ (VNO). The heterotrimeric GTP-binding protein Go is widely expressed in the MOE and VNO of mice. Early studies indicated that Go expression in VNO sensory neurons is critical for directing social and sexual behaviors in female mice [Oboti L, et al. (2014) BMC Biol 12:31]. However, the physiological functions of Go in the MOE have remained poorly defined. Here, we examined the role of Go in the MOE using mice lacking the α subunit of Go Development of the olfactory bulb (OB) was perturbed in mutant mice as a result of reduced neurogenesis and increased cell death. The balance between cell types of OB interneurons was altered in mutant mice, with an increase in the number of tyrosine hydroxylase-positive interneurons at the expense of calbindin-positive interneurons. Sexual behavior toward female mice and preference for female urine odors by olfactory sensory neurons in the MOE were abolished in mutant male mice. Our data suggest that Go signaling is essential for the structural and functional integrity of the MOE and for specification of OB interneurons, which in turn are required for the transmission of pheromone signals and the initiation of mating behavior with the opposite sex.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Olfactory Mucosa/metabolism , Sexual Behavior, Animal , Animals , Apoptosis/genetics , Cell Count , Cells, Cultured , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Gene Deletion , Interneurons/metabolism , Male , Mice , Models, Biological , Neurogenesis/genetics , Olfactory Bulb/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Tyrosine 3-Monooxygenase/metabolism , Vomeronasal Organ/metabolism
7.
Eur J Pharmacol ; 780: 115-21, 2016 Jun 05.
Article in English | MEDLINE | ID: mdl-27025291

ABSTRACT

Mast cells activated by IgE-dependent and -independent mechanisms play important roles in innate and acquired immune responses. Activation of pertussis toxin (PTX)-sensitive Gi/o proteins is the key step in mast cell degranulation and release of de novo synthesized inflammatory mediators through IgE-independent mechanism. However, the roles of Gi and Go proteins in mast cells activation have not yet been differentiated. In the current study, the functional roles of Go proteins in the activities of LAD2 cells, a human mast cell line, are identified. Knockdown of Gαo expression significantly inhibited the synthesis of IL-8 and TNF-α from substance P activated LAD2 cells but demonstrated no effect on degranulation. This effect was associated with the activation of Erk and JNK/MAPKs signaling, whereas PI3K-Akt, calcium mobilization and NFAT translocation remained unchanged. These results suggest that Gi and Go proteins differentially regulate human mast cells activities through activating distinct signaling cascades.


Subject(s)
Cell Degranulation , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Interleukin-8/metabolism , Mast Cells/cytology , Mast Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Calcium/metabolism , Cell Degranulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Gene Knockdown Techniques , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mast Cells/drug effects , NFATC Transcription Factors/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Signal Transduction/drug effects , Substance P/pharmacology
8.
Clinics ; 70(3): 157-161, 03/2015. tab
Article in English | LILACS | ID: lil-747105

ABSTRACT

OBJECTIVE: To determine whether Tai Chi Chuan or ballroom dancing promotes better performance with respect to postural balance, gait, and postural transfer among elderly people. METHODS: We evaluated 76 elderly individuals who were divided into two groups: the Tai Chi Chuan Group and the Dance Group. The subjects were tested using the NeuroCom Balance Master¯ force platform system with the following protocols: static balance tests (the Modified Clinical Tests of Sensory Interaction on Balance and Unilateral Stance) and dynamic balance tests (the Walk Across Test and Sit-to-stand Transfer Test). RESULTS: In the Modified Clinical Test of Sensory Interaction on Balance, the Tai Chi Chuan Group presented a lower sway velocity on a firm surface with open and closed eyes, as well as on a foam surface with closed eyes. In the Modified Clinical Test of Sensory Interaction on Unilateral Stance, the Tai Chi Chuan Group presented a lower sway velocity with open eyes, whereas the Dance Group presented a lower sway velocity with closed eyes. In the Walk Across Test, the Tai Chi Chuan Group presented faster walking speeds than those of the Dance Group. In the Sit-to-stand Transfer Test, the Tai Chi Chuan Group presented shorter transfer times from the sitting to the standing position, with less sway in the final standing position. CONCLUSION: The elderly individuals who practiced Tai Chi Chuan had better bilateral balance with eyes open on both types of surfaces compared with the Dance Group. The Dance Group had better unilateral postural balance with eyes closed. The Tai Chi Chuan Group had faster walking speeds, shorter transfer times, and better postural balance in the final standing position during the Sit-to-stand Test. .


Subject(s)
/metabolism , Cyclic AMP/metabolism , Dictyostelium/enzymology , Dictyostelium/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Protozoan Proteins/metabolism , /genetics , Dictyostelium/growth & development , Dictyostelium/metabolism , Down-Regulation/drug effects , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Folic Acid/pharmacology , /deficiency , /genetics , /metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Mutation , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Protozoan Proteins/genetics , Signal Transduction , Spores, Protozoan/enzymology , Spores, Protozoan/genetics , Vitamin B Complex/pharmacology
9.
Cell Signal ; 26(2): 409-18, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24511612

ABSTRACT

Cyclic AMP (cAMP) is an important intracellular signaling molecule for many G protein-mediated signaling pathways but the specificity of cAMP signaling in cells with multiple signaling pathways is not well-understood. In Dictyostelium, at least two different G protein signaling pathways, mediated by the Gα2 and Gα4 subunits, are involved with cAMP accumulation, spore production, and chemotaxis and the stimulation of these pathways results in the activation of ERK2, a mitogen-activated protein kinase that can down regulate the cAMP-specific phosphodiesterase RegA. The regA gene was disrupted in gα2(−) and gα4(−) cells to determine if the absence of this phosphodiesterase rescues the development of these G protein mutants as it does for erk2(−) mutants. There gA(−) mutation had no major effects on developmental morphology but enriched the distribution of the Gα mutant cells to the prespore/prestalk border in chimeric aggregates. The loss of RegA function had no effect on Gα4- mediated folate chemotaxis. However, the regA gene disruption in gα4(−) cells, but not in gα2(−) cells, resulted in a substantial rescue and acceleration of spore production. This rescue in sporulation required cell autonomous signaling because the precocious sporulation could not be induced through intercellular signaling in chimeric aggregates. However, intercellular signals from regA(−) strains increased the expression of the prestalk gene ecmB and accelerated the vacuolization of stalk cells. Intercellular signaling from the gα4(−)regA(−) strain did not induce ecmA gene expression indicating cell-type specificity in the promotion of prestalk cell development. regA gene disruption in a Gα4(HC) (Gα4 overexpression) strain did not result in precocious sporulation or stalk cell development indicating that elevated Gα4 subunit expression can mask regA(−) associated phenotypes even when provided with wild-type intercellular signaling. These findings indicate that the Gα2 and Gα4-mediated pathways provide different contributions to the development of spores and stalk cells and that the absence of RegA function can bypass some but not all defects in G protein regulated spore development.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cyclic AMP/metabolism , Dictyostelium/enzymology , Dictyostelium/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Protozoan Proteins/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Dictyostelium/growth & development , Dictyostelium/metabolism , Down-Regulation/drug effects , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Folic Acid/pharmacology , GTP-Binding Protein alpha Subunit, Gi2/deficiency , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mutation , Protozoan Proteins/genetics , Signal Transduction , Spores, Protozoan/enzymology , Spores, Protozoan/genetics , Vitamin B Complex/pharmacology
10.
PLoS One ; 8(8): e72596, 2013.
Article in English | MEDLINE | ID: mdl-23977324

ABSTRACT

B lymphocytes are compartmentalized within lymphoid organs. The organization of these compartments depends upon signaling initiated by G-protein linked chemoattractant receptors. To address the importance of the G-proteins Gαi2 and Gαi3 in chemoattractant signaling we created mice lacking both proteins in their B lymphocytes. While bone marrow B cell development and egress is grossly intact; mucosal sites, splenic marginal zones, and lymph nodes essentially lack B cells. There is a partial block in splenic follicular B cell development and a 50-60% reduction in splenic B cells, yet normal numbers of splenic T cells. The absence of Gαi2 and Gαi3 in B cells profoundly disturbs the architecture of lymphoid organs with loss of B cell compartments in the spleen, thymus, lymph nodes, and gastrointestinal tract. This results in a severe disruption of B cell function and a hyper-IgM like syndrome. Beyond the pro-B cell stage, B cells are refractory to chemokine stimulation, and splenic B cells are poorly responsive to antigen receptor engagement. Gαi2 and Gαi3 are therefore critical for B cell chemoattractant receptor signaling and for normal B cell function. These mice provide a worst case scenario of the consequences of losing chemoattractant receptor signaling in B cells.


Subject(s)
B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Compartmentation , GTP-Binding Protein alpha Subunit, Gi2/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , Hyper-IgM Immunodeficiency Syndrome/immunology , Hyper-IgM Immunodeficiency Syndrome/pathology , Animals , Antibody Formation/drug effects , B-Lymphocytes/drug effects , Bone Marrow/drug effects , Bone Marrow/pathology , Cell Compartmentation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemokines/metabolism , Chemotactic Factors/pharmacology , Disease Progression , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Germinal Center/drug effects , Germinal Center/pathology , Immunization , Integrases/metabolism , Integrins/metabolism , Lipopolysaccharides/pharmacology , Lymph Nodes/drug effects , Lymph Nodes/pathology , Lymphocyte Count , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/metabolism , Mice, Inbred C57BL , Mice, Knockout , Peritoneum/drug effects , Peritoneum/pathology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/drug effects , Spleen/pathology
11.
Proc Natl Acad Sci U S A ; 109(52): 21366-71, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23236180

ABSTRACT

129/SvEv mice with a loss-of-function mutation in the heterotrimeric G protein α-subunit gene Gnai3 have fusions of ribs and lumbar vertebrae, indicating a requirement for Gα(i) (the "inhibitory" class of α-subunits) in somite derivatives. Mice with mutations of Gnai1 or Gnai2 have neither defect, but loss of both Gnai3 and one of the other two genes increases the number and severity of rib fusions without affecting the lumbar fusions. No myotome defects are observed in Gnai3/Gnai1 double-mutant embryos, and crosses with a conditional allele of Gnai2 indicate that Gα(i) is specifically required in cartilage precursors. Penetrance and expressivity of the rib fusion phenotype is altered in mice with a mixed C57BL/6 × 129/SvEv genetic background. These phenotypes reveal a previously unknown role for G protein-coupled signaling pathways in development of the axial skeleton.


Subject(s)
Bone Development , Bone and Bones/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Mammals/growth & development , Signal Transduction , Alleles , Animals , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Lumbar Vertebrae/abnormalities , Lumbar Vertebrae/growth & development , Mice , Mice, 129 Strain , Mice, Mutant Strains , Mutation/genetics , Phenotype , Ribs/abnormalities , Ribs/growth & development , Spinal Fusion , Sternum/abnormalities , Sternum/growth & development
12.
J Immunol ; 189(2): 980-7, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22706085

ABSTRACT

Various heterotrimeric G(i) proteins are considered to be involved in cell migration and effector function of immune cells. The underlying mechanisms, how they control the activation of myeloid effector cells, are not well understood. To elucidate isoform-redundant and -specific roles for Gα(i) proteins in these processes, we analyzed mice genetically deficient in Gα(i2) or Gα(i3). First, we show an altered distribution of tissue macrophages and blood monocytes in the absence of Gα(i2) but not Gα(i3). Gα(i2)-deficient but not wild-type or Gα(i3)-deficient mice exhibited reduced recruitment of macrophages in experimental models of thioglycollate-induced peritonitis and LPS-triggered lung injury. In contrast, genetic ablation of Gα(i2) had no effect on Gα(i)-dependent peritoneal cytokine production in vitro and the phagocytosis-promoting function of the Gα(i)-coupled C5a anaphylatoxin receptor by liver macrophages in vivo. Interestingly, actin rearrangement and CCL2- and C5a anaphylatoxin receptor-induced chemotaxis but not macrophage CCR2 and C5a anaphylatoxin receptor expression were reduced in the specific absence of Gα(i2). Furthermore, knockdown of Gα(i2) caused decreased cell migration and motility of RAW 264.7 cells, which was rescued by transfection of Gα(i2) but not Gα(i3). These results indicate that Gα(i2), albeit redundant to Gα(i3) in some macrophage activation processes, clearly exhibits a Gα(i) isoform-specific role in the regulation of macrophage migration.


Subject(s)
Cell Migration Inhibition/immunology , GTP-Binding Protein alpha Subunit, Gi2/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Macrophages/immunology , Macrophages/pathology , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Cell Migration Inhibition/genetics , GTP-Binding Protein alpha Subunit, Gi2/genetics , Lipopolysaccharides/toxicity , Macrophages/metabolism , Mice , Mice, 129 Strain , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Peritonitis/chemically induced , Peritonitis/immunology , Peritonitis/pathology , Thioglycolates/toxicity
13.
PLoS One ; 6(9): e24376, 2011.
Article in English | MEDLINE | ID: mdl-21931697

ABSTRACT

BACKGROUND: Ocular albinism type 1, an X-linked disease characterized by the presence of enlarged melanosomes in the retinal pigment epithelium (RPE) and abnormal crossing of axons at the optic chiasm, is caused by mutations in the OA1 gene. The protein product of this gene is a G-protein-coupled receptor (GPCR) localized in RPE melanosomes. The Oa1-/- mouse model of ocular albinism reproduces the human disease. Oa1 has been shown to immunoprecipitate with the Gαi subunit of heterotrimeric G proteins from human skin melanocytes. However, the Gαi subfamily has three highly homologous members, Gαi1, Gαi2 and Gαi3 and it is possible that one or more of them partners with Oa1. We had previously shown by in-vivo studies that Gαi3-/- and Oa1-/- mice have similar RPE phenotype and decussation patterns. In this paper we analyze the specificity of the Oa1-Gαi interaction. METHODOLOGY: By using the genetic mouse models Gαi1-/-, Gαi2-/-, Gαi3-/- and the double knockout Gαi1-/-, Gαi3-/- that lack functional Gαi1, Gαi2, Gαi3, or both Gαi1 and Gαi3 proteins, respectively, we show that Gαi3 is critical for the maintenance of a normal melanosomal phenotype and that its absence is associated with changes in melanosomal size and density. GST-pull-down and immunoprecipitation assays conclusively demonstrate that Gαi3 is the only Gαi that binds to Oa1. Western blots show that Gαi3 expression is barely detectable in the Oa1-/- RPE, strongly supporting a previously unsuspected role for Gαi3 in melanosomal biogenesis. CONCLUSION: Our results identify the Oa1 transducer Gαi3 as the first downstream component in the Oa1 signaling pathway.


Subject(s)
Eye Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Melanosomes/metabolism , Membrane Glycoproteins/metabolism , Organelle Size , Receptors, G-Protein-Coupled/metabolism , Retinal Pigment Epithelium/metabolism , Adenosine Diphosphate Ribose/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Chromatography, Liquid , Electroretinography , Eye Proteins/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , Humans , Mass Spectrometry , Melanosomes/ultrastructure , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Molecular Sequence Data , Organelle Shape , Protein Binding , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/deficiency , Retinal Pigment Epithelium/physiology , Retinal Pigment Epithelium/ultrastructure
14.
Neuropsychopharmacology ; 36(10): 2041-53, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21654736

ABSTRACT

Opioid analgesics elicit their effects via activation of the mu-opioid receptor (MOR), a G protein-coupled receptor known to interact with Gα(i/o)-type G proteins. Work in vitro has suggested that MOR couples preferentially to the abundant brain Gα(i/o) isoform, Gα(o). However, studies in vivo evaluating morphine-mediated antinociception have not supported these findings. The aim of the present work was to evaluate the contribution of Gα(o) to MOR-dependent signaling by measuring both antinociceptive and biochemical endpoints in a Gα(o) null transgenic mouse strain. Male wild-type and Gα(o) heterozygous null (Gα(o) ⁺/⁻) mice were tested for opioid antinociception in the hot plate test or the warm-water tail withdrawal test as measures of supraspinal or spinal antinociception, respectively. Reduction in Gα(o) levels attenuated the supraspinal antinociception produced by morphine, methadone, and nalbuphine, with the magnitude of suppression dependent on agonist efficacy. This was explained by a reduction in both high-affinity MOR expression and MOR agonist-stimulated G protein activation in whole brain homogenates from Gα(o) ⁺/⁻ and Gα(o) homozygous null (Gα(o)⁻/⁻) mice, compared with wild-type littermates. On the other hand, morphine spinal antinociception was not different between Gα(o) ⁺/⁻ and wild-type mice and high-affinity MOR expression was unchanged in spinal cord tissue. However, the action of the partial agonist nalbuphine was compromised, showing that reduction in Gα(o) protein does decrease spinal antinociception, but suggesting a higher Gα(o) protein reserve. These results provide the first in vivo evidence that Gα(o) contributes to maximally efficient MOR signaling and antinociception.


Subject(s)
Analgesics, Opioid/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Pain Measurement , Receptors, Opioid, mu/metabolism , Animals , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Male , Mice , Mice, 129 Strain , Mice, Knockout , Mice, Transgenic , Pain Measurement/drug effects , Pain Measurement/methods , Protein Binding/genetics , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/physiology , Signal Transduction/genetics
15.
J Neurosci ; 30(14): 5092-101, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20371829

ABSTRACT

The rapidly activating and inactivating voltage-gated K(+) (Kv) current, I(A), is broadly expressed in neurons and is a key regulator of action potential repolarization, repetitive firing, backpropagation (into dendrites) of action potentials, and responses to synaptic inputs. Interestingly, results from previous studies on a number of neuronal cell types, including hippocampal, cortical, and spinal neurons, suggest that macroscopic I(A) is composed of multiple components and that each component is likely encoded by distinct Kv channel alpha-subunits. The goals of the experiments presented here were to test this hypothesis and to determine the molecular identities of the Kv channel alpha-subunits that generate I(A) in cortical pyramidal neurons. Combining genetic disruption of individual Kv alpha-subunit genes with pharmacological approaches to block Kv currents selectively, the experiments here revealed that Kv1.4, Kv4.2, and Kv4.3 alpha-subunits encode distinct components of I(A) that together underlie the macroscopic I(A) in mouse (male and female) cortical pyramidal neurons. Recordings from neurons lacking both Kv4.2 and Kv4.3 (Kv4.2(-/-)/Kv4.3(-/-)) revealed that, although Kv1.4 encodes a minor component of I(A), the Kv1.4-encoded current was found in all the Kv4.2(-/-)/Kv4.3(-/-) cortical pyramidal neurons examined. Of the cortical pyramidal neurons lacking both Kv4.2 and Kv1.4, 90% expressed a Kv4.3-encoded I(A) larger in amplitude than the Kv1.4-encoded component. The experimental findings also demonstrate that the targeted deletion of the individual Kv alpha-subunits encoding components of I(A) results in electrical remodeling that is Kv alpha-subunit specific.


Subject(s)
Cerebral Cortex/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Ion Channel Gating/genetics , Kv1.4 Potassium Channel/genetics , Protein Subunits/genetics , Pyramidal Cells/physiology , Shal Potassium Channels/genetics , Action Potentials/drug effects , Action Potentials/genetics , Animals , Cerebral Cortex/drug effects , Female , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , Gene Targeting , Kv1.4 Potassium Channel/antagonists & inhibitors , Kv1.4 Potassium Channel/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/physiology , Potassium Channel Blockers/pharmacology , Protein Subunits/antagonists & inhibitors , Protein Subunits/deficiency , Pyramidal Cells/drug effects , Shal Potassium Channels/antagonists & inhibitors , Shal Potassium Channels/deficiency
16.
Am J Physiol Regul Integr Comp Physiol ; 295(6): R1822-30, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18832081

ABSTRACT

Multiple isoforms of inhibitory Galpha-subunits (Galphai1,2,3, as well as Galphao) are present within the heart, and their role in modulating pacemaker function remains unresolved. Do inhibitory Galpha-subunits selectively modulate parasympathetic heart rate responses? Published findings using a variety of experimental approaches have implicated roles for Galphai2, Galphai3, and Galphao in parasympathetic signal transduction. We have compared in vivo different groups of mice with global genetic deletion of Gialpha1/Galphai3, Galphai2, and Galphao against littermate controls using implanted ECG telemetry. Significant resting tachycardia was observed in Galphai2(-/-) and Galphao(-/-) mice compared with control and Galphai1(-/-)/Galphai3(-/-) mice (P < 0.05). Loss of diurnal heart rate variation was seen exclusively in Galphao(-/-) mice. Using heart rate variability (HRV) analysis, compared with littermate controls (4.02 ms2 +/- 1.17; n = 6, Galphai2(-/-)) mice have a selective attenuation of high-frequency (HF) power (0.73 ms2 +/- 0.31; n = 5, P < 0.05). Galphai1(-/-)/Galphai3(-/-) and Galphao(-/-) cohorts have nonsignificant changes in HF power. Galphao(-/-) mice have a different basal HRV signature. The observed HRV phenotype in Galphai2(-/-) mice was qualitatively similar to atropine (1 mg/kg)-treated controls [and mice treated with the GIRK channel blocker tertiapinQ (0.05 mg/kg)]. Maximal cardioinhibitory response to the M(2)-receptor agonist carbachol (0.5 mg/kg) compared with basal heart rate was attenuated in Galphai2(-/-) mice (0.08 +/- 0.04; n = 6) compared to control (0.27 +/- 0.04; n = 7 P < 0.05). Our data suggest a selective defect of parasympathetic heart rate modulation in mice with Galphai2 deletion. Mice with Galphao deletion also have a defect in short-term heart rate dynamics, but this is qualitatively different to the effects of atropine, tertiapinQ, and Galphai2 deletion. In contrast, Galphai1 and Galphai3 do not appear to be essential for parasympathetic responses in vivo.


Subject(s)
Circadian Rhythm , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Heart Rate , Heart/innervation , Myocardium/metabolism , Parasympathetic Nervous System/physiology , Animals , Atropine/pharmacology , Bee Venoms/pharmacology , Carbachol/pharmacology , Electrocardiography, Ambulatory , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Genotype , Heart Rate/drug effects , Mice , Mice, Knockout , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Parasympathetic Nervous System/drug effects , Phenotype , Potassium Channel Blockers/pharmacology , Telemetry
17.
Pharmacol Res ; 58(3-4): 222-31, 2008.
Article in English | MEDLINE | ID: mdl-18812224

ABSTRACT

The aim of this study was to investigate the relevance of mouse ex vivo cultures as a first screening model for new therapeutic agents of Inflammatory Bowel Disease (IBD). Two murine models (dextran sodium sulphate (DSS)-induced colitis and Galphai2-deficient mice) and two anti-inflammatory agents (methyl-prednisolone and the proteasome inhibitor MG132) were evaluated. The in vivo effects of methyl-prednisolone were assessed in both models. Ex vivo colonic tissue from both mouse models were cultured in the presence or absence of the drugs and TaqMan Low-Density arrays were used to assess the regulation of inflammatory genes before and after drug treatment. Colitis induced a similar inflammatory gene profile in both mouse models in in vivo studies and in ex vivo cultures. The differences encountered reflected the different phases of colitis in the models, e.g. innate cytokine/chemokine profile in the DSS model and T cell related markers in Galphai2-deficient mice. After steroid treatment, a similar pattern of genes was suppressed in the two mouse models. We confirmed the suppression of inflammatory gene expression for IL-1beta, IL-6 and iNOS in ex vivo and in vivo colons from both mouse models by quantitative RT-PCR. Importantly, the inflammatory responses in the murine ex vivo culture system reflected the in vivo response in the inflamed colonic tissue as assessed by changes in inflammatory gene expression, suggesting that the murine culture system can be used for validation of future IBD therapies.


Subject(s)
Colitis/drug therapy , Inflammatory Bowel Diseases/drug therapy , Animals , Anti-Inflammatory Agents/therapeutic use , Cells, Cultured , Colitis/chemically induced , Colitis/genetics , DNA Primers , Dextran Sulfate , Female , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Leupeptins/pharmacology , Methylprednisolone/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/biosynthesis , Organ Culture Techniques , Reverse Transcriptase Polymerase Chain Reaction
18.
Invest Ophthalmol Vis Sci ; 49(7): 3245-52, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18378571

ABSTRACT

PURPOSE: Ocular albinism type 1 (OA1) is characterized by abnormalities in retinal pigment epithelium (RPE) melanosomes and misrouting of optic axons. The OA1 gene encodes a G-protein-coupled receptor (GPCR) that coimmunoprecipitates with the G alpha i-subunit of heterotrimeric G-proteins from human melanocyte extracts. This study was undertaken to test whether one of the G alpha i proteins, G alpha i3, signals in the same pathway as OA1 to regulate melanosome biogenesis and axonal growth through the optic chiasm. METHODS: Adult G alpha i3(-/-) and Oa1(-/-) mice were compared with their respective control mice (129Sv and B6/NCrl) to study the effects of the loss of G alpha i3 or Oa1 function. Light and electron microscopy were used to analyze the morphology of the retina and the size and density of RPE melanosomes, electroretinograms to study retinal function, and retrograde labeling to investigate the size of the uncrossed optic pathway. RESULTS: Although G alpha i3(-/-) and Oa1(-/-) photoreceptors were comparable to those of the corresponding control retinas, the density of their RPE melanosomes was significantly lower than in control RPEs. In addition, the RPE cells of G alpha i3(-/-) and Oa1(-/-) mice showed abnormal melanosomes that were far larger than the largest 129Sv and B6/NCrl melanosomes, respectively. Although G alpha i3(-/-) and Oa1(-/-) mice had normal results on electroretinography, retrograde labeling showed a significant reduction from control in the size of their ipsilateral retinofugal projections. CONCLUSIONS: These results indicate that G alpha i3, like Oa1, plays an important role in melanosome biogenesis. Furthermore, they suggest a common Oa1-G alpha i3 signaling pathway that ultimately affects axonal growth through the optic chiasm.


Subject(s)
Eye Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Melanosomes/physiology , Membrane Glycoproteins/metabolism , Optic Chiasm/physiology , Pigment Epithelium of Eye/physiology , Receptors, G-Protein-Coupled/metabolism , Animals , Axons/physiology , Carrier Proteins/metabolism , Cell Count , Electroretinography , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , Intracellular Membranes/metabolism , Melanosomes/ultrastructure , Membrane Glycoproteins/deficiency , Mice , Mice, Knockout , Microscopy, Electron , Phenotype , Pigment Epithelium of Eye/ultrastructure , Receptors, G-Protein-Coupled/deficiency , Retina/cytology , Retina/physiology , Retinal Ganglion Cells/cytology , Signal Transduction/physiology , Synaptic Transmission , Visual Pathways/cytology , Visual Pathways/growth & development , Visual Pathways/physiology
19.
J Biol Chem ; 282(13): 9547-9555, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17289675

ABSTRACT

G protein-coupled receptors (GPCRs) convey extracellular stimulation into dynamic intracellular action, leading to the regulation of cell migration and differentiation. T lymphocytes express G alpha(i2) and G alpha(i3), two members of the G alpha(i/o) protein family, but whether these two G alpha(i) proteins have distinguishable roles guiding T cell migration remains largely unknown because of a lack of member-specific inhibitors. This study details distinct G alpha(i2) and G alpha(i3) effects on chemokine receptor CXCR3-mediated signaling. Our data showed that G alpha(i2) was indispensable for T cell responses to three CXCR3 ligands, CXCL9, CXCL10, and CXCL11, as the lack of G alpha(i2) abolished CXCR3-stimulated migration and guanosine 5'-3-O-(thio)triphosphate (GTPgammaS) incorporation. In sharp contrast, T cells isolated from G alpha(i3) knock-out mice displayed a significant increase in both GTPgammaS incorporation and migration as compared with wild type T cells when stimulated with CXCR3 agonists. The increased GTPgammaS incorporation was blocked by G alpha(i3) protein in a dose-dependent manner. G alpha(i3)-mediated blockade of G alpha(i2) activation did not result from G alpha(i3) activation, but instead resulted from competition or steric hindrance of G alpha(i2) interaction with the CXCR3 receptor via the N terminus of the second intracellular loop. A mutation in this domain abrogated not only G alpha(i2) activation induced by a CXCR3 agonist but also the interaction of G alpha(i3) to the CXCR3 receptor. These findings reveal for the first time an interplay of G alpha(i) proteins in transmitting G protein-coupled receptor signals. This interplay has heretofore been masked by the use of pertussis toxin, a broad inhibitor of the G alpha(i/o) protein family.


Subject(s)
GTP-Binding Protein alpha Subunit, Gi2/antagonists & inhibitors , GTP-Binding Protein alpha Subunit, Gi2/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Receptors, Chemokine/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Chemokine CXCL10 , Chemokine CXCL11 , Chemokine CXCL9 , Chemokines, CXC/metabolism , Female , GTP-Binding Protein alpha Subunit, Gi2/deficiency , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3 , Receptors, Chemokine/metabolism , Signal Transduction/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
20.
J Clin Invest ; 115(11): 3045-56, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16276415

ABSTRACT

Ang II type 1 (AT1) receptors activate both conventional heterotrimeric G protein-dependent and unconventional G protein-independent mechanisms. We investigated how these different mechanisms activated by AT1 receptors affect growth and death of cardiac myocytes in vivo. Transgenic mice with cardiac-specific overexpression of WT AT1 receptor (AT1-WT; Tg-WT mice) or an AT1 receptor second intracellular loop mutant (AT1-i2m; Tg-i2m mice) selectively activating G(alpha)q/G(alpha)i-independent mechanisms were studied. Tg-i2m mice developed more severe cardiac hypertrophy and bradycardia coupled with lower cardiac function than Tg-WT mice. In contrast, Tg-WT mice exhibited more severe fibrosis and apoptosis than Tg-i2m mice. Chronic Ang II infusion induced greater cardiac hypertrophy in Tg-i2m compared with Tg-WT mice whereas acute Ang II administration caused an increase in heart rate in Tg-WT but not in Tg-i2m mice. Membrane translocation of PKCepsilon, cytoplasmic translocation of G(alpha)q, and nuclear localization of phospho-ERKs were observed only in Tg-WT mice while activation of Src and cytoplasmic accumulation of phospho-ERKs were greater in Tg-i2m mice, consistent with the notion that G(alpha)q/G(alpha)i-independent mechanisms are activated in Tg-i2m mice. Cultured myocytes expressing AT1-i2m exhibited a left and upward shift of the Ang II dose-response curve of hypertrophy compared with those expressing AT1-WT. Thus, the AT1 receptor mediates downstream signaling mechanisms through G(alpha)q/G(alpha)i-dependent and -independent mechanisms, which induce hypertrophy with a distinct phenotype.


Subject(s)
Bradycardia/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Hypertrophy, Left Ventricular/genetics , Mutation , Myocytes, Cardiac/metabolism , Receptor, Angiotensin, Type 1/deficiency , Receptor, Angiotensin, Type 1/genetics , Animals , Apoptosis/genetics , Bradycardia/metabolism , Bradycardia/pathology , Cells, Cultured , Electrocardiography , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis/genetics , Fibrosis/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/deficiency , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Mice , Mice, Transgenic , Myocytes, Cardiac/pathology , Phenotype , Protein Kinase C-epsilon/metabolism , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL