Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biol Direct ; 18(1): 78, 2023 11 20.
Article in English | MEDLINE | ID: mdl-37986113

ABSTRACT

BACKGROUND: Regulator of G protein signaling 5 (RGS5), as a negative regulator of G protein-coupled receptor (GPCR) signaling, is highly expressed in arterial VSMCs and pericytes, which is involved in VSMC phenotypic heterogeneity and vascular remodeling in tumors. However, its role in normal and tumor vascular remodeling is controversial. METHODS: RGS5 knockout (Rgs5-KO) mice and RGS5 overexpression or knockdown in VSMCs in vivo by adeno-associated virus type 9 (AAV) carrying RGS5 cDNA or small hairpin RNA (shRNA) targeting RGS5 were used to determine the functional significance of RGS5 in vascular inflammation. RGS5 expression in the triple-negative (TNBCs) and non-triple-negative breast cancers (Non-TNBCs) was determined by immunofluorescent and immunohistochemical staining. The effect of breast cancer cell-conditioned media (BC-CM) on the pro-inflammatory phenotype of VSMCs was measured by phagocytic activity assays, adhesion assay and Western blot. RESULTS: We identified that knockout and VSMC-specific knockdown of RGS5 exacerbated accumulation and pyroptosis of pro-inflammatory VSMCs, resulting in vascular remodeling, which was negated by VSMC-specific RGS5 overexpression. In contrast, in the context of breast cancer tissues, the role of RGS5 was completely disrupted. RGS5 expression was increased in the triple-negative breast cancer (TNBC) tissues and in the tumor blood vessels, accompanied with an extensive vascular network. VSMCs treated with BC-CM displayed enhanced pro-inflammatory phenotype and higher adherent with macrophages. Furthermore, tumor-derived RGS5 could be transferred into VSMCs. CONCLUSIONS: These findings suggest that tumor microenvironment shifts the function of RGS5 from anti-inflammation to pro-inflammation and induces the pro-inflammatory phenotype of VSMCs that is favorable for tumor metastasis.


Subject(s)
Neoplasms , RGS Proteins , Mice , Animals , RGS Proteins/genetics , RGS Proteins/metabolism , Vascular Remodeling/genetics , Muscle, Smooth, Vascular/metabolism , Tumor Microenvironment , Mice, Knockout , Homeostasis , Inflammation , Cell Proliferation
2.
Stem Cells ; 41(5): 493-504, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36888549

ABSTRACT

Regulator of G protein signaling 5 (RGS5) is a GTPase activator for heterotrimeric G-protein α-subunits, shown to be a marker of pericytes. Bone marrow stromal cell population (BMSCs) is heterogeneous. Populations of mesenchymal progenitors, cells supportive of hematopoiesis, and stromal cells regulating bone remodeling have been recently identified. Periosteal and bone marrow mesenchymal stem cells (MSCs) are participating in fracture healing, but it is difficult to distinguish the source of cells within the callus. Considering that perivascular cells exert osteoprogenitor potential, we generated an RGS5 transgenic mouse model (Rgs5-CreER) which when crossed with Ai9 reporter animals (Rgs5/Tomato), is suitable for lineage tracing during growth and post-injury. Flow cytometry analysis and histology confirmed the presence of Rgs5/Tomato+ cells within CD31+ endothelial, CD45+ hematopoietic, and CD31-CD45- mesenchymal/perivascular cells. A tamoxifen chase showed expansion of Rgs5/Tomato+ cells expressing osterix within the trabeculae positioned between mineralized matrix and vasculature. Long-term chase showed proportion of Rgs5/Tomato+ cells contributes to mature osteoblasts expressing osteocalcin. Following femoral fracture, Rgs5/Tomato+ cells are observed around newly formed bone within the BM cavity and expressed osterix and osteocalcin, while contribution within periosteum was low and limited to fibroblastic callus with very few positive chondrocytes. In addition, BM injury model confirmed that RGS5-Cre labels population of BMSCs expands during injury and participates in osteogenesis. Under homeostatic conditions, lineage-traced RGS5 cells within the trabecular area demonstrate osteoprogenitor capacity that in an injury model contributes to new bone formation primarily within the BM niche.


Subject(s)
Bony Callus , RGS Proteins , Mice , Animals , Osteocalcin/metabolism , Bony Callus/metabolism , Bony Callus/pathology , Osteogenesis , Fracture Healing/physiology , Chondrocytes/metabolism , Mice, Transgenic , Osteoblasts/metabolism , RGS Proteins/genetics , RGS Proteins/metabolism
3.
Int J Mol Sci ; 24(6)2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36982744

ABSTRACT

The brain needs sufficient oxygen in order to function normally. This is achieved by a large vascular capillary network ensuring that oxygen supply meets the changing demand of the brain tissue, especially in situations of hypoxia. Brain capillaries are formed by endothelial cells and perivascular pericytes, whereby pericytes in the brain have a particularly high 1:1 ratio to endothelial cells. Pericytes not only have a key location at the blood/brain interface, they also have multiple functions, for example, they maintain blood-brain barrier integrity, play an important role in angiogenesis and have large secretory abilities. This review is specifically focused on both the cellular and the molecular responses of brain pericytes to hypoxia. We discuss the immediate early molecular responses in pericytes, highlighting four transcription factors involved in regulating the majority of transcripts that change between hypoxic and normoxic pericytes and their potential functions. Whilst many hypoxic responses are controlled by hypoxia-inducible factors (HIF), we specifically focus on the role and functional implications of the regulator of G-protein signaling 5 (RGS5) in pericytes, a hypoxia-sensing protein that is regulated independently of HIF. Finally, we describe potential molecular targets of RGS5 in pericytes. These molecular events together contribute to the pericyte response to hypoxia, regulating survival, metabolism, inflammation and induction of angiogenesis.


Subject(s)
Endothelial Cells , Pericytes , Humans , Pericytes/metabolism , Endothelial Cells/metabolism , Brain/metabolism , Hypoxia/metabolism , Blood-Brain Barrier/metabolism , Oxygen/metabolism
4.
Int J Mol Sci ; 22(17)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34502263

ABSTRACT

The regulator of G protein signaling (RGS) represents a widespread system of controllers of cellular responses. The activities of the R4 subfamily of RGSs have been elucidated in allergic pulmonary diseases. However, the R4 signaling in other inflammatory lung diseases, with a strong cellular immune response, remained unexplored. Thus, our study aimed to discern the functional relevance of the R4 family member, RGS5, as a potential modulating element in this context. Gene profiling of the R4 subfamily showed increased RGS5 expression in human fibrosing lung disease samples. In line with this, RGS5 was markedly increased in murine lungs following bleomycin injury. RGS knock-out mice (RGS-/-) had preserved lung function while control mice showed significant combined ventilatory disorders three days after bleomycin application as compared to untreated control mice. Loss of RGS5 was associated with a significantly reduced neutrophil influx and tissue myeloperoxidase expression. In the LPS lung injury model, RGS5-/- mice also failed to recruit neutrophils into the lung, which was accompanied by reduced tissue myeloperoxidase levels after 24 h. Our in-vitro assays showed impaired migration of RGS5-/- neutrophils towards chemokines despite preserved Ca2+ signaling. ERK dephosphorylation might play a role in reduced neutrophil migration in our model. As a conclusion, loss of RGS5 preserves lung function and attenuates hyperinflammation in the acute phase of bleomycin-induced pulmonary fibrosis and LPS-induced lung injury. Targeting RGS5 might alleviate the severity of exacerbations in interstitial lung diseases.


Subject(s)
Inflammation/metabolism , Lung Injury/metabolism , Neutrophils/metabolism , RGS Proteins/genetics , RGS Proteins/metabolism , Animals , Bleomycin/toxicity , Chemotaxis/genetics , Disease Models, Animal , Fibrosis/genetics , Humans , Inflammation/chemically induced , Lipopolysaccharides/toxicity , Lung Diseases, Interstitial/genetics , Lung Diseases, Interstitial/metabolism , Lung Diseases, Interstitial/pathology , Lung Injury/chemically induced , Lung Injury/pathology , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Neutrophils/cytology , RGS Proteins/deficiency , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/metabolism
5.
Front Physiol ; 12: 622084, 2021.
Article in English | MEDLINE | ID: mdl-33815137

ABSTRACT

AIMS: This study investigated the contribution of the regulator of G-protein signaling 5 (Rgs5) knockout to the alteration of the action potential duration (APD) restitution and repolarizing dispersion in ventricle. METHODS AND RESULTS: The effects of Rgs5-/- were investigated by QT variance (QTv) and heart rate variability analysis of Rgs5-/- mice. Monophasic action potential analysis was investigated in isolated Rgs5-/- heart. Rgs5-/- did not promote ventricular remodeling. The 24-h QTv and QT variability index (QTVI) of the Rgs5-/- mice were higher than those of wild-type (WT) mice (P < 0.01). In WT mice, a positive correlation was found between QTv and the standard deviation of all NN intervals (r = 0.62; P < 0.01), but not in Rgs5-/- mice (R = 0.01; P > 0.05). The absence of Rgs5 resulted in a significant prolongation of effective refractory period and APD in isolated ventricle. In addition, compared with WT mice, the knockout of Rgs5 significantly deepened the slope of the APD recovery curve at all 10 sites of the heart (P < 0.01) and increased the spatial dispersions of Smax (COV-Smax) (WT: 0.28 ± 0.03, Rgs5-/-: 0.53 ± 0.08, P < 0.01). Compared with WT heart, Rgs5-/- increased the induced S1-S2 interval at all sites of heart and widened the window of vulnerability of ventricular tachyarrhythmia (P < 0.05). CONCLUSION: Our findings indicate that Rgs5-/- is an important regulator of ventricular tachyarrhythmia in mice by prolonging ventricular repolarization and increasing spatial dispersion in ventricle.

6.
Exp Neurol ; 305: 139-150, 2018 07.
Article in English | MEDLINE | ID: mdl-29630897

ABSTRACT

Microvascular changes have recently been described for several neurodegenerative disorders, including Huntington's disease (HD). HD is characterized by a progressive neuronal cell loss due to a mutation in the Huntingtin gene. However, the temporal and spatial microvascular alterations in HD remain unclear. Also, knowledge on the implication of pericytes in HD pathology is still sparse and existing findings are contradictory. Here we examine alterations in brain pericytes in the R6/2 mouse model of HD and in human post mortem HD brain sections. To specifically track activated pericytes, we crossbred R6/2 mice with transgenic mice expressing the Green fluorescent protein gene under the Regulator of G-protein signaling 5 (Rgs5) promoter. We demonstrate an increase in activated pericytes in the R6/2 brain and in post mortem HD brain tissue. Importantly, pericyte changes are already detected before striatal neuronal cell loss, weight loss or behavioural deficits occur in R6/2 mice. This is associated with vascular alterations, whereby striatal changes precede cortical changes. Our findings suggest that pericyte activation may be one of the initial steps contributing to the observed vascular modifications in HD. Thus, pericytes may constitute an important target to address early microvascular changes contributing to disease progression in HD.


Subject(s)
Brain/metabolism , Brain/pathology , Huntington Disease/metabolism , Huntington Disease/pathology , Pericytes/metabolism , Pericytes/pathology , Adult , Aged , Animals , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged
7.
Oncol Lett ; 15(2): 1763-1770, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29434872

ABSTRACT

Portal vein invasion (PVI) is a major prognostic factor in hepatocellular carcinoma (HCC). The aim of the present study was to identify molecules that regulate PVI. Sections of cancerous tissue, paired noncancerous tissue and the PVI area were collected from 3 frozen HCC sections, using laser microdissection. The present study focused on 3 upregulated molecules, integrin ß3 (ITGB3), secreted phosphoprotein 1 (SPP1) and regulator of G-protein signaling 5 (RGS5), and 2 molecules that were downregulated in PVI tissue compared with cancer tissue, metallothionein 1G (MT1G) and metallothionein 1H (MT1H), as determined by cDNA microarray analysis. Reverse transcription-quantitative polymerase chain reaction analysis of 32 HCC cases revealed that RGS5 mRNA levels were significantly increased and MT1 G and MT1H mRNA levels were significantly decreased in cancerous tissue compared with noncancerous tissue. However, there was no significant difference in ITGB3 and SPP1 expression. There were no significant differences between the expression of these molecules and any clinicopathologic factors, including PVI. Immunohistochemical staining for RGS5 in 60 HCC cases demonstrated that RGS5 protein levels were higher in cancerous tissue compared with paired noncancerous tissue in 63.3% of HCC cases. Furthermore, high expression of RGS5 in cancerous tissue was significantly associated with PVI and tended to be associated with intrahepatic metastasis. Confluent multinodular type was significantly more frequent in cases with high expression of RGS5 in the cancerous tissue. Therefore, RGS5 may be a useful prognostic biomarker as well as a potential target of molecular therapy to treat HCC.

8.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-515217

ABSTRACT

Objective To observe the effect of endostatin(Endostar) combined with radiotheraphy on the growth,microvessel density (MVD),the expressions of regulators of G-protein signaling 5 (RGSS),vascular endothelial growth factor (VEGF) and hypoxia-induced factor-1 α (HIF-1 α) ingastric transplantation tumor,and to explore underlying mechanism.Methods Thirty-two tumor-bearing mice were randomly divided into four groups including control group,ES group,RT group and ES + RT group.Then the tumor-bearing mice survival and tumor volume alterations were observed.After treatments of drug and RT,the inhibition rate of tumor growth and tumor MVD were measured.Immunohistochemical staining method was used to detect the expressions of RGS5,VEGF and HIF-1α.Results Compared with the ES group and control group,the ES + RT and RT treatment had effective anti-tumor effect(t=7.4,5.6,P < 0.05),where as the ES + RT was more obvious in anti-tumor with the lowest value of MVD in tumor.Compared with the control group,the expression of HIF-1α in ES group,RT group,and especially ES + RT group was significantly reduced (t =6.5,8.2,13.1,P < 0.05).The expression levels of VEGF and RGS5 wereincreased in RT group but was reduced in ES group and ES + RT group,and the expressions of VEGF and RGS5 in ES group and ES + RT group had no significant difference (P > 0.05).Rank correlation analysis showed the expression of HIF-1α and the expressions of VEGF and RGS5 were positively correlated (r =0.57,0.71,P < 0.05).Conclusions A possible mechanism of Endostar combined with radiotherapy on tumor growth inhibition may due to the inhibition of HIF-1α and its downstream VEGF and RGS5,and then inhibit tumor angiogenesis that results in the recovery of tumor blood vessels and tumor oxygen supplement.

9.
J Am Heart Assoc ; 5(3): e002783, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26961238

ABSTRACT

BACKGROUND: The regulator of G-protein signaling 5 (Rgs5), which functions as the regulator of G-protein-coupled receptor (GPCR) including muscarinic receptors, has a potential effect on atrial muscarinic receptor-activated IKA ch current. METHODS AND RESULTS: In the present study, hearts of Rgs5 knockout (KO) mice had decreased low-frequency/high-frequency ratio in spectral measures of heart rate variability. Loss of Rgs5 provoked dramatically exaggerated bradycardia and significantly (P<0.05) prolonged sinus nodal recovery time in response to carbachol (0.1 mg/kg, intraperitoneally). Compared to those from wild-type (WT) mice, Langendorff perfused hearts from Rgs5 KO mice had significantly (P<0.01) abbreviated atrial effective refractory periods and increased dominant frequency after administration of acetylcholine (ACh; 1 µmol/L). In addition, whole patch clamp analyses of single atrial myocytes revealed that the ACh-regulated potassium current (IKA ch) was significant increased in the time course of activation and deactivation (P<0.01) in Rgs5 KO, compared to those in WT, mice. To further determine the effect of Rgs5, transgenic mice with cardiac-specific overexpression of human Rgs5 were found to be resistant to ACh-related effects in bradycardia, atrial electrophysiology, and atrial tachyarrhythmia (AT). CONCLUSION: The results of this study indicate that, as a critical regulator of parasympathetic activation in the heart, Rgs5 prevents vagal-related bradycardia and AT through negatively regulating the IKA ch current.


Subject(s)
Bradycardia/prevention & control , Heart Rate , Heart/innervation , Myocytes, Cardiac/metabolism , RGS Proteins/metabolism , Tachycardia, Supraventricular/prevention & control , Vagus Nerve/physiopathology , Acetylcholine/pharmacology , Action Potentials , Animals , Bradycardia/genetics , Bradycardia/metabolism , Bradycardia/physiopathology , Carbachol , Cholinergic Agonists/pharmacology , Disease Models, Animal , Humans , Isolated Heart Preparation , Male , Mice, Knockout , Myocytes, Cardiac/drug effects , Potassium/metabolism , RGS Proteins/deficiency , RGS Proteins/genetics , Receptors, Muscarinic/metabolism , Refractory Period, Electrophysiological , Tachycardia, Supraventricular/genetics , Tachycardia, Supraventricular/metabolism , Tachycardia, Supraventricular/physiopathology , Time Factors
10.
Prog Mol Biol Transl Sci ; 133: 93-121, 2015.
Article in English | MEDLINE | ID: mdl-26123304

ABSTRACT

The cardiovascular system including the heart and intricate network of arteries, veins, and capillaries is essential for normal organ homeostasis and tightly controlled by G protein-coupled receptor (GPCR) signaling cascades. Imbalances of these signaling systems can manifest in cardiovascular disease. There has been a recent surge in studies on modulators of GPCR activity, so-called regulator of G protein signaling (RGS) molecules, due to their potential as pharmacological targets. Among RGS proteins, RGS5 is prominently expressed in arterial vascular smooth muscle cells (vSMC) suggesting an important role in vascular function. Although apparently dispensable for embryonic development, RGS5 has now emerged as a crucial regulator of adaptive cardiovascular processes, including remodeling of the vascular wall under stress. RGS5 has been shown to regulate signaling pathways which shape vSMC differentiation, migration, contraction, as well as tissue inflammation and fibrosis. Indeed, studies in RGS5 mutant mice have confirmed a crucial and nonredundant role as regulator of cardiac function, blood pressure homeostasis, and adult neovascularization such as angiogenesis and arteriogenesis. In response to environmental cues, RGS5 is dynamically controlled at both mRNA and protein levels. This enables direct, precise, and rapid modulation of Gαq- and Gαi-coupled GPCR signaling which also integrates receptor tyrosine kinases (RTK) and Gαs/Gα12/13-mediated GPCR signal transduction. Although RGS5's endogenous role in a spatiotemporal context is still largely unknown, its prominence in vascular tissue makes it an important molecule to study and a prime candidate for therapeutic intervention.


Subject(s)
Blood Vessels/physiology , RGS Proteins/metabolism , Signal Transduction , Animals , Blood Vessels/metabolism , Cardiovascular System/metabolism , Cardiovascular System/pathology , Humans , Myocytes, Smooth Muscle/metabolism , Phenotype
11.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-440549

ABSTRACT

Regulator of G-protein signaling 5 (RGS5) belongs to RGS family,which can negatively regulate the conduction of this signaling pathway.RGS5 mainly expresses in vascular pericyte,and is closely related to the occurrence,development and maturation of the blood vessels.Loss of RGS5 results in pericyte maturation,tumor vascular normalization,and these changes can improve the curative effect combined with chemotherapy and immunotherapy,indicating that RGS5 may become a new target of anti-tumor treatment.In addition,RGS5 involves in tumor metastasis and apoptosis,which can improve antineoplastic effect by inducing tumor cells apoptosis.

SELECTION OF CITATIONS
SEARCH DETAIL
...