Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Country/Region as subject
Publication year range
1.
EMBO J ; 41(13): e109996, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35767364

ABSTRACT

Helicobacter pylori is a pathogen that colonizes the stomach and causes chronic gastritis. Helicobacter pylori can colonize deep inside gastric glands, triggering increased R-spondin 3 (Rspo3) signaling. This causes an expansion of the "gland base module," which consists of self-renewing stem cells and antimicrobial secretory cells and results in gland hyperplasia. The contribution of Rspo3 receptors Lgr4 and Lgr5 is not well explored. Here, we identified that Lgr4 regulates Lgr5 expression and is required for H. pylori-induced hyperplasia and inflammation, while Lgr5 alone is not. Using conditional knockout mice, we reveal that R-spondin signaling via Lgr4 drives proliferation of stem cells and also induces NF-κB activity in the proliferative stem cells. Upon exposure to H. pylori, the Lgr4-driven NF-κB activation is responsible for the expansion of the gland base module and simultaneously enables chemokine expression in stem cells, resulting in gland hyperplasia and neutrophil recruitment. This demonstrates a connection between R-spondin-Lgr and NF-κB signaling that links epithelial stem cell behavior and inflammatory responses to gland-invading H. pylori.


Subject(s)
Helicobacter pylori , Animals , Hyperplasia/metabolism , Hyperplasia/pathology , Inflammation/pathology , Mice , NF-kappa B/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Stomach
2.
Am J Physiol Gastrointest Liver Physiol ; 326(4): G460-G472, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38440827

ABSTRACT

Current therapy for hepatic injury induced by the accumulation of bile acids is limited. Leucine-rich repeat G protein-coupled receptor 4 (LGR4), also known as GPR48, is critical for cytoprotection and cell proliferation. Here, we reported a novel function for the LGR4 in cholestatic liver injury. In the bile duct ligation (BDL)-induced liver injury model, hepatic LGR4 expression was significantly downregulated. Deficiency of LGR4 in hepatocytes (Lgr4LKO) notably decreased BDL-induced liver injury measured by hepatic necrosis, fibrosis, and circulating liver enzymes and total bilirubin. Levels of total bile acids in plasma and liver were markedly reduced in these mice. However, deficiency of LGR4 in macrophages (Lyz2-Lgr4MKO) demonstrated no significant effect on liver injury induced by BDL. Deficiency of LGR4 in hepatocytes significantly attenuated S1PR2 and the phosphorylation of protein kinase B (AKT) induced by BDL. Recombinant Rspo1 and Rspo3 potentiated the taurocholic acid (TCA)-induced upregulation in S1PR2 and phosphorylation of AKT in hepatocytes. Inhibition of S1PR2-AKT signaling by specific AKT or S1PR2 inhibitors blocked the increase of bile acid secretion induced by Rspo1/3 in hepatocytes. Our studies indicate that the R-spondins (Rspos)-LGR4 signaling in hepatocytes aggravates the cholestatic liver injury by potentiating the production of bile acids in a S1PR2-AKT-dependent manner.NEW & NOTEWORTHY Deficiency of LGR4 in hepatocytes alleviates BDL-induced liver injury. LGR4 in macrophages demonstrates no effect on BDL-induced liver injury. Rspos-LGR4 increases bile acid synthesis and transport via potentiating S1PR2-AKT signaling in hepatocytes.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Cholestasis , Mice , Animals , Proto-Oncogene Proteins c-akt/metabolism , Liver/metabolism , Cholestasis/complications , Cholestasis/metabolism , Hepatocytes/metabolism , Bile Acids and Salts/metabolism , Bile Ducts/metabolism , Ligation , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
3.
Circ Res ; 131(12): 1037-1054, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36354004

ABSTRACT

BACKGROUND: Vascular calcification is closely related to the all-cause mortality of cardiovascular events. Basement membrane protein nidogen-2 is a key component of the vascular extracellular matrix microenvironment and we recently found it is pivotal for the maintenance of contractile phenotype in vascular smooth muscle cells (VSMCs). However, whether nidogen-2 is involved in VSMCs osteochondrogenic transition and vascular calcification remains unclear. METHODS: VSMCs was treated with high-phosphate to study VSMC calcification in vitro. Three different mice models (5/6 nephrectomy-induced chronic renal failure, cholecalciferol-overload, and periadventitially administered with CaCl2) were used to study vascular calcification in vivo. Membrane protein interactome, coimmunoprecipitation, flow cytometric binding assay, surface plasmon resonance, G protein signaling, VSMCs calcium assays were performed to clarify the phenotype and elucidate the molecular mechanisms. RESULTS: Nidogen-2 protein levels were significantly reduced in calcified VSMCs and aortas from mice in different vascular calcification model. Nidogen-2 deficiency exacerbated high-phosphate-induced VSMC calcification, whereas the addition of purified nidogen-2 protein markedly alleviated VSMC calcification in vitro. Nidogen-2-/- mice exhibited aggravated aorta calcification compared to wild-type (WT) mice in response to 5/6 nephrectomy, cholecalciferol-overload, and CaCl2 administration. Further unbiased coimmunoprecipitation and interactome analysis of purified nidogen-2 and membrane protein in VSMCs revealed that nidogen-2 directly binds to LGR4 (leucine-rich repeat G-protein-coupled receptor 4) with KD value 26.77 nM. LGR4 deficiency in VSMCs in vitro or in vivo abolished the protective effect of nidogen-2 on vascular calcification. Of interest, nidogen-2 biased activated LGR4-Gαq-PKCα (protein kinase Cα)-AMPKα1 (AMP-activated protein kinase α1) signaling to counteract VSMCs osteogenic transition and mineralization. CONCLUSIONS: Nidogen-2 is a novel endogenous ligand of LGR4 that biased activated Gαq- PKCα-AMPKα1 signaling and inhibited vascular calcification.


Subject(s)
Membrane Glycoproteins , Muscle, Smooth, Vascular , Receptors, G-Protein-Coupled , Vascular Calcification , Animals , Mice , Calcium Chloride , Cells, Cultured , Cholecalciferol/pharmacology , Cholecalciferol/metabolism , Ligands , Membrane Glycoproteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phosphates/adverse effects , Protein Kinase C-alpha/metabolism , Receptors, G-Protein-Coupled/metabolism , Vascular Calcification/prevention & control , Vascular Calcification/genetics
4.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33443181

ABSTRACT

Taste bud cells regenerate throughout life. Taste bud maintenance depends on continuous replacement of senescent taste cells with new ones generated by adult taste stem cells. More than a century ago it was shown that taste buds degenerate after their innervating nerves are transected and that they are not restored until after reinnervation by distant gustatory ganglion neurons. Thus, neuronal input, likely via neuron-supplied factors, is required for generation of differentiated taste cells and taste bud maintenance. However, the identity of such a neuron-supplied niche factor(s) remains unclear. Here, by mining a published RNA-sequencing dataset of geniculate ganglion neurons and by in situ hybridization, we demonstrate that R-spondin-2, the ligand of Lgr5 and its homologs Lgr4/6 and stem-cell-expressed E3 ligases Rnf43/Znrf3, is expressed in nodose-petrosal and geniculate ganglion neurons. Using the glossopharyngeal nerve transection model, we show that systemic delivery of R-spondin via adenovirus can promote generation of differentiated taste cells despite denervation. Thus, exogenous R-spondin can substitute for neuronal input for taste bud cell replenishment and taste bud maintenance. Using taste organoid cultures, we show that R-spondin is required for generation of differentiated taste cells and that, in the absence of R-spondin in culture medium, taste bud cells are not generated ex vivo. Thus, we propose that R-spondin-2 may be the long-sought neuronal factor that acts on taste stem cells for maintaining taste tissue homeostasis.


Subject(s)
Regeneration , Taste Buds/physiology , Thrombospondins/metabolism , Animals , Cell Differentiation , Mice , Organoids , Taste Buds/cytology
5.
Int J Mol Sci ; 25(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38891922

ABSTRACT

Vascular calcification has a global health impact that is closely linked to bone loss. The Receptor Activator of Nuclear Factor Kappa B (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) system, fundamental for bone metabolism, also plays an important role in vascular calcification. The Leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4), a novel receptor for RANKL, regulates bone remodeling, and it appears to be involved in vascular calcification. Besides RANKL, LGR4 interacts with R-spondins (RSPOs), which are known for their roles in bone but are less understood in vascular calcification. Studies were conducted in rats with chronic renal failure fed normal or high phosphorus diets for 18 weeks, with and without control of circulating parathormone (PTH) levels, resulting in different degrees of aortic calcification. Additionally, vascular smooth muscle cells (VSMCs) were cultured under non-calcifying (1 mM phosphate) and calcifying (3 mM phosphate) media with different concentrations of PTH. To explore the role of RANKL in VSMC calcification, increasing concentrations of soluble RANKL were added to non-calcifying and calcifying media. The effects mediated by RANKL binding to its receptor LGR4 were investigated by silencing the LGR4 receptor in VSMCs. Furthermore, the gene expression of the RANK/RANKL/OPG system and the ligands of LGR4 was assessed in human epigastric arteries obtained from kidney transplant recipients with calcification scores (Kauppila Index). Increased aortic calcium in rats coincided with elevated systolic blood pressure, upregulated Lgr4 and Rankl gene expression, downregulated Opg gene expression, and higher serum RANKL/OPG ratio without changes in Rspos gene expression. Elevated phosphate in vitro increased calcium content and expression of Rankl and Lgr4 while reducing Opg. Elevated PTH in the presence of high phosphate exacerbated the increase in calcium content. No changes in Rspos were observed under the conditions employed. The addition of soluble RANKL to VSMCs induced genotypic differentiation and calcification, partly prevented by LGR4 silencing. In the epigastric arteries of individuals presenting vascular calcification, the gene expression of RANKL was higher. While RSPOs show minimal impact on VSMC calcification, RANKL, interacting with LGR4, drives osteogenic differentiation in VSMCs, unveiling a novel mechanism beyond RANKL-RANK binding.


Subject(s)
Muscle, Smooth, Vascular , RANK Ligand , Receptors, G-Protein-Coupled , Vascular Calcification , RANK Ligand/metabolism , RANK Ligand/genetics , Animals , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Vascular Calcification/metabolism , Vascular Calcification/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Rats , Humans , Male , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Osteoprotegerin/metabolism , Osteoprotegerin/genetics , Parathyroid Hormone/metabolism , Cells, Cultured , Rats, Sprague-Dawley
6.
Connect Tissue Res ; 64(1): 40-52, 2023 01.
Article in English | MEDLINE | ID: mdl-35726900

ABSTRACT

INTRODUCTION: RANKL plays an important role in the differentiation and maturation process of preosteoclast cells. The osteoclast is a multinucleated cell that can have various sizes and a variable number of nuclei. However, there are no models that allow us to understand how successive cell fusions have a limit, or how cell fusion is regulated. METHODOLOGY: The present investigation was aimed to determine whether fusing U937 cells with PEG to generate osteoclast-like cells expresses LGR4 and whether applying RANKL to these cells modifies osteoclastic activity compared to non-PEG-fused and RANKL-treated cells. RESULTS: By fusing U937 cells with PEG, it was found that the LGR4 receptor expression was promoted as early as 24 hours of culture. Applying RANKL before or after fusion inhibits osteoclastic activity. Interfering RANKL interaction with LGR4 in PEG-treated cells recovers and increases cell fusion and osteoclastic activity. PEG-fused U937 cells show osteoclast markers similar to those observed in the classical RANKL-stimulated cell model. CONCLUSION: Our model allows us to understand that RANKL has fusogenic activity during the first days of culture and in fused cells modulates fusion, contributing to differentiate the role of RANKL before and after fusion through LGR4.


Subject(s)
Bone Resorption , Osteogenesis , Humans , Bone Resorption/metabolism , U937 Cells , Osteoclasts/metabolism , Cell Differentiation , RANK Ligand , Receptors, G-Protein-Coupled/metabolism
7.
EMBO Rep ; 22(5): e51415, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33786993

ABSTRACT

The tumour suppressors RNF43 and ZNRF3 play a central role in development and tissue homeostasis by promoting the turnover of the Wnt receptors LRP6 and Frizzled (FZD). The stem cell growth factor R-spondin induces auto-ubiquitination and membrane clearance of ZNRF3/RNF43 to promote Wnt signalling. However, the deubiquitinase stabilising ZNRF3/RNF43 at the plasma membrane remains unknown. Here, we show that the USP42 antagonises R-spondin by protecting ZNRF3/RNF43 from ubiquitin-dependent clearance. USP42 binds to the Dishevelled interacting region (DIR) of ZNRF3 and stalls the R-spondin-LGR4-ZNRF3 ternary complex by deubiquitinating ZNRF3. Accordingly, USP42 increases the turnover of LRP6 and Frizzled (FZD) receptors and inhibits Wnt signalling. Furthermore, we show that USP42 functions as a roadblock for paracrine Wnt signalling in colon cancer cells and mouse small intestinal organoids. We provide new mechanistic insights into the regulation R-spondin and conclude that USP42 is crucial for ZNRF3/RNF43 stabilisation at the cell surface.


Subject(s)
Thrombospondins , Ubiquitin-Protein Ligases , Animals , Mice , Receptors, G-Protein-Coupled/genetics , Thrombospondins/genetics , Thrombospondins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Wnt Signaling Pathway
8.
Exp Cell Res ; 405(2): 112713, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34181940

ABSTRACT

Obesity, a global epidemic, is one of the critical causes of chronic kidney disease (CKD). R-spondin1 (RSPO1) possessing the potential to activate Wnt/ß-catenin pathway was reported to be elevated in circulation of obesity objects. However, the function of RSPO1 and the latent mechanism in obesity-related CKD are still left to be revealed. In the present study, renal RSPO1 expression was increased in mice fed on high-fat diet (HFD) for 12 weeks. Lentivirus-mediated RSPO1 knockdown partly recovered obesity-related metabolic symptoms, while distinctly remitted kidney dysfunction and renal fibrosis in obesity mice. In vitro, recombinant RSPO1 was found to elevate leucine-rich repeat-containing G protein coupled receptor 4 (LGR4) expression, promote Wnt/ß-catenin signaling pathway activation, facilitate epithelial-mesenchymal transition (EMT) and increase collagen deposition in HK2 renal tubular cells. Such pro-fibrotic effect of RSPO1 was diminished by LGR4 siRNA in HK2 cells. In summary, we demonstrate that RSPO1/LGR4 axis is involved in obesity-related renal fibrosis at least through activating Wnt/ß-catenin signaling pathway, providing a potential therapeutic target for this disease.


Subject(s)
Fibrosis/metabolism , Obesity/complications , Thrombospondins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/physiology , Male , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/metabolism
9.
J Cell Physiol ; 236(6): 4273-4289, 2021 06.
Article in English | MEDLINE | ID: mdl-33452710

ABSTRACT

While mesenchymal stem cells (MSCs) have been widely used to repair radiation-induced bone damage, the molecular mechanism underlying the effects of MSCs in the maintenance of bone homeostasis under radiation stress remains largely unknown. In this study, the role and mechanisms of R-spondin 1 (Rspo1)-leucine-rich repeat-containing G protein-coupled receptor 4 (LGR4) axis on the initiation of self-defense of bone mesenchymal stem cells (BMSCs) and maintenance of bone homeostasis under radiation stress were investigated. Interestingly, radiation increased levels of Rspo1 and LGR4 in BMSCs. siRNA knockdown of Rspo1 or LGR4 aggravated radiation-induced impairment of self-renewal ability and osteogenic differentiation potential of BMSCs. However, exogenous Rspo1 significantly attenuated radiation-induced depletion of BMSCs, and promoted the lineage shift towards osteoblasts. This alteration was associated with the reversal of mammalian target of rapamycin (mTOR) activation and autophagy decrement. Pharmacological and genetic blockade of autophagy attenuated the radio-protective effects of Rspo1, rendering BMSCs more vulnerable to radiation-induced injury. Then bone radiation injury was induced in C57BL6J mice to further determine the radio-protective effects of Rspo1. In mice, administration of Rspo1 recombinant protein alleviated radiation-induced bone loss. Our results uncover that Rspo1-LGR4-mTOR-autophagy axis are key mechanisms by which BMSCs initiate self-defense against radiation and maintain bone homeostasis. Targeting Rspo1-LGR4 may provide a novel strategy for the intervention of radiation-induced bone damage.


Subject(s)
Autophagy , Bone Diseases/prevention & control , Mesenchymal Stem Cells/enzymology , Radiation Injuries/prevention & control , Receptors, G-Protein-Coupled/metabolism , TOR Serine-Threonine Kinases/metabolism , Thrombospondins/metabolism , Animals , Autophagy/radiation effects , Bone Diseases/enzymology , Bone Diseases/genetics , Bone Diseases/pathology , Cell Differentiation , Cell Proliferation , Cell Self Renewal , Cells, Cultured , DNA Damage , Disease Models, Animal , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/radiation effects , Mice, Inbred C57BL , Osteogenesis , Radiation Injuries/enzymology , Radiation Injuries/genetics , Radiation Injuries/pathology , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Thrombospondins/genetics
10.
Calcif Tissue Int ; 108(4): 439-451, 2021 04.
Article in English | MEDLINE | ID: mdl-33586001

ABSTRACT

In the course of chronic kidney disease (CKD), alterations in the bone-vascular axis augment the risk of bone loss, fractures, vascular and soft tissue calcification, left ventricular hypertrophy, renal and myocardial fibrosis, which markedly increase morbidity and mortality rates. A major challenge to improve skeletal and cardiovascular outcomes in CKD patients requires a better understanding of the increasing complex interactions among the main modulators of the bone-vascular axis. Serum parathyroid hormone (PTH), phosphorus (P), calcium (Ca), fibroblast growth factor 23 (FGF23), calcidiol, calcitriol and Klotho are involved in this axis interact with RANK/RANKL/OPG system and the Wnt/ß-catenin pathway. The RANK/RANKL/OPG system controls bone remodeling by inducing osteoblast synthesis of RANKL and downregulating OPG production and it is also implicated in vascular calcification. The complexity of this system has recently increased due the discovery of LGR4, a novel RANKL receptor involved in bone formation, but possibly also in vascular calcification. The Wnt/ß-catenin pathway plays a key role in bone formation: when this pathway is activated, bone is formed, but when it is inhibited, bone formation is stopped. In the progression of CKD, a downregulation of the Wnt/ß-catenin pathway has been described which occurs mainly through the not coincident elevations of sclerostin, Dickkopf1 (Dkk1) and the secreted Frizzled Related Proteins (sFRPs). This review analyzes the interactions of PTH, P, Ca, FGF23, calcidiol, calcitriol and Klotho with the RANKL/RANKL/OPG system and the Wnt/ß-catenin, pathway and their implications in bone and cardiovascular disorders in CKD.


Subject(s)
Catenins , Renal Insufficiency, Chronic , Bone Remodeling , Bone and Bones , Fibroblast Growth Factor-23 , Humans , Osteoprotegerin , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B
11.
FASEB J ; 34(11): 14946-14959, 2020 11.
Article in English | MEDLINE | ID: mdl-32926477

ABSTRACT

R-spondins (Rspos) are endogenous ligands of leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4). Rspos-LGR4 signaling plays important roles in embryogenesis, gastrointestinal homeostasis, and food intake. Here, we investigated the impacts of Rspos-LGR4 on hepatic cholesterol synthesis. Rspo1/3 and Lgr4 knockdown mice were used to investigate the impacts of Rspo1/3-LGR4 on hepatic cholesterol synthesis. AMPKα agonist, antagonist, and shRNA were used to explore the downstream targets of Rspos-LGR4 signaling. In our study, we reported that LGR4, Rspo1, and Rspo3 were highly expressed in hepatocytes and their expressions were sensitive to energy states. Rspo1 and Rspo3 reversed OA-induced cholesterol synthesis, accompanying with increased the phosphorylation of AMPKα Thr172, reduced SREBP2 nuclear translocation, and Srebf2 mRNA expression. Conversely, hepatic LGR4 knockdown increased hepatic cholesterol synthesis and decreased the phosphorylation of AMPKα both in vitro and in vivo. Activation or inhibition of AMPKα significantly abolished the effects of LGR4 deficiency or Rspos, respectively, on cholesterol synthesis. Knockdown of AMPKα1 or/and AMPKα2 repressed Rspos-induced inhibition on cholesterol synthesis. Our study indicates that Rspo1/Rspo3-LGR4 signaling in hepatocytes suppresses cholesterol synthesis via the AMPKα-SREBP2 pathway.


Subject(s)
Cholesterol/biosynthesis , Hepatocytes/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Thrombospondins/metabolism , AMP-Activated Protein Kinase Kinases , Active Transport, Cell Nucleus , Animals , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , Energy Metabolism , Hepatocytes/drug effects , Mice , Mice, Inbred C57BL , Oleic Acid/pharmacology , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Receptors, G-Protein-Coupled/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Thrombospondins/genetics
12.
FASEB J ; 34(11): 15605-15620, 2020 11.
Article in English | MEDLINE | ID: mdl-33001511

ABSTRACT

Mammalian LGR5 and LGR4, markers of adult stem cells, are involved in many physiological functions by enhancing WNT signaling. However, whether LGR5 and LGR4 are coupled to other intracellular signaling pathways to regulate stem cell function remains unknown. Here, we show that LGR5 and LGR4 can constitutively activate NF-κB signaling in a ligand-independent manner, which is dependent on their C-termini, but independent of receptor endocytosis. Moreover, the C-termini of LGR5/4 interact with TROY, which is required for activating NF-κB signaling. In small intestinal crypt organoids, overexpression of a C-terminal deletion mutant of LGR5 inhibits the growth and bud formation of organoids, whereas overexpression of the R-spondin-binding mutant of LGR5 that is defective for WNT signaling can still promote organoid growth. Our study reveals that NF-κB signaling, regulated by LGR5 and LGR4, plays an important role in the survival of colon cancer cells and the growth of intestinal crypts. Our findings also suggest that LGR5/4-induced NF-κB signaling and WNT signaling may co-regulate the growth of LGR5+ adult stem cells and intestinal crypts.


Subject(s)
Colonic Neoplasms/pathology , Intestinal Mucosa/cytology , NF-kappa B/metabolism , Receptors, G-Protein-Coupled/metabolism , Stem Cells/cytology , Cells, Cultured , Colonic Neoplasms/metabolism , Humans , Intestinal Mucosa/metabolism , NF-kappa B/genetics , Receptors, G-Protein-Coupled/genetics , Stem Cells/metabolism , Wnt Signaling Pathway
13.
Nephrol Dial Transplant ; 36(4): 618-631, 2021 03 29.
Article in English | MEDLINE | ID: mdl-33367746

ABSTRACT

BACKGROUND: In chronic kidney disease, serum phosphorus (P) elevations stimulate parathyroid hormone (PTH) production, causing severe alterations in the bone-vasculature axis. PTH is the main regulator of the receptor activator of nuclear factor κB (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) system, which is essential for bone maintenance and also plays an important role in vascular smooth muscle cell (VSMC) calcification. The discovery of a new RANKL receptor, leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4), which is important for osteoblast differentiation but with an unknown role in vascular calcification (VC), led us to examine the contribution of LGR4 in high P/high PTH-driven VC. METHODS: In vivo studies were conducted in subtotally nephrectomized rats fed a normal or high P diet, with and without parathyroidectomy (PTX). PTX rats were supplemented with PTH(1-34) to achieve physiological serum PTH levels. In vitro studies were performed in rat aortic VSMCs cultured in control medium, calcifying medium (CM) or CM plus 10-7 versus 10-9 M PTH. RESULTS: Rats fed a high P diet had a significantly increased aortic calcium (Ca) content. Similarly, Ca deposition was higher in VSMCs exposed to CM. Both conditions were associated with increased RANKL and LGR4 and decreased OPG aorta expression and were exacerbated by high PTH. Silencing of LGR4 or parathyroid hormone receptor 1 (PTH1R) attenuated the high PTH-driven increases in Ca deposition. Furthermore, PTH1R silencing and pharmacological inhibition of protein kinase A (PKA), but not protein kinase C, prevented the increases in RANKL and LGR4 and decreased OPG. Treatment with PKA agonist corroborated that LGR4 regulation is a PTH/PKA-driven process. CONCLUSIONS: High PTH increases LGR4 and RANKL and decreases OPG expression in the aorta, thereby favouring VC. The hormone's direct pro-calcifying actions involve PTH1R binding and PKA activation.


Subject(s)
Myocytes, Smooth Muscle/metabolism , Osteoprotegerin/metabolism , Parathyroid Hormone/pharmacology , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, G-Protein-Coupled/metabolism , Vascular Calcification/metabolism , Animals , Calcium-Regulating Hormones and Agents/pharmacology , Gene Expression Regulation/drug effects , Ligands , Male , NF-kappa B/metabolism , Osteoprotegerin/genetics , RANK Ligand/genetics , Rats , Rats, Wistar , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptors, G-Protein-Coupled/genetics
14.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946652

ABSTRACT

Leucine-rich repeats containing G protein-coupled receptor 4 (LGR4) is a receptor that belongs to the superfamily of G protein-coupled receptors that can be activated by R-spondins (RSPOs), Norrin, circLGR4, and the ligand of the receptor activator of nuclear factor kappa-B (RANKL) ligands to regulate signaling pathways in normal and pathological processes. LGR4 is widely expressed in different tissues where it has multiple functions such as tissue development and maintenance. LGR4 mainly acts through the Wnt/ß-catenin pathway to regulate proliferation, survival, and differentiation. In cancer, LGR4 participates in tumor progression, invasion, and metastasis. Furthermore, recent evidence reveals that LGR4 is essential for the regulation of the cancer stem cell population by controlling self-renewal and regulating stem cell properties. This review summarizes the function of LGR4 and its ligands in normal and malignant processes.


Subject(s)
Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Carcinogenesis , Female , Humans , Ligands , Male , Mice , MicroRNAs/genetics , Models, Biological , Neoplasms/etiology , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Tissue Distribution , Wnt Signaling Pathway
15.
Int J Mol Sci ; 23(1)2021 Dec 30.
Article in English | MEDLINE | ID: mdl-35008827

ABSTRACT

The significance of cancer stem cells (CSCs) in initiation and progression of colon cancer (CC) has been established. In this study, we investigated the utility of measuring mRNA expression levels of CSC markers EpCAM, LGR5 and LGR4 for predicting survival outcome in surgically treated CC patients. Expression levels were determined in 5 CC cell lines, 66 primary CC tumors and 382 regional lymph nodes of 121 CC patients. Prognostic relevance was determined using Kaplan-Meier survival and Cox regression analyses. CC patients with lymph nodes expressing high levels of EpCAM, LGR5 or LGR4 (higher than a clinical cutoff of 0.07, 0.06 and 2.558 mRNA copies/18S rRNA unit, respectively) had a decreased mean survival time of 32 months for EpCAM and 42 months for both LGR5 and LGR4 at a 12-year follow-up (p = 0.022, p = 0.005 and p = 0.011, respectively). Additional patients at risk for recurrence were detected when LGR5 was combined with the biomarkers CXCL17 or CEA plus CXCL16. In conclusion, the study underscores LGR5 as a particularly useful prognostic biomarker and illustrates the strength of combining biomarkers detecting different subpopulations of cancer cells and/or cells in the tumor microenvironment for predicting recurrence.


Subject(s)
Biomarkers, Tumor/genetics , Colonic Neoplasms/genetics , Epithelial Cell Adhesion Molecule/genetics , Gene Expression Regulation, Neoplastic , Lymph Nodes/metabolism , Neoplastic Stem Cells/metabolism , Receptors, G-Protein-Coupled/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Colonic Neoplasms/surgery , Epithelial Cell Adhesion Molecule/metabolism , Humans , Kaplan-Meier Estimate , Lymph Nodes/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Risk Factors
16.
Gynecol Oncol ; 159(3): 839-849, 2020 12.
Article in English | MEDLINE | ID: mdl-32980127

ABSTRACT

OBJECTIVE: High-grade serous ovarian cancer (HGSOC) is lethal mainly due to extensive metastasis. Cancer cell stem-like properties are responsible for HGSOC metastasis. LGR4, a G-protein-coupled receptor, is involved in the maintenance of stem cell self-renewal and activity in some human organs. METHODS: TCGA and CCLE databases were interrogated for gene mRNA in ovarian cancer tissues and cell lines. Gain and loss of functions of LGR4, ELF3, FZD5 and WNT7B were performed to identify their roles in ovarian cancer cell epithelial phenotype and stem-like properties. In vivo experiments were performed to observe the effect of LGR4 on ovarian cancer cell growth and peritoneal seeding. The binding of ELF3 to LGR4 gene promoter was investigated by dual-luciferase reporter assays and ChIP. RESULTS: LGR4 was shown to be overexpressed in HGSOCs and maintain the epithelial phenotype of HGSOC cells. LGR4 knockdown suppressed POU5F1, SOX2, PROM1 (CD133) and ALDH1A2 expression. Furthermore, LGR4 knockdown reduced CD133+ and ALDH+ subpopulations and impaired tumorisphere formation. To the contrary, LGR4 overexpression enhanced POU5F1 and SOX2 expression and tumorisphere formation capacity. LGR4 knockdown inhibited HGSOC cell growth and peritoneal seeding in xenograft models. Mechanistically, LGR4 and ELF3, an epithelium-specific transcription factor, formed a reciprocal regulatory loop, which was positively modulated by WNT7B/FZD5 ligand-receptor pair. Consistently, knockdown of ELF3, WNT7B, and FZD5, respectively, disrupted HGSOC cell epithelial phenotype and stem-like properties. CONCLUSION: Together, these data demonstrate that WNT7B/FZD5-LGR4/ELF3 axis maintains HGSOC cell epithelial phenotype and stem-like traits; targeting this axis may prevent HGSOC metastasis.


Subject(s)
Carcinoma, Ovarian Epithelial/secondary , Epithelial Cells/pathology , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Receptors, G-Protein-Coupled/metabolism , Animals , Carcinoma, Ovarian Epithelial/diagnosis , Cell Line, Tumor , Cell Self Renewal , DNA-Binding Proteins/metabolism , Female , Frizzled Receptors/metabolism , Gene Knockdown Techniques , Humans , Mice , Neoplasm Grading , Ovarian Neoplasms/diagnosis , Ovary/cytology , Ovary/pathology , Peritoneal Neoplasms/diagnosis , Peritoneum/cytology , Peritoneum/pathology , Proto-Oncogene Proteins c-ets/metabolism , Receptors, G-Protein-Coupled/genetics , Transcription Factors/metabolism , Wnt Proteins/metabolism , Xenograft Model Antitumor Assays
17.
J Bone Miner Metab ; 38(5): 658-669, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32399675

ABSTRACT

INTRODUCTION: Leucine-rich repeat-containing G protein-coupled receptor 4 (LGR4) could affect differentiation of osteoblasts and bone mass through potentiating Wnt/ß-catenin signaling. LGR4 is also relevant to glycolipid metabolism. The present study aims to explore the relationship between genetic variations in LGR4 gene and peak bone mineral density (peak BMD) and body composition phenotypes in Chinese nuclear families. MATERIALS AND METHODS: 22 single-nucleotide polymorphisms (SNPs) were selected and five blocks were constructed in LGR4. Body composition (lean mass and fat mass) and peak BMD were measured by dual-energy X-ray absorptiometry (DXA). Quantitative transmission disequilibrium test (QTDT) analysis was used to explore the relationship between LGR4 genotypes and the mentioned phenotypes. RESULTS: For QTDT analysis after 1000 permutations, significant within-family associations were observed between rs11029986 and total fat mass (TFM) and percentage of TFM (PFM) (P = 0.014 and 0.011, respectively), rs12787344, rs4128868, rs4923445, and rs7936621 and body mass index (BMI) (P = 0.008, 0.003, 0.046, and 0.003, respectively), rs11029986 and total hip BMD (P = 0.026), and rs12796247, rs2219783, and lumbar spine BMD (P = 0.013 and 0.027, respectively). Haplotypes GCGT and AAGC (both in block 3) were observed in significant within-family association with BMI (P = 0.003 and 0.002, respectively). CONCLUSION: It is the first family-based association analysis to explore and demonstrate significant associations between LGR4 genotypes and variations of peak BMD and body composition in young Chinese men. The results are consistent with the findings that recent studies revealed, and confirm the critical relationship between LGR4 gene and both BMD and body composition.


Subject(s)
Body Composition/genetics , Bone Density/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide/genetics , Receptors, G-Protein-Coupled/genetics , Absorptiometry, Photon , Adiposity , Asian People/genetics , Female , Haplotypes/genetics , Humans , Linkage Disequilibrium/genetics , Male , Middle Aged
18.
Proc Natl Acad Sci U S A ; 114(2): 376-381, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28028233

ABSTRACT

The unrestrained growth of tumor cells is generally attributed to mutations in essential growth control genes, but tumor cells are also affected by, or even addicted to, signals from the microenvironment. As therapeutic targets, these extrinsic signals may be equally significant as mutated oncogenes. In multiple myeloma (MM), a plasma cell malignancy, most tumors display hallmarks of active Wnt signaling but lack activating Wnt-pathway mutations, suggesting activation by autocrine Wnt ligands and/or paracrine Wnts emanating from the bone marrow (BM) niche. Here, we report a pivotal role for the R-spondin/leucine-rich repeat-containing G protein-coupled receptor 4 (LGR4) axis in driving aberrant Wnt/ß-catenin signaling in MM. We show that LGR4 is expressed by MM plasma cells, but not by normal plasma cells or B cells. This aberrant LGR4 expression is driven by IL-6/STAT3 signaling and allows MM cells to hijack R-spondins produced by (pre)osteoblasts in the BM niche, resulting in Wnt (co)receptor stabilization and a dramatically increased sensitivity to auto- and paracrine Wnts. Our study identifies aberrant R-spondin/LGR4 signaling with consequent deregulation of Wnt (co)receptor turnover as a driver of oncogenic Wnt/ß-catenin signaling in MM cells. These results advocate targeting of the LGR4/R-spondin interaction as a therapeutic strategy in MM.


Subject(s)
Membrane Glycoproteins/metabolism , Multiple Myeloma/metabolism , Osteoblasts/metabolism , Receptors, G-Protein-Coupled/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Interleukin-6/metabolism , Ligands , Mice , Protein Binding/physiology , STAT3 Transcription Factor/metabolism , beta Catenin/metabolism
19.
Dev Biol ; 441(1): 42-51, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29859889

ABSTRACT

Coronary artery anomalies are common congenital disorders with serious consequences in adult life. Coronary circulation begins when the coronary stems form connections between the aorta and the developing vascular plexus. We recently identified the WNT signaling modulator R-spondin 3 (Rspo3), as a crucial regulator of coronary stem proliferation. Using expression analysis and tissue-specific deletion we now demonstrate that Rspo3 is primarily produced by cardiomyocytes. Moreover, we have employed CRISPR/Cas9 technology to generate novel Lgr4-null alleles that showed a significant decrease in coronary stem proliferation and thus phenocopied the coronary artery defects seen in Rspo3 mutants. Interestingly, Lgr4 mutants displayed slightly hypomorphic right ventricles, an observation also made after myocardial specific deletion of Rspo3. These results shed new light on the role of Rspo3 in heart development and demonstrate that LGR4 is the principal R-spondin 3 receptor in the heart.


Subject(s)
Coronary Vessels/embryology , Heart/embryology , Myocytes, Cardiac/metabolism , Receptors, G-Protein-Coupled/metabolism , Thrombospondins/metabolism , Wnt Signaling Pathway/physiology , Animals , Coronary Circulation/physiology , Coronary Vessels/cytology , Mice , Mice, Transgenic , Myocytes, Cardiac/cytology , Receptors, G-Protein-Coupled/genetics , Thrombospondins/genetics
20.
J Biol Chem ; 293(25): 9759-9769, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29752411

ABSTRACT

The four R-spondins (RSPO1-4) strongly potentiate Wnt signaling and play critical roles in normal development, adult stem cell survival, and cancer development and aggressiveness. All four RSPOs have been suggested to potentiate Wnt signaling by binding to three related receptors, i.e. leucine-rich repeat-containing, G protein-coupled receptors 4, 5, and 6 (LGR4/5/6), and then inducing the clearance of two E3 ubiquitin ligases (RNF43 and ZNRF3) that otherwise would ubiquitinate Wnt receptors for degradation. Here, we show that RSPO1-4 have differential dependence on LGRs in potentiating Wnt/ß-catenin signaling and that RSPO2 can enhance this pathway without any LGR. LGR4 knockout (LGR4KO) in HEK293 cells completely abrogated the Wnt/ß-catenin signaling response to RSPO1 and RSPO4 and strongly impaired the response to RSPO3. RSPO2, however, retained robust activity albeit with decreased potency. Complete rescue of RSPO1-4 activity in LGR4KO cells required the seven-transmembrane domain of LGR4. Furthermore, an RSPO2 mutant with normal binding affinity to ZNRF3 but no or little binding to LGR4 or LGR5 still potentiated Wnt/ß-catenin signaling in vitro, supported the growth of intestinal organoids ex vivo, and stimulated intestinal crypt growth in vivo Mechanistically, RSPO2 could increase Wnt receptor levels in the absence of any LGR without affecting ZNRF3 endocytosis and stability. These findings suggest that RSPO1-4 use distinct mechanisms in regulating Wnt and other signaling pathways, which have important implications for understanding the pleiotropic functions of RSPOs and LGRs in both normal and cancer development.


Subject(s)
Signal Transduction , Thrombospondins/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Organoids/cytology , Organoids/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Thrombospondins/genetics , Wnt Proteins/genetics , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL