Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Bioorg Chem ; 143: 107066, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38185009

ABSTRACT

Repeated chromatography of the CH2Cl2 and EtOAc soluble fractions from the methanol extract of Belamcanda chinensis root yielded six new sucrosephenylpropanoid esters (1-6) and twenty-one known compounds (7-27). The structures of 1-6 were elucidated using diverse nuclear magnetic resonance (NMR) techniques and high-resolution mass spectrometry (HRMS) data analysis, together with chemical methods. All the twenty-seven isolated compounds were evaluated for their anti-osteoclastogenic activities. Preliminary screening results revealed that compounds 1 and 19 exhibited strong effects against RANKL-induced osteoclast formation in RAW264.7 cells. In addition, the treatment of mouse bone marrow macrophages (BMMs) with compounds 1 and 19 significantly decreased RANKL-induced TRAP-positive multinucleated osteoclast formation in a concentration-dependent manner without affecting cell viability. Further bioassay investigation showed that compounds 1 and 19 inhibited the expression of some osteoclast-specific marker genes and the transcription factor nuclear factor of activated T cells cytoplasmic 1 (NFATc1) in response to RANKL. To the best of our knowledge, this is the first investigation of anti-osteoclastogenic activity for compounds isolated from B. chinensis.


Subject(s)
Bone Resorption , Isoflavones , Animals , Mice , Bone Resorption/drug therapy , Bone Resorption/metabolism , Bone Resorption/prevention & control , Cell Differentiation , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoclasts , Osteogenesis/drug effects , Isoflavones/chemistry , Isoflavones/pharmacology , Plant Roots/chemistry
2.
Food Chem Toxicol ; 168: 113407, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36075474

ABSTRACT

Glucocorticoid (GC) exposure can lead to deterioration of the structure and function of hippocampal neurons and is closely involved in Alzheimer's disease (AD). Amyloid-ß (Aß) overproduction is an important aspect of AD pathogenesis. Our study mainly investigated the mechanism of chronic GC exposure in accelerating Aß production in primary cultured hippocampal neurons from APP/PS1 mice. The results indicated that chronic dexamethasone (DEX, 1 µM) significantly accelerated neuronal damage and Aß accumulation in hippocampal neurons from APP/PS1 mice. Meanwhile, DEX exposure markedly upregulated APP, NCSTN, BACE1 and p-Tau/Tau expression in hippocampal neurons from APP/PS1 mice. Our study also indicated that chronic DEX exposure significantly increased intracellular Ca2+ ([Ca2+]i) levels and the expressions of p-PLC, CN and NFAT1 in hippocampal neurons from APP/PS1 mice. We further found that stabilizing intracellular calcium homeostasis with 2-APB (50 µM) and SKF-96365 (10 µM) significantly alleviated neuronal damage and Aß accumulation in chronic DEX-induced hippocampal neurons from APP/PS1 mice. Additionally, dual luciferase assays showed that NFAT1 upregulated NCSTN transactivation, which was further increased upon DEX treatment. This study suggests that chronic DEX exposure accelerates Aß accumulation by activating calcium-mediated CN-NFAT1 signaling in hippocampal neurons from APP/PS1 mice, which may be closely related to the acceleration of AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Glucocorticoids , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/metabolism , Calcium/metabolism , Dexamethasone/toxicity , Disease Models, Animal , Glucocorticoids/adverse effects , Glucocorticoids/toxicity , Hippocampus/metabolism , Mice , Mice, Transgenic , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/metabolism , Neurons/metabolism , Neurotoxicity Syndromes/metabolism
3.
J Cell Physiol ; 236(12): 8239-8252, 2021 12.
Article in English | MEDLINE | ID: mdl-34192358

ABSTRACT

Many bone diseases such as osteoporosis and periodontitis are caused by hyperactivation of osteoclasts. Calcium (Ca2+ ) signals are crucial for osteoclast differentiation and function. Thus, the blockade of Ca2+ signaling may be a strategy for regulating osteoclast activity and has clinical implications. Flunarizine (FN) is a Ca2+ channel antagonist that has been used for reducing migraines. However, the role of FN in osteoclast differentiation and function remains unknown. Here, we investigated whether FN regulates osteoclastogenesis and elucidated the molecular mechanism. FN inhibited osteoclast differentiation along with decreased expression of nuclear factor of activated T cells, cytoplasmic 1 (NFATc1), and attenuated osteoclast maturation and bone resorption. FN inhibition of osteoclast differentiation was restored by ectopic expression of constitutively active NFATc1. FN reduced calcium oscillations and its inhibition of osteoclast differentiation and resorption function was reversed by ionomycin, an ionophore that binds Ca2+ . FN also inhibited Ca2+ /calmodulin-dependent protein kinase IV (CaMKIV) and calcineurin leading to a decrease in the cAMP-responsive element-binding protein-dependent cFos and peroxisome proliferator-activated receptor-γ coactivator 1ß expression, and NFATc1 nuclear translocation. These results indicate that FN inhibits osteoclastogenesis via regulating CaMKIV and calcineurin as a Ca2+ channel blocker. In addition, FN-induced apoptosis in osteoclasts and promoted osteogenesis. Furthermore, FN protected lipopolysaccharide- and ovariectomy-induced bone destruction in mouse models, suggesting that it has therapeutic potential for treating inflammatory bone diseases and postmenopausal osteoporosis.


Subject(s)
Calcium Signaling/drug effects , Flunarizine/antagonists & inhibitors , Osteoclasts/drug effects , Osteogenesis/drug effects , Animals , Bone Resorption/drug therapy , Bone Resorption/metabolism , Calcineurin/metabolism , Cell Differentiation/drug effects , Flunarizine/metabolism , Humans , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Osteogenesis/physiology , Osteoporosis/drug therapy , Osteoporosis/metabolism , RANK Ligand/metabolism
4.
Arthritis Rheumatol ; 73(7): 1145-1154, 2021 07.
Article in English | MEDLINE | ID: mdl-33512089

ABSTRACT

OBJECTIVE: We have previously reported that stimulation of mouse bone marrow-derived macrophages with tumor necrosis factor (TNF) and interleukin-6 (IL-6) induces differentiation of osteoclast-like cells. We undertook this study to clarify the characterization and function of human TNF and IL-6-induced osteoclasts using peripheral blood collected from patients with rheumatoid arthritis (RA) and healthy donors. METHODS: Peripheral blood monocytes were cultured with a combination of TNF and IL-6, TNF alone, IL-6 alone, or with RANKL, and their bone resorption ability was evaluated. Expression levels of NFATc1, proinflammatory cytokines, and matrix metalloproteinase 3 were analyzed. The effects of NFAT inhibitor and JAK inhibitor were examined. Furthermore, the relationship between the number of TNF and IL-6-induced osteoclasts or RANKL-induced osteoclasts differentiated from peripheral blood mononuclear cells (PBMCs) in patients with RA and the modified total Sharp score (mTSS) or whole-body bone mineral density (BMD) was examined. RESULTS: Peripheral blood monocytes stimulated with a TNF and IL-6-induced osteoclasts were shown to demonstrate the ability to absorb bone matrix. Cell differentiation was not inhibited by the addition of osteoprotegerin. Stimulation with a combination of TNF and IL-6 promoted NFATc1 expression, whereas the NFAT and JAK inhibitors prevented TNF and IL-6-induced osteoclast formation. Expression levels of IL1ß, TNF, IL12p40, and MMP3 were significantly increased in TNF and IL-6-induced osteoclasts, but not in RANKL-induced osteoclasts. The number of TNF and IL-6-induced osteoclasts differentiated from PBMCs in patients with RA positively correlated with the mTSS, whereas RANKL-induced osteoclast numbers negatively correlated with the whole-body BMD of the same patients. CONCLUSION: Our results demonstrate that TNF and IL-6-induced osteoclasts may contribute to the pathology of inflammatory arthritis associated with joint destruction, such as RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Bone Resorption/immunology , Interleukin-6/immunology , Osteoclasts/immunology , Tumor Necrosis Factor-alpha/immunology , Aged , Arthritis, Rheumatoid/diagnostic imaging , Arthritis, Rheumatoid/metabolism , Bone Density , Bone Resorption/diagnostic imaging , Bone Resorption/metabolism , Case-Control Studies , Cytokines/drug effects , Cytokines/immunology , Cytokines/metabolism , Female , Humans , Interleukin-12 Subunit p40/drug effects , Interleukin-12 Subunit p40/immunology , Interleukin-12 Subunit p40/metabolism , Interleukin-1beta/drug effects , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-6/pharmacology , Male , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/immunology , Matrix Metalloproteinase 3/metabolism , Middle Aged , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Osteogenesis/immunology , RANK Ligand/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
5.
Drug Des Devel Ther ; 14: 4189-4203, 2020.
Article in English | MEDLINE | ID: mdl-33116407

ABSTRACT

INTRODUCTION: Osteoporosis is a metabolic bone disease characterized by reduced bone quantity and microstructure, typically owing to increased osteoclastogenesis and/or enhanced osteoclastic bone resorption, resulting in uncontrolled bone loss, which primarily affects postmenopausal women. In consideration of the severe side effects of current drugs for osteoporosis, new safe and effective medications are necessary. Pristimerin (Pri), a quinone methide triterpene extracted from Celastraceae and Hippocrateaceae members, exhibits potent antineoplastic and anti-inflammatory effects. However, its effect on osteoclasts remains unknown. MATERIALS AND METHODS: We evaluated the anti-osteoclastogenic and anti-resorptive effect of Pri on bone marrow-derived osteoclasts and its underlying mechanism in vitro. In addition, the protective effect of Pri on ovariectomy model was also explored in vivo. RESULTS: In vitro, Pri inhibited osteoclast differentiation and mature osteoclastic bone resorption in a time- and dose-dependent manner. Further, Pri suppressed the expression of osteoclast-related genes and the activation of key proteins. Pri also inhibited the early activation of ERK, JNK MAPK, and AKT signaling pathways in bone marrow-derived macrophages (BMMs), ultimately inhibiting the induction and activation of the crucial osteoclast transcriptional factor nuclear factor of activated T-cell cytoplasmic 1 (NFATc1). In vivo, consistent with our in vitro data, Pri clearly prevented ovariectomy-induced bone loss. CONCLUSION: Our data showed that Pri inhibits the differentiation and activation of osteoclasts in vitro and in vivo, and could be a promising candidate for treating osteoporosis.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone Resorption/prevention & control , Cell Differentiation/drug effects , Osteoclasts/drug effects , Osteoporosis, Postmenopausal/prevention & control , Ovariectomy , Pentacyclic Triterpenes/pharmacology , Animals , Bone Resorption/diagnostic imaging , Dose-Response Relationship, Drug , Female , Humans , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/drug effects , Osteogenesis/drug effects , Signal Transduction/drug effects , X-Ray Microtomography
6.
Sci Rep ; 10(1): 10914, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32616823

ABSTRACT

Plant cell cultures have been exploited to provide stable production and new secondary metabolites for better pharmacological activity. Fractionation of adventitious root cultures of Echinacea purpurea resulted in the isolation of eleven constituents, including three new compounds. The structures of the three new compounds were determined to be an alkylamide (1), a polyacetylene (2) and a lignan (3) on the basis of combined spectroscopic analysis. To discover new types of antiresorptive agents, we screened for new compounds that regulate osteoclast differentiation, and survival. Among three new compounds, echinalkamide (compound 1) had considerably inhibitory effects on RANKL-induced osteoclast differentiation, and on proliferation of osteoclasts and efficiently attenuated osteoclastic bone resorption without toxicity. In addition, echinalamide treatment inhibited the osteoclast-specific gene expression level. Echinalkamide achieved this inhibitory effect by disturbing phosphorylation of MAPK and activation of osteoclast transcription factors c-Fos and NFATc1. Conclusionally, our study investigated that echinalkamide remarkably inhibited osteoclast differentiation and osteoclast specific gene expression through repression of the MAPK-c-Fos-NFATC1 cascade.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone Resorption/prevention & control , Echinacea/chemistry , Osteogenesis/drug effects , Phytotherapy , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacology , Antioxidants/isolation & purification , Antioxidants/pharmacology , Bone Density Conservation Agents/isolation & purification , Bone Resorption/drug therapy , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/metabolism , Nitric Oxide/biosynthesis , Phosphorylation/drug effects , Plant Roots/chemistry , Protein Processing, Post-Translational/drug effects , RANK Ligand/pharmacology , RAW 264.7 Cells , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
7.
Exp Cell Res ; 394(1): 112115, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32473224

ABSTRACT

Postmenopausal osteoporosis is very common in women. Currently, many kinds of new drugs are being developed for this disease. Postmenopausal osteoporosis is closely related to overactivity of osteoclasts in body. Shikonin is purple red naphthoquinone pigment extracted from lithospermum, which has anti-inflammation, antivirus, anticancer and other bioactivities. At the same time, it has been proved that shikonin can promote the proliferation and differentiation of osteoblasts, but its influence on osteoclasts and molecular mechanism are unknown. Our study showed that shikonin could inhibit the activity and formation of RANKL-mediated osteoclasts depending on dose without affecting the activity of bone marrow macrophages (BMM). In addition, we have also found that shikonin can inhibit the expression of specific marker gene of osteoclasts, including nuclear factor of activated T cells cytoplasmic 1 (NFATc1), cathepsin K (Ctsk), tartrate resistant acid phosphatase (TRAcP) and calcitonin receptor. Shikonin also could promote the proliferation of MC3T3-E1, increasing the expression of mRNA related to osteogenesis, like the expression of bone morphogenetic protein-2 (BMP-2), alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2) and osteocalcin (OCN). Luciferase reporter gene assay and Western blot analysis further indicated that shikonin could inhibit the activity of RANKL-induced NF-κB and NFAT receptors. Moreover, shikonin can also slow down bone loss of ovariectomized (OVX) mice by inhibiting the activity of osteoclasts. This work explains the molecular mechanism of shikonin in RANKL-mediated formation of osteoclasts, and reveals the potential of further developing shikonin into a new drug for prevention and treatment of postmenopausal osteoporosis.


Subject(s)
NF-kappa B/drug effects , NFATC Transcription Factors/drug effects , Naphthoquinones/pharmacology , Osteoclasts/drug effects , Osteogenesis/drug effects , Animals , Bone Resorption/drug therapy , Bone Resorption/metabolism , Cell Differentiation , Mice , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Naphthoquinones/metabolism , Osteoclasts/metabolism , Osteogenesis/genetics , Osteoporosis/metabolism
8.
J Neurochem ; 154(5): 519-529, 2020 09.
Article in English | MEDLINE | ID: mdl-31755556

ABSTRACT

Hair cell (HC) degeneration causes hearing loss in millions of people worldwide. Aminoglycoside exposure is one major cause of sensory HC damage. Aminoglycosides generate free radicals within the inner ear, permanently damaging sensory cells, and thus causing hearing loss. Hearing protection requires strategies to overcome the apparently irreversible loss of HCs in mammals. The nuclear factor of activated T cells (NFAT) inhibitor 11R-VIVIT reportedly protects HCs from gentamicin toxicity. Here we investigated whether the combination of 11R-VIVIT with the antioxidant L-carnitine or N-acetylcysteine could protect mouse cochlear HCs from gentamicin damage. Compared to single-component treatment, combined treatment with 11R-VIVIT plus L-carnitine yielded significant protection from gentamicin, and 11R-VIVIT plus N-acetylcysteine provided almost complete protection of HCs from gentamicin. Caspase activity in organ of Corti was significantly reduced by combined treatment with 11R-VIVIT + N-acetylcysteine + gentamicin, compared to 11R-VIVIT + gentamicin or gentamicin alone. Analysis of relative gene expression by qPCR revealed down-regulation of the pro-apoptotic genes Fasl and Casp9, and up-regulation of the antioxidant genes Hmox1 and Nrf2 after treatment with 11R-VIVIT + N-acetylcysteine + gentamicin, compared to single-compound treatment or gentamicin alone in cultures. Selective NFAT inhibition by 11R-VIVIT may be a good strategy for preventing gentamicin-induced HC damage. L-carnitine and N-acetylcysteine, with their ROS-reducing properties, contribute to the synergistic effectiveness with 11R-VIVIT by decreasing ROS-induced NFAT translocation. Our data suggest that a combined approach of NFAT inhibition together with an antioxidant, like N-acetylcysteine, could be useful for hearing loss treatment and/or prevention. Cover Image for this issue: https://doi.org/10.1111/jnc.14759.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Hair Cells, Auditory/drug effects , Organ of Corti/drug effects , Aminoglycosides/metabolism , Animals , Antioxidants/metabolism , Gentamicins/metabolism , Hair Cells, Auditory/metabolism , Mice , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/metabolism , Organ of Corti/metabolism , Protective Agents/pharmacology , T-Lymphocytes/metabolism
9.
Diabetes ; 69(3): 424-435, 2020 03.
Article in English | MEDLINE | ID: mdl-31806622

ABSTRACT

Central to the development of diabetic macro- and microvascular disease is endothelial dysfunction, which appears well before any clinical sign but, importantly, is potentially reversible. We previously demonstrated that hyperglycemia activates nuclear factor of activated T cells (NFAT) in conduit and medium-sized resistance arteries and that NFAT blockade abolishes diabetes-driven aggravation of atherosclerosis. In this study, we test whether NFAT plays a role in the development of endothelial dysfunction in diabetes. NFAT-dependent transcriptional activity was elevated in skin microvessels of diabetic Akita (Ins2 +/- ) mice when compared with nondiabetic littermates. Treatment of diabetic mice with the NFAT blocker A-285222 reduced NFATc3 nuclear accumulation and NFAT-luciferase transcriptional activity in skin microvessels, resulting in improved microvascular function, as assessed by laser Doppler imaging and iontophoresis of acetylcholine and localized heating. This improvement was abolished by pretreatment with the nitric oxide (NO) synthase inhibitor l-N G-nitro-l-arginine methyl ester, while iontophoresis of the NO donor sodium nitroprusside eliminated the observed differences. A-285222 treatment enhanced dermis endothelial NO synthase expression and plasma NO levels of diabetic mice. It also prevented induction of inflammatory cytokines interleukin-6 and osteopontin, lowered plasma endothelin-1 and blood pressure, and improved mouse survival without affecting blood glucose. In vivo inhibition of NFAT may represent a novel therapeutic modality to preserve endothelial function in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Endothelium, Vascular/drug effects , Microvessels/drug effects , NFATC Transcription Factors/antagonists & inhibitors , Pyrazoles/pharmacology , Acetylcholine/pharmacology , Animals , Blood Pressure/drug effects , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Endothelin-1/drug effects , Endothelin-1/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Enzyme Inhibitors/pharmacology , Insulin/genetics , Interleukin-6/metabolism , Iontophoresis , Mice , Microvessels/metabolism , Microvessels/physiopathology , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/drug effects , Nitric Oxide Synthase Type III/metabolism , Nitroprusside/pharmacology , Osteopontin/drug effects , Osteopontin/metabolism , Skin/blood supply , Survival Rate , Ultrasonography, Doppler , Vasodilator Agents/pharmacology
10.
Neurochem Int ; 131: 104548, 2019 12.
Article in English | MEDLINE | ID: mdl-31539560

ABSTRACT

3-Nitropropionic acid (3-NP) induces a spectrum of Huntington's disease (HD)-like neuropathologies in the rat striatum. The present study aimed to demonstrate the neuroprotective effect of lercanidipine (LER) in rats with 3-NP-induced neurotoxicity, address the possible additional protective effect of combined treatment with bone marrow-derived mesenchymal stem cells (BM-MSCs) and LER, and investigate the possible involvement of the Ca2+/calcineurin (CaN)/nuclear factor of activated T cells c4 (NFATc4) and Wnt/ß-catenin signalling pathways. Rats were injected with 3-NP (10 mg/kg/day, i.p.) for two weeks and were divided into four subgroups; the first served as the control HD group, the second received a daily dose of LER (0.5 mg/kg, i.p.), the third received a single injection of BM-MSCs (1 x 106/rat, i.v.) and the last received a combination of both BM-MSCs and LER. The combined therapy improved motor and behaviour performance. Meanwhile, this treatment led to a marked reduction in striatal cytosolic Ca2+, CaN, tumour necrosis factor-alpha, and NFATc4 expression and the Bax/Bcl2 ratio. Combined therapy also increased striatal brain-derived neurotrophic factor, FOXP3, Wnt, and ß-catenin protein expression. Furthermore, haematoxylin-eosin and Nissl staining revealed an amelioration of striatum tissue injury with the combined treatment. In conclusion, the current study provides evidence for a neuroprotective effect of LER and/or BM-MSCs in 3-NP-induced neurotoxicity in rats. Interestingly, combined LER/BM-MSC therapy was superior to cell therapy alone in inhibiting 3-NP-induced neurological insults via modulation of the Ca2+/CaN/NFATc4 and Wnt/ß-catenin signalling pathways. LER/BM-MSC combined therapy may represent a feasible approach for improving the beneficial effects of stem cell therapy in HD.


Subject(s)
Calcium Channel Blockers/therapeutic use , Dihydropyridines/therapeutic use , Huntington Disease/therapy , Mesenchymal Stem Cell Transplantation/methods , Signal Transduction/drug effects , Animals , Behavior, Animal/drug effects , Calcineurin/drug effects , Calcium Signaling/drug effects , Cytosol/drug effects , Cytosol/metabolism , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Male , NFATC Transcription Factors/drug effects , Nerve Tissue Proteins/drug effects , Nitro Compounds , Propionates , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism , Wnt Signaling Pathway/drug effects , bcl-2-Associated X Protein/metabolism , beta Catenin/drug effects
11.
Med Sci Monit ; 25: 2228-2237, 2019 Mar 26.
Article in English | MEDLINE | ID: mdl-30913205

ABSTRACT

BACKGROUND The inhibitory effect of arsenic trioxide (As2O3) on lung cancer has been reported in some preclinical studies. However, its effect on small cell lung cancer (SCLC) has been poorly explored. Calcineurin and its substrate, nuclear factor of activated T cells (NFAT), mediate the downstream signaling of VEGF, and is critical in the process endothelium activation and tumor metastasis. In this study, we aimed to evaluate whether As2O3 had inhibitory effects on endothelial cells activation and the metastasis of SCLC, and to explore the possible mechanisms. MATERIAL AND METHODS In vitro, human umbilical vein endothelial cells (HUVECs) were used. Cell Counting Kit-8 assay and cell migration assay were performed to determine the effect of As2O3 on HUVECs proliferation and migration. The level of calcineurin, NFAT, downstream factors for Down syndrome candidate region 1 (DSCR1), and the endogenous inhibitor of calcineurin, were evaluated by quantitative PCR and western blotting. In vivo, SCLC metastasis models were established by injecting NCI-H446 cells into tail veins of nude mice. Tumor-bearing mice were treated with As2O3 or calcineurin inhibitor for 10 days, after which tumor metastasis in target organs was evaluated. RESULTS As2O3 significantly inhibited the proliferation and migration of endothelial cells. Also, As2O3 inhibited the expression levels of calcineurin, NFAT, and the downstream target genes CXCR7 and RND1, while it upregulated the level of DSCR1. Both As2O3 and calcineurin inhibitor exhibited notable inhibitory effect on the metastasis of SCLC, without obvious side effects. CONCLUSIONS These findings suggested that As2O3 had remarkable inhibitory effects on the endothelial cell activation and SCLC metastasis, and the mechanism might be related to the blocking of calcineurin-NFAT signaling by upregulating DSCR1.


Subject(s)
Arsenic Trioxide/pharmacology , NFATC Transcription Factors/drug effects , Small Cell Lung Carcinoma/drug therapy , Animals , Arsenic Trioxide/metabolism , Calcineurin/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , China , DNA-Binding Proteins , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intracellular Signaling Peptides and Proteins/drug effects , Male , Mice , Mice, Nude , Muscle Proteins/drug effects , NFATC Transcription Factors/metabolism , Neoplasm Metastasis/drug therapy , Neovascularization, Pathologic/metabolism , Receptors, CXCR/drug effects , Signal Transduction , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/drug effects , rho GTP-Binding Proteins/drug effects
12.
Arthritis Rheumatol ; 71(8): 1308-1318, 2019 08.
Article in English | MEDLINE | ID: mdl-30791224

ABSTRACT

OBJECTIVE: Lupus nephritis (LN) is a major determinant of morbidity and mortality in systemic lupus erythematosus (SLE). Pim-1 regulates lymphocyte proliferation and activation. The role of Pim-1 in autoimmune disease remains unclear. This study was undertaken to test the hypothesis that inhibition of Pim-1 would have therapeutic potential in patients with LN. METHODS: Pim-1 expression was analyzed in lupus-prone (NZB × NZW)F1 mice (n = 6), human peripheral blood mononuclear cells (PBMCs) from SLE patients (n = 10), and glomeruli from patients with LN (n = 8). The therapeutic effect of the Pim-1 inhibitor AZD1208 was assessed in the same murine lupus model (n = 10 mice per group). In vitro analysis was conducted to explore the mechanisms of action of Pim-1 in mouse and human podocytes after Pim-1 expression had been induced by anti-double-stranded DNA (anti-dsDNA) antibody-positive serum. Finally, MRL/lpr mice were used to confirm the therapeutic effects of Pim-1 inhibition in vivo (n = 10 mice per group). RESULTS: Up-regulation of Pim-1 was seen in renal lysates from diseased (NZB × NZW)F1 mice and in PBMCs from patients with SLE and renal biopsy tissue from patients with LN, relative to their control counterparts (each P < 0.05). The Pim-1 inhibitor AZD1208 reduced the severity of proteinuria, glomerulonephritis, renal immune complex deposits, and serum anti-dsDNA antibody levels, concomitant with the suppression of NFATc1 expression and NLRP3 inflammasome activation, in diseased (NZB × NZW)F1 mice (each P < 0.05 versus controls). Moreover, in mouse and human podocytes, Pim-1 knockdown with targeted small interfering RNA (siRNA) suppressed NFATc1 and NLRP3 inflammasome signaling in the presence of anti-dsDNA-positive serum (each P < 0.05 versus control siRNA). Mechanistically, Pim-1 modulated NLRP3 inflammasome activation through intracellular Ca2+ (P < 0.05 versus normal controls). The therapeutic effect of Pim-1 blockade was replicated in MRL/lpr mice. CONCLUSION: These data identify Pim-1 as a critical regulator of LN pathogenesis in patients with SLE. Targeting of the Pim-1/NFATc1/NLRP3 pathway might therefore have therapeutic potential in human LN.


Subject(s)
Cell Proliferation/drug effects , Lupus Erythematosus, Systemic/complications , Lupus Nephritis/drug therapy , Lymphocyte Activation/drug effects , Proto-Oncogene Proteins c-pim-1/pharmacology , Animals , Antibodies, Antinuclear/immunology , Disease Models, Animal , Humans , Inflammasomes/drug effects , Kidney/cytology , Kidney Glomerulus/metabolism , Leukocytes, Mononuclear , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/immunology , Mice , Mice, Inbred MRL lpr , NFATC Transcription Factors/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Podocytes/metabolism , Proto-Oncogene Proteins c-pim-1/immunology , Signal Transduction/drug effects
13.
J Cell Physiol ; 234(5): 6477-6488, 2019 05.
Article in English | MEDLINE | ID: mdl-30341897

ABSTRACT

Excessive osteoclast formation and function are considered as the main causes of bone lytic disorders such as osteoporosis and osteolysis. Therefore, the osteoclast is a potential therapeutic target for the treatment of osteoporosis or other osteoclast-related diseases. Helvolic acid (HA), a mycotoxin originally isolated from Aspergillus fumigatus , has been discovered as an effective broad-spectrum antibacterial agent and has a wide range of pharmacological properties. Herein, for the first time, HA was demonstrated to be capable of significantly inhibiting receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis and bone resorption in vitro by suppressing nuclear factor of activated T cells 1 (NFATc1) activation. This inhibition was followed by the dramatically decreased expression of NFATc1-targeted genes including Ctr (encoding calcitonin receptor), Acp5 (encoding tartrate-resistant acid phosphatase [TRAcP]), Ctsk (encoding cathepsin K), Atp6v0d2 (encoding the vacuolar H+ ATPase V0 subunit d2 [V-ATPase-d2]) and Mmp9 (encoding matrix metallopeptidase 9) which are osteoclastic-specific genes required for osteoclast formation and function. Mechanistically, HA was shown to greatly attenuate multiple upstream pathways including extracellular signal-regulated kinase (ERK) phosphorylation, c-Fos signaling, and intracellular Ca 2+ oscillation, but had little effect on nuclear factor-κB (NF-κB) activation. In addition, HA also diminished the RANKL-induced generation of intracellular reactive oxygen species. Taken together, our study indicated HA effectively suppressed RANKL-induced osteoclast formation and function. Thus, we propose that HA can be potentially used in the development of a novel drug for osteoclast-related bone diseases.


Subject(s)
Fusidic Acid/analogs & derivatives , NFATC Transcription Factors/drug effects , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , RANK Ligand/drug effects , Animals , Bone Resorption/metabolism , Fusidic Acid/pharmacology , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Osteogenesis/physiology , RANK Ligand/metabolism
14.
J Cell Physiol ; 234(7): 11951-11959, 2019 07.
Article in English | MEDLINE | ID: mdl-30552680

ABSTRACT

Osteoporosis, arthritis, Peget's disease, bone tumor, periprosthetic joint infection, and periprosthetic loosening have a common characteristic of osteolysis, which is characterized by the enhanced osteoclastic bone resorptive function. At present, the treatment target of these diseases is to interfere with osteoclastic formation and function. Scutellarein (Scu), a flavonoids compound, can inhibit the progress of tumor and inflammation. However, the role of Scu in inflammatory osteolysis isn't elucidated clearly. Our study showed that Scu inhibited bone destruction induced by LPS in vivo and OC morphology and function induced by RANKL in vitro. Mechanistic studies revealed that Scu suppressed osteoclastic marker gene expression by RANKL-induced, such as Ctsk9, Mmp9, Acp5, and Atp6v0d2. In addition, we found that the inhibition effects of osteoclastogenesis and bone resorption function of Scu were mediated via attenuating NF-κB and NFAT signaling pathways. In conclusion, the results showed that Scu may become a potential new drug for the treatment of inflammatory osteolysis.


Subject(s)
Apigenin/pharmacology , Bone Resorption/drug therapy , Cell Differentiation/drug effects , Osteoclasts/drug effects , Animals , Bone Diseases, Metabolic/metabolism , Bone Resorption/pathology , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice, Inbred C57BL , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Osteogenesis/drug effects
15.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(10): 1270-1276, 2018 Sep 30.
Article in Chinese | MEDLINE | ID: mdl-30377135

ABSTRACT

OBJECTIVE: To determine whether hyperglycemia activates NFAT2 in cultured podocytes to cause podocyte apoptosis and explore the role of NFAT2 in high glucose-induced podocyte apoptosis. METHODS: Immortalized mouse podocytes were cultured in the presence of normal (5.3 mmol/L) or high glucose (10, 20, 30, and 40 mmol/L) or pretreated with 11R-vivit (100 nmol/L) or cyclosporine A (500 nmol/L) before exposure to 20 mmol/L glucose for different durations (0.5-48 h). The activation of NFAT2 in the podocytes was detected using Western blotting and immunofluorescence assay. The role of NFAT2 in hyperglycemia-induced podocyte apoptosis was explored by observing the inhibition of NFAT2 activation by 11R-vivit using flow cytometry. Intracellular Ca2+ was monitored in high glucose-treated podocytes using Fluo-3/AM. The mRNA and protein expressions of the apoptosis gene Bax were detected using real time-qPCR and Western blotting. RESULTS: Exposure to high glucose in the medium time- and dose-dependently activated NFAT2 in cultured podocytes. Pretreatment with cyclosporine A or 11R- VIVIT completely blocked nuclear accumulation of NFAT2. Treatment with 11R- vivit also inhibited high glucoseinduced apoptosis in cultured podocytes. Exposure to high glucose obviously increased [Ca2 +]I in the podocytes to cause activation of calcineurin and the subsequent increment of nuclear accumulation of NFAT2 and Bax expression. CONCLUSIONS: High glucose-induced apoptosis in podocytes is mediated by calcineurin/NFAT2/Bax signaling pathway, which may serve as a potential target for therapeutic intervention.


Subject(s)
Apoptosis , Glucose/pharmacology , Hyperglycemia/metabolism , NFATC Transcription Factors/metabolism , Podocytes/drug effects , Animals , Calcineurin/metabolism , Cells, Cultured , Cyclosporine/pharmacology , Mice , NFATC Transcription Factors/drug effects , Oligopeptides , Podocytes/cytology , Podocytes/metabolism , Signal Transduction , bcl-2-Associated X Protein/metabolism
16.
Mem Inst Oswaldo Cruz ; 113(9): e180171, 2018 Jul 16.
Article in English | MEDLINE | ID: mdl-30020318

ABSTRACT

BACKGROUND: The anti-inflammatory and cardioprotective properties of curcumin (Cur), a natural polyphenolic flavonoid isolated from the rhizomes of Curcuma longa, are increasingly considered to have beneficial effects on the progression of Chagas heart disease, caused by the protozoan parasite Trypanosoma cruzi. OBJECTIVE: To evaluate the effects of oral therapy with Cur on T. cruzi-mediated cardiovasculopathy in acutely infected mice and analyse the in vitro response of parasite-infected human microvascular endothelial cells treated with this phytochemical. METHODS: Inflammation of heart vessels from Cur-treated and untreated infected mice were analysed by histology, with benznidazole (Bz) as the reference compound. Parasitaemia was monitored by the direct method. Capillary permeability was visualised by Evans-blue assay. Myocardial ET-1, IL-6, and TNF-α mRNA expressions were measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Microvascular endothelial HMEC-1 cells were infected in vitro with or without addition of Cur or Bz. Induction of the Ca2+/NFAT pathway was assessed by fluorometry, immunoblotting, and reporter assay. FINDINGS: Oral Cur therapy of recently infected mice reduced inflammatory cell infiltration of myocardial arteries without lowering parasite levels. Compared to that of the phosphate-buffered saline-receiving group, hearts from Cur-treated mice showed significantly decreased vessel inflammation scores (p < 0.001), vascular permeabilities (p < 0.001), and levels of IL-6/TNF-α (p < 0.01) and ET-1 (p < 0.05) mRNA. Moreover, Cur significantly (p < 0.05 for transcript; p < 0.01 for peptide) downregulated ET-1 secretion from infected HMEC-1 cells. Remarkably, Cur addition significantly (p < 0.05 at 27.0 µM) interfered with T. cruzi-dependent activation of the Ca2+/NFATc1 signalling pathway that promotes generation of inflammatory agents in HMEC-1 cells. CONCLUSIONS: Oral treatment with Cur dampens cardiovasculopathy in acute Chagas mice. Cur impairs the Ca2+/NFATc1-regulated release of ET-1 from T. cruzi-infected vascular endothelium. These findings identify new perspectives for exploring the potential of Cur-based interventions to ameliorate Chagas heart disease.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chagas Cardiomyopathy/drug therapy , Curcumin/pharmacology , Endothelin-1/drug effects , NFATC Transcription Factors/drug effects , Acute Disease , Animals , Blotting, Western , Capillary Permeability/drug effects , Cells, Cultured , Chagas Cardiomyopathy/metabolism , Chagas Cardiomyopathy/parasitology , Disease Progression , Endothelial Cells/drug effects , Endothelial Cells/parasitology , Endothelin-1/analysis , Endothelin-1/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/parasitology , Enzyme-Linked Immunosorbent Assay , Fluorescent Dyes , Interleukin-6/blood , Male , Mice, Inbred C57BL , NFATC Transcription Factors/analysis , NFATC Transcription Factors/metabolism , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Trypanosoma cruzi/drug effects , Tumor Necrosis Factor-alpha/blood
17.
Med Sci Monit ; 24: 2569-2577, 2018 Apr 26.
Article in English | MEDLINE | ID: mdl-29698379

ABSTRACT

BACKGROUND Neogambogic acid (NGA) is used in traditional Chinese medicine. The aim of this study was to investigate the effects of NGA on gene signaling pathways involved in osteoclastogenesis in mouse bone marrow-derived monocyte/macrophages (BMMs) and on bone resorption in vitro. MATERIAL AND METHODS Primary mouse BMMs were cultured with increasing concentrations of NGA. Real-time polymerase chain reaction was used to study the expression of mRNAs corresponding to gene products specific to receptor activator of NF-κB ligand (RANKL)-induced osteoclast differentiation, including tartrate-resistant acid phosphatase (TRAP), calcitonin receptor (CTR), cathepsin K (CTSK), and nuclear factor of activated T cells c1 (NFATc1). A cell counting kit-8 assay was used to evaluate cell proliferation. Western blotting and confocal immunofluorescence microscopy were used to investigate the signaling pathways. A bone resorption model was used to quantify bone resorption. RESULTS An NGA dose of ≤0.4 µg/ml had no significant effect on the proliferation of mouse BMMs in vitro (P>0.05); concentrations of between 0.1-0.4 µg/ml significantly inhibited RANKL-induced osteoclastogenesis (P<0.01) in a dose-dependent manner. Compared with the control group, NGA significantly reduced RANKL-induced bone resorption in vitro (P <0.01), and downregulated the expression of osteoclast-related mRNAs of TRAP, CTR, CTSK, and NFATc1. NGA suppressed the activation of JNK but not the p38 signaling pathway and significantly reduced NF-κB p65 phosphorylation and the nuclear transport of NF-κB molecules, which inhibited NFATc1 expression. CONCLUSIONS NGA suppressed RANKL-induced osteoclastogenesis by inhibiting the JNK and NF-κB pathways in mouse BMMs in vitro and reduced osteoclastic bone resorption.


Subject(s)
Macrophages/drug effects , Osteogenesis/drug effects , Xanthenes/pharmacology , Animals , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Cathepsin K/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , MAP Kinase Signaling System/drug effects , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Monocytes/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , NFATC Transcription Factors/drug effects , Osteoclasts/metabolism , RANK Ligand/metabolism , RANK Ligand/pharmacology , Receptors, Calcitonin/drug effects , Signal Transduction/drug effects , Tartrate-Resistant Acid Phosphatase/drug effects , Transcriptome/drug effects , Xanthenes/metabolism
18.
Endocrinology ; 159(9): 3143-3157, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29514186

ABSTRACT

Pharmacologic expansion of endogenous ß cells is a promising therapeutic strategy for diabetes. To elucidate the molecular pathways that control ß-cell growth we screened ∼2400 bioactive compounds for rat ß-cell replication-modulating activity. Numerous hit compounds impaired or promoted rat ß-cell replication, including CC-401, an advanced clinical candidate previously characterized as a c-Jun N-terminal kinase inhibitor. Surprisingly, CC-401 induced rodent (in vitro and in vivo) and human (in vitro) ß-cell replication via dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) 1A and 1B inhibition. In contrast to rat ß cells, which were broadly growth responsive to compound treatment, human ß-cell replication was only consistently induced by DYRK1A/B inhibitors. This effect was enhanced by simultaneous glycogen synthase kinase-3ß (GSK-3ß) or activin A receptor type II-like kinase/transforming growth factor-ß (ALK5/TGF-ß) inhibition. Prior work emphasized DYRK1A/B inhibition-dependent activation of nuclear factor of activated T cells (NFAT) as the primary mechanism of human ß-cell-replication induction. However, inhibition of NFAT activity had limited effect on CC-401-induced ß-cell replication. Consequently, we investigated additional effects of CC-401-dependent DYRK1A/B inhibition. Indeed, CC-401 inhibited DYRK1A-dependent phosphorylation/stabilization of the ß-cell-replication inhibitor p27Kip1. Additionally, CC-401 increased expression of numerous replication-promoting genes normally suppressed by the dimerization partner, RB-like, E2F and multivulval class B (DREAM) complex, which depends upon DYRK1A/B activity for integrity, including MYBL2 and FOXM1. In summary, we present a compendium of compounds as a valuable resource for manipulating the signaling pathways that control ß-cell replication and leverage a DYRK1A/B inhibitor (CC-401) to expand our understanding of the molecular pathways that control ß-cell growth.


Subject(s)
Cell Proliferation/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Insulin-Secreting Cells/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazolones/pharmacology , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Adult , Animals , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p27/drug effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Forkhead Box Protein M1/drug effects , Forkhead Box Protein M1/metabolism , Humans , In Vitro Techniques , Kv Channel-Interacting Proteins/drug effects , Kv Channel-Interacting Proteins/metabolism , Male , Middle Aged , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/metabolism , Rats , Repressor Proteins/drug effects , Repressor Proteins/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism , Dyrk Kinases
19.
Brain Behav Immun ; 68: 158-168, 2018 02.
Article in English | MEDLINE | ID: mdl-29056557

ABSTRACT

Antitubulin chemotherapeutics agents, such as paclitaxel, are effective chemotherapy drugs for cancer treatment. However, painful neuropathy is a major adverse effect limiting the wider application of chemotherapeutics. In this study, we found that A-kinase anchor protein 150 (AKAP150) was significantly upregulated after paclitaxel injection. Inhibition of AKAP150 via siRNA or AKAP150flox/flox in rodents alleviated the pain behavior induced by paclitaxel, and partly restored the decreased calcineurin (CN) phosphatase activity after paclitaxel treatment. Paclitaxel decreased the expression of anti-inflammatory cytokine interleukin-4 (IL-4), and intrathecal injections of IL-4 effectively alleviated paclitaxel-induced hypersensitivity and the frequency of dorsal root ganglion (DRG) neurons action potential. The decreased CN enzyme activity, resulted in reduced protein expression of nuclear factor of activated T cells 2 (NFAT2) in cell nuclei. Chromatin immunoprecipitation showed that, NFAT2 binds to the IL-4 gene promoter regulating the protein expression of IL-4. Overexpression of NFAT2 by intrathecal injection of the AAV5-NFAT2-GFP virus alleviated the pain behavior induced by paclitaxel via increasing the expression of IL-4. Knocked down AKAP150 by siRNA or AAV5-Cre-GFP partly restored the expression of IL-4 in DRG. Our results indicated that regulation of IL-4 via the CN/NFAT2 pathway mediated by AKAP150 could be a pivotal treatment target for paclitaxel-induced neuropathic pain and or other neuropsychiatric disorders.


Subject(s)
A Kinase Anchor Proteins/metabolism , Neuralgia/metabolism , A Kinase Anchor Proteins/physiology , Animals , Calcineurin/drug effects , Calcineurin/metabolism , Cytokines/metabolism , Down-Regulation , Ganglia, Spinal/metabolism , Hyperalgesia/metabolism , Injections, Spinal , Interleukin-4/metabolism , Male , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/metabolism , Neuralgia/physiopathology , Paclitaxel/adverse effects , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Up-Regulation
20.
Nat Med ; 22(10): 1170-1179, 2016 10.
Article in English | MEDLINE | ID: mdl-27595322

ABSTRACT

The use of selective serotonin-reuptake inhibitors (SSRIs) has been associated with an increased risk of bone fracture, raising concerns about their increasingly broader usage. This deleterious effect is poorly understood, and thus strategies to avoid this side effect remain elusive. We show here that fluoxetine (Flx), one of the most-prescribed SSRIs, acts on bone remodeling through two distinct mechanisms. Peripherally, Flx has anti-resorptive properties, directly impairing osteoclast differentiation and function through a serotonin-reuptake-independent mechanism that is dependent on intracellular Ca2+ levels and the transcription factor Nfatc1. With time, however, Flx also triggers a brain-serotonin-dependent rise in sympathetic output that increases bone resorption sufficiently to counteract its local anti-resorptive effect, thus leading to a net effect of impaired bone formation and bone loss. Accordingly, neutralizing this second mode of action through co-treatment with the ß-blocker propranolol, while leaving the peripheral effect intact, prevents Flx-induced bone loss in mice. Hence, this study identifies a dual mode of action of SSRIs on bone remodeling and suggests a therapeutic strategy to block the deleterious effect on bone homeostasis from their chronic use.


Subject(s)
Bone Remodeling/drug effects , Bone and Bones/drug effects , Fluoxetine/pharmacology , Osteogenesis/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Sympathetic Nervous System/drug effects , Adrenergic beta-Antagonists/pharmacology , Animals , Bone Resorption , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Calcium/metabolism , Gene Expression/drug effects , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Knockout , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/metabolism , Optical Imaging , Propranolol/pharmacology , Serotonin Plasma Membrane Transport Proteins/genetics , Tryptophan Hydroxylase/genetics , X-Ray Microtomography
SELECTION OF CITATIONS
SEARCH DETAIL
...