Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Cell Death Discov ; 10(1): 86, 2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38368392

ABSTRACT

The key target for treating inflammatory osteolysis is osteoclasts. In an inflammatory environment, osteoclast differentiation increases, and bone resorption is enhanced. Periplogenin (Ppg) is a traditional Chinese medicine. It has anti-inflammatory and antitumor effects, but its impact on inflammatory osteolysis is unknown. This study found that Ppg prevented LPS-induced skull osteolysis by inhibiting the expression of inflammatory cytokines and osteoclast production. In vitro, Ppg blocked the RANKL-induced generation of osteoclasts, the development of pseudopodia bands, and bone resorption. Ppg also attenuated the expression of NFATc1, c-Fos, CTSK, and Atp6v0d2 proteins by inhibiting the NFATc1 signaling pathway. In addition, Ppg inhibited the expression of osteoclast-specific genes, including NFATc1, c-Fos, CTSK, Atp6v0d2, and Mmp9. Moreover, Ppg also inhibited NF-κB and MAPK pathways. In vivo, Ppg reduced the number of osteoclasts on the surface of the bone and suppressed LPS-induced osteolysis of the skull. These outcomes suggest that Ppg can serve as a new alternative therapy for treating inflammatory osteolysis by inhibiting inflammation and osteoclasts.

2.
Biomed Pharmacother ; 171: 116166, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38244329

ABSTRACT

Osteoporosis is a systemic disease characterized by an imbalance in bone homeostasis, where osteoblasts fail to fully compensate for the bone resorption induced by osteoclasts. Corylifol A, a flavonoid extracted from Fructus psoraleae, has been identified as a potential treatment for this condition. Predictions from network pharmacology and molecular docking studies suggest that Corylifol A exhibits strong binding affinity with NFATc1, Nrf2, PI3K, and AKT1. Empirical evidence from in vivo experiments indicates that Corylifol A significantly mitigates systemic bone loss induced by ovariectomy by suppressing both the generation and activation of osteoclasts. In vitro studies further showed that Corylifol A inhibited the activation of PI3K-AKT and MAPK pathways and calcium channels induced by RANKL in a time gradient manner, and specifically inhibited the phosphorylation of PI3K, AKT, GSK3 ß, ERK, CaMKII, CaMKIV, and Calmodulin. It also diminishes ROS production through Nrf2 activation, leading to a decrease in the expression of key regulators such as NFATcl, C-Fos, Acp5, Mmp9, and CTSK that are involved in osteoclastogenesis. Notably, our RNA-seq analysis suggests that Corylifol A primarily impacts mitochondrial energy metabolism by suppressing oxidative phosphorylation. Collectively, these findings demonstrate that Corylifol A is a novel inhibitor of osteoclastogenesis, offering potential therapeutic applications for diseases associated with excessive bone resorption.


Subject(s)
Bone Resorption , Flavones , Osteogenesis , Female , Humans , Animals , Mice , Reactive Oxygen Species/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Glycogen Synthase Kinase 3/metabolism , Molecular Docking Simulation , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Osteoclasts/metabolism , Bone Resorption/metabolism , Ovariectomy , RANK Ligand/metabolism , NFATC Transcription Factors/metabolism , Mice, Inbred C57BL , Cell Differentiation
3.
Cell Prolif ; 57(1): e13535, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37551727

ABSTRACT

Periodontal disease and arthroplasty prosthesis loosening and destabilization are both associated with osteolysis, which is predominantly caused by abnormal bone resorption triggered by pro-inflammatory cytokines. Osteoclasts (OCs) are critical players in the process. Concerns regarding the long-term efficacy and side effects of current frontline therapies, however, remain. Alternative therapies are still required. The aim of this work was to investigate the involvement of Tenacissoside H (TDH) in RANKL-mediated OC differentiation, as well as inflammatory osteolysis and associated processes. In vitro, bone marrow-derived macrophages (BMMs) cultured with RANKL and M-CSF were used to detect TDH in the differentiation and function of OCs. Real-time quantitative PCR was used to measure the expression of specific genes and inflammatory factors in OCs. Western blot was used to identify NFATc1, IKK, NF-κB, MAPK pathway, and oxidative stress-related components. Finally, an LPS-mediated calvarial osteolysis mouse model was employed to explore TDH's role in inflammatory osteolysis. The results showed that in vivo TDH inhibited the differentiation and resorption functions of OCs and down-regulated the transcription of osteoclast-specific genes, as well as Il-1ß, Il-6 and Tnf-α. In addition, TDH inhibited the IKK and NF-κB signalling pathways and down-regulated the level of ROS. In vivo studies revealed that TDH improves the bone loss caused by LPS. TDH may be a new candidate or treatment for osteoclast-associated inflammatory osteolytic disease.


Subject(s)
Osteolysis , Animals , Mice , Osteolysis/chemically induced , NF-kappa B/metabolism , Lipopolysaccharides/pharmacology , Reactive Oxygen Species/metabolism , Osteoclasts/metabolism , RANK Ligand/metabolism , Cell Differentiation , NFATC Transcription Factors/metabolism
4.
Biomed Pharmacother ; 167: 115605, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37801901

ABSTRACT

The second most common cancer among men is prostate cancer, which is also the fifth leading reason for male cancer deaths worldwide. Bone metastases are the main factor affecting the prognosis of prostate cancer. Consequently, antitumor and anti-prostate cancer-induced bone destruction medicines are urgently needed. We previously discovered that aminooxyacetic acid hemihydrochloride (AOAA) suppressed bone resorption and osteoclast growth by decreasing adenosine triphosphate (ATP) production and limiting oxidative phosphorylation (OXPHOS). Here, we evaluated the impacts of AOAA on prostate cancer RM-1 cells in vitro. It's found that AOAA significantly inhibited cell proliferation, migration, and invasiveness, decreased ATP levels, increased ROS, halted the cell cycle phase, and triggered apoptosis. AOAA also decreased mitochondrial membrane potential and the ability to uptake glucose, suggesting that the antitumor effects of AOAA were expressed through the inhibition of OXPHOS and glycolysis. Furthermore, we assessed the effects of AOAA in vivo using a prostate cancer-induced bone osteolysis mice model. AOAA also delayed tumor growth and bone destruction in vivo. On the whole, our findings imply that AOAA may potentially have therapeutic effects on prostate cancer and prostate cancer-induced osteolysis.


Subject(s)
Osteolysis , Prostatic Neoplasms , Mice , Animals , Male , Humans , Aminooxyacetic Acid/pharmacology , Adenosine Triphosphate/metabolism , Energy Metabolism , Prostatic Neoplasms/drug therapy , Cell Cycle , Cell Line, Tumor
5.
iScience ; 26(10): 107760, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37720109

ABSTRACT

Osteoporosis is a prevalent systemic metabolic disease in modern society, in which patients often suffer from bone loss due to over-activation of osteoclasts. Currently, amelioration of bone loss through modulation of osteoclast activity is a major therapeutic strategy. Ataxia telangiectasia mutated (ATM) inhibitor CGK733 (CG) was reported to have a sensitizing impact in treating malignancies. However, its effect on osteoporosis remains unclear. In this study, we investigated the effects of CG on osteoclast differentiation and function, as well as the therapeutic effects of CG on osteoporosis. Our study found that CG inhibits osteoclast differentiation and function. We further found that CG inhibits the activation of NFATc1 and ultimately osteoclast formation by inhibiting RANKL-mediated Ca2+ oscillation and the NF-κB/MAPK signaling pathway. Next, we constructed an ovariectomized mouse model and demonstrated that CG improved bone loss in ovariectomized mice. Therefore, CG may be a potential drug for the prevention and treatment of osteoporosis.

6.
Biomed Pharmacother ; 165: 114898, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37352699

ABSTRACT

Osteoporosis, as a severe public health problem worldwide, causes systemic damage to bone mass, strength, and microstructure with an increased propensity for fragility fractures. Given the inherent adverse effects associated with long-term use of current prescription medications for osteoporosis treatment, identifying natural alternatives to existing treatment methods is imperative. Pteryxin (PTX), a natural coumarin, is isolated from the Peucedanum species belonging to the family Apiaceae. PTX has been reported to have antioxidant, anti-inflammatory and anti-obesity properties. However, its effect on osteoporosis has not been clarified. Our study confirmed that PTX could attenuate the formation of osteoclasts and bone resorption on a dose-dependent basis in vitro. Consistently, in vivo ovariectomy (OVX)-induced osteoporosis models simulating the physiological characteristics of postmenopausal women showed that PTX could partially reverse the bone loss caused by OVX. Further study of its mechanism revealed that PTX might block the MAPK and Ca2+-calcineurin-NFATc1 signaling pathways by decreasing the reactive oxygen species (ROS) level in osteoclasts to dampen the expression of critical transcriptional NFATc1 and downstream osteoclast-specific genes. Overall, PTX may present a new or alternative treatment option for osteoporosis.


Subject(s)
Osteogenesis , Osteoporosis , Female , Humans , Reactive Oxygen Species/metabolism , Osteoclasts , Signal Transduction , Coumarins/pharmacology , Coumarins/therapeutic use , Osteoporosis/drug therapy , Osteoporosis/prevention & control , Osteoporosis/etiology , Ovariectomy/adverse effects , RANK Ligand/metabolism , Cell Differentiation , NF-kappa B/metabolism
7.
Int J Mol Med ; 51(5)2023 May.
Article in English | MEDLINE | ID: mdl-37052260

ABSTRACT

Postmenopausal osteoporosis is a systemic metabolic disease that chronically endangers public health and is typically characterized by low bone mineral density and marked bone fragility. The excessive bone resorption activity of osteoclasts is a major factor in the pathogenesis of osteoporosis; therefore, strategies aimed at inhibiting osteoclast activity may prevent bone decline and attenuate the process of osteoporosis. Casticin (Cas), a natural compound, has anti­inflammatory and antitumor properties. However, the role of Cas in bone metabolism remains largely unclear. The present study found that the receptor activator of nuclear factor­κΒ (NF­κB) ligand­induced osteoclast activation and differentiation were inhibited by Cas. Tartrate­resistant acid phosphatase staining revealed that Cas inhibited osteoclast differentiation, and bone resorption pit assays demonstrated that Cas affected the function of osteoclasts. Cas significantly reduced the expression of osteoclast­specific genes and related proteins, such as nuclear factor of activated T cells, cytoplasmic 1 and c­Fos at the mRNA and protein level in a concentration­dependent manner. Cas inhibited osteoclast formation by blocking the AKT/ERK and NF­κB signaling pathways, according to the intracellular signaling analysis. The microcomputed tomography and tissue staining of tibiae from ovariectomized mice revealed that Cas prevented the bone loss induced by estrogen deficiency and reduced osteoclast activity in vivo. Collectively, these findings indicated that Cas may be used to prevent osteoporosis.


Subject(s)
Bone Diseases, Metabolic , Bone Resorption , Osteoporosis , Female , Animals , Mice , Humans , Osteogenesis , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , X-Ray Microtomography/adverse effects , Signal Transduction , Osteoclasts/metabolism , Bone Resorption/drug therapy , Bone Resorption/etiology , Bone Resorption/prevention & control , Cell Differentiation , Osteoporosis/drug therapy , Osteoporosis/etiology , Osteoporosis/prevention & control , Bone Diseases, Metabolic/complications , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Ovariectomy/adverse effects , RANK Ligand/metabolism
8.
Biochem Pharmacol ; 210: 115463, 2023 04.
Article in English | MEDLINE | ID: mdl-36849060

ABSTRACT

BACKGROUND: Osteoporosis, a systemic metabolic bone disease, is often caused by the disruption of dynamic equilibrium between osteoclasts and osteoblasts. Overactive bone resorption, in which osteoclasts play a major role, is one of the most common and major causes of osteoporosis. Less costly and more effective drug treatments for this disease are needed. Based on the combination of molecular docking techniques and in vitro cell assays, this study aimed to explore the mechanism by which Isoliensinine (ILS) protects the bone loss by inhibiting osteoclast differentiation. METHODS: A virtual docking model based on molecular docking technology was used to investigate the interactions between ILS and the Receptor Activator of Nuclear Kappa-B (RANK)/Receptor Activator of Nuclear Kappa-B Ligand (RANKL).In this study, we determined the effective dose of action of ILS to inhibit osteoclast differentiation in vitro and, using bone resorption experiments, RT-CPR and Western Blot investigated the effects of ILS on bone resorption function and normal expression of osteoclast-associated genes and proteins, and validated potential mechanistic pathways. In vivo experiments revealed that ILS could inhibit bone loss through Micro-CT results. Finally, the molecular interaction between ILS and RANK/RANKL was investigated using biomolecular interaction experiments to verify the correctness and accuracy of the computational results. RESULTS: ILS binds to RANK and RANKL proteins, respectively, through virtual molecular docking. The Surface Plasmon Resonance (SPR) experiment results revealed that phosphorylated JNK, ERK, P38, and P65 expression was significantly downregulated when ILS were targeted to inhibit RANKL/RANK binding. At the same time, the expression of IKB-a was significantly increased under the stimulation of ILS, which rescued the degradation of IKB-a. ILS can significantly inhibit the levels of Reactive Oxygen Species (ROS) and Ca2 + concentration in vitro. Finally, the results of Micro-CT showed that ILS can significantly inhibit bone loss in vivo, indicating that ILS has a potential role in the treatment of osteoporosis. CONCLUSION: ILS inhibits osteoclast differentiation and bone loss by preventing the normal binding of RANKL/RANK, affecting downstream signaling pathways, including MAPK.NF-KB, ROS, Ca2+, genes, and proteins.


Subject(s)
Bone Density Conservation Agents , Bone Diseases, Metabolic , Bone Resorption , Osteoporosis , Humans , Reactive Oxygen Species/metabolism , Molecular Docking Simulation , Cell Differentiation , Osteoclasts , Bone Resorption/drug therapy , Bone Resorption/prevention & control , Bone Resorption/metabolism , NF-kappa B/metabolism , Bone Density Conservation Agents/pharmacology , Bone Diseases, Metabolic/metabolism , Osteoporosis/metabolism , RANK Ligand/pharmacology , Osteogenesis
9.
Biomed Pharmacother ; 160: 114347, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36746095

ABSTRACT

The formation of osteoclasts and their hyperactive bone resorption are related to the aggregation of intracellular reactive oxygen species (ROS). Flavonoids, derived from plant active ingredients, can alleviate the symptoms of osteoporosis (OP). Isosinensetin (Iss) is a flavonoid with antioxidant effects obtained mainly from citrus fruits, and its effect on osteoclastogenesis has not been reported. In this study, we investigated the antioxidant activity of Iss on osteoclast differentiation and function, as well as the therapeutic impact of Iss on OP. We found that Iss inhibited osteoclastogenesis and suppressed the bone resorption function of osteoclasts. Additionally, Iss reduced receptor activator of nuclear factor-κB ligand (RANKL)-induced intracellular ROS. Using quantitative real-time polymerase chain reaction and western blot, we further found that Iss inhibited osteoclast-specific genes and related proteins, while promoting the expression of antioxidant enzyme-related genes and proteins. Mechanistically, Iss reduces intracellular ROS by activating nuclear factor-erythroid 2-related factor 2 (Nrf2) and its related antioxidant enzymes and inhibits the downstream nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways of ROS, which in turn inhibits nuclear factor of activated T cells 1 (NFATc1), and ultimately inhibits osteoclastogenesis. In vivo, by micro-computed tomography (Micro-CT) assay and histological analyses, we found that Iss could reduce bone loss in ovariectomized (OVX) mice. Therefore, Iss has the potential as an OP preventative and therapeutic drug option.


Subject(s)
Bone Resorption , Osteoporosis , Animals , Mice , NF-kappa B/metabolism , Reactive Oxygen Species/pharmacology , Antioxidants/pharmacology , Antioxidants/metabolism , X-Ray Microtomography , Cell Differentiation , Osteoclasts , Bone Resorption/metabolism , MAP Kinase Signaling System , Osteogenesis , Osteoporosis/drug therapy , Osteoporosis/pathology , Estrogens/pharmacology , RANK Ligand/metabolism
10.
Biomed Pharmacother ; 159: 114101, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36640671

ABSTRACT

4-Methylcatechol (4-MC) is an agonist of various neurotrophic factors, which can upregulate the expression of Heme oxygenase 1 (HO-1) protein by activating nuclear factor erythroid 2-related factor 2 (Nrf2), thereby inhibiting oxidative stress-induced neural stem cell death. During RANKL-stimulated osteoclast differentiation, intracellular reactive oxygen species (ROS) levels were increased. Nonetheless, the effect of 4-MC on osteoclast formation and bone resorption function has not been researched. In this study, we investigated the effect of HO-1 upregulation by 4-MC on RANKL-induced osteoclastogenesis and explored the molecular mechanism of HO-1 upregulation by 4-MC. We found that the small molecule compound 4-MC could bind to Keap1 amino acid residue of glycine GLY 367, isoleucine ILE 559 and valine VAL 606, with a predicted binding energy of -4.99 kcal/mol. 4-MC was found to inhibit osteoclast differentiation in vitro by activating Nrf2 to scavenge ROS, inhibiting NF-κB phosphorylation, and alleviating osteoporosis in ovariectomized (OVX) mice. Taken together, 4-MC reduces ROS by inhibiting Keap1, thereby preventing OVX-induced bone loss.


Subject(s)
NF-E2-Related Factor 2 , Osteogenesis , Animals , Mice , Reactive Oxygen Species/metabolism , NF-E2-Related Factor 2/metabolism , Molecular Docking Simulation , Kelch-Like ECH-Associated Protein 1/metabolism , Osteoclasts , NF-kappa B/metabolism , RANK Ligand/metabolism
11.
Int Immunopharmacol ; 113(Pt A): 109370, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36327872

ABSTRACT

As a common disease in modern society, osteoporosis is caused by osteoclast hyperactivation, leading to enhanced bone resorption. Reactive oxygen species (ROS) metobolism and nuclear factor-activated T cells 1 (NFATc1) activities are two crucial processes during osteoclastogenesis. AZD1390 (AZD), an inhibitor of ataxia telangiectasia mutated (ATM), has been reported for antitumor effects, but little is known about how it plays a function in metabolic bone disease. Here, we found that AZD inhibitsthe generation, function and ROS-scavenging enzyme activity of mature osteoclast induced by RANKL stimulation, in a dose-dependent manner.Mechanistic analysis shows thatAZD affects osteoclast function and differentiation by inhibiting RANKL-induced NFATc1 signaling pathway and by increasing ROS-scavenging enzymes production in oxidative stress pathways. Preclinical studies have shown that AZD protects against bone loss in an ovariectomy (OVX) mouse model. Finally, our data confirm that AZD may prevent OVX-induced bone loss by abrogating RANKL-induced AKT/GSK3ß/NFATc1 signaling pathways, and by promoting the expression of ROS scavenging enzymes in oxidative stress pathways.Collectively, our research shows that AZD has the potential as a new therapeutic agent for osteoporosis.


Subject(s)
Bone Resorption , Osteoporosis , Mice , Animals , Female , Osteoclasts , Reactive Oxygen Species/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , NFI Transcription Factors/metabolism , NFI Transcription Factors/pharmacology , T-Lymphocytes/metabolism , RANK Ligand/metabolism , Bone Resorption/prevention & control , Osteoporosis/metabolism , Osteogenesis , Cell Differentiation , NFATC Transcription Factors/metabolism
12.
Front Pharmacol ; 13: 896108, 2022.
Article in English | MEDLINE | ID: mdl-36110547

ABSTRACT

Osteoporosis affects around 200 million people globally, with menopausal women accounting for the bulk of cases. In the occurrence and development of osteoporosis, a key role is played by osteoclasts. Excessive osteoclast-mediated bone resorption activity reduces bone mass and increases bone fragility, resulting in osteoporosis. Thus, considerable demand exists for designing effective osteoporosis treatments based on targeting osteoclasts. Eltanexor (Elt; KPT-8602) is a selective nuclear-export inhibitor that covalently binds to and blocks the function of the nuclear-export protein exportin-1 (XPO1), which controls the nucleus-to-cytoplasm transfer of certain critical proteins related to growth regulation and tumor suppression, such as p53, IκBα [nuclear factor-κB (NF-κB) inhibitor α] and FOXO1; among these proteins, IκBα, a critical component of the NF-κB signaling pathway that primarily governs NF-κB activation and transcription. How Elt treatment affects osteoclasts remains poorly elucidated. Elt inhibited the growth and activity of RANKL-induced osteoclasts in vitro in a dose-dependent manner, and Elt exerted no cell-killing effect within the effective inhibitory concentration. Mechanistically, Elt was found to trap IκBα in the nucleus and thus protect IκBα from proteasome degradation, which resulted in the blocking of the translocation of IκBα and NF-κB p65 and the consequent inhibition of NF-κB activity. The suppression of NF-κB activity, in turn, inhibited the activity of two transcription factors (NFATc1 and c-Fos) essential for osteoclast formation and led to the downregulation of genes and proteins related to bone resorption. Our study thus provides a newly identified mechanism for targeting in the treatment of osteoporosis.

13.
Eur J Pharmacol ; 923: 174908, 2022 May 15.
Article in English | MEDLINE | ID: mdl-35405113

ABSTRACT

Osteoporosis is an osteolytic disease with a disrupted balance between the resorption and formation of bone as well as bone microstructure degeneration, leading to bone loss and increased fracture risk, which greatly affects patients' quality of life. Currently, inhibition of osteoclast bone resorption remains the mainstream treatment for osteoporosis. Onc201, a new compound, induces the gene expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and has an efficient anticancer effect in clinical trials. However, its effects on osteolytic disease and the mechanism of action are unclear. We examined the effect of Onc201 on nuclear factor κB ligand-receptor activator (RANKL)-induced osteoclasts via Cell Counting Kit-8, bone resorption assay, luciferase reporter assay, immunofluorescence staining, calcium ion intensity assay and employed an ovariectomy model to investigate the effect of Onc201 on osteoporosis in the mice. Results showed that Onc201 inhibited the function and formation of osteoclasts induced by RANKL in a manner that was dependent on time and concentration, and did not cause cytotoxicity. Mechanistically, Onc201 inhibited osteoclast-relevant genes and NFATc1 expression, the main transcriptional regulatory factor of the formation of osteoclasts induced by RANKL; meanwhile, downregulating the expressions of the osteoclast cytoskeleton key signal molecules integrin αvß3, focal adhesion kinase (FAK), c-Src, and spleen-associated tyrosine kinase (SYK). In addition, Onc201 had a protective effect on the mouse model of bone loss caused by ovariectomy-induced estrogen deficiency, which is consistent with the in vitro results. Our findings suggest that the new small-molecular compound Onc201 has the potential to prevent osteoclast-related osteolytic diseases.


Subject(s)
Bone Resorption , Osteoporosis , Animals , Bone Resorption/metabolism , Bone Resorption/prevention & control , Cell Differentiation , Female , Humans , Integrins/metabolism , Ligands , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Osteoclasts , Osteogenesis , Osteoporosis/etiology , Osteoporosis/metabolism , Osteoporosis/prevention & control , Ovariectomy/adverse effects , Quality of Life , RANK Ligand/metabolism , RANK Ligand/pharmacology , Receptor Activator of Nuclear Factor-kappa B , Signal Transduction
14.
Mol Med Rep ; 19(3): 1728-1738, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30628641

ABSTRACT

The present study aimed to investigate the potent inhibitory effects and possible biochemical basis of the novel phosphodiesterase 4 (PDE4) inhibitor ciclamilast, which is a derivative of piclamilast (RP 73401), on PDE4 and allergic inflammation. Ciclamilast was orally administered to allergic rats, their lungs and bronchoalveolar lavage fluid (BALF) were harvested, and their levels of inflammation and goblet cell hyperplasia, particularly cAMP­PDE activity, and expression and distribution of PDE4 subtypes were determined. The results suggested that oral administration of ciclamilast significantly reduced the total leukocyte number and eosinophil number in BALF and suppressed lung histology changes, including the infiltration of inflammatory cells into the perivascular and peribronchial spaces, structural changes and goblet cell hyperplasia. For eosinophil infiltration, ciclamilast exhibited improved selectivity compared with piclamilast. Furthermore, ciclamilast significantly inhibited the upregulated activity of cAMP­PDE and showed improved selective inhibition of the protein expression of PDE4B than piclamilast in a dose­dependent manner. The mRNA expression of PDE4D was significantly increased in allergic rats, but PDE4B was not. PDE4B was mainly distributed in the cytoplasm, whereas PDE4D was mainly distributed in the cell membrane. The improved anti­inflammatory activity of ciclamilast compared with piclamilast may be due to its higher level of inhibition of the activity, mRNA and protein expression of PDE4, particularly its effect on PDE4B.


Subject(s)
Benzamides/administration & dosage , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Hypersensitivity/drug therapy , Inflammation/drug therapy , Pyridines/administration & dosage , Animals , Blood Cell Count , Bronchoalveolar Lavage Fluid , Cyclic AMP/genetics , Disease Models, Animal , Eosinophils/drug effects , Gene Expression Regulation/drug effects , Goblet Cells/drug effects , Goblet Cells/pathology , Humans , Hyperplasia/drug therapy , Hyperplasia/pathology , Hypersensitivity/genetics , Hypersensitivity/pathology , Inflammation/genetics , Inflammation/pathology , Leukocytes/drug effects , Lung/drug effects , Lung/pathology , Phosphodiesterase Inhibitors/administration & dosage , Rats
15.
Front Pharmacol ; 8: 212, 2017.
Article in English | MEDLINE | ID: mdl-28473770

ABSTRACT

Objective: A generic vaginal progesterone gel has recently been developed in China. Little is known about its pharmacokinetic properties in Chinese subjects. The purpose of our study was to investigate the pharmacokinetics of three forms of vaginal progesterone gel (test formulations at 4 and 8% strength vs. a reference formulation: Crinone 8%) in Chinese healthy post-menopausal women. Methods: This study consisted of two parts study. The part 1 study was a single-center, open-label, 3-period study. Twelve healthy post-menopausal women were to evaluate the safety and pharmacokinetics of 45 mg vaginal progesterone gel (Test 4%) following single dose and multiple doses administered once every other day (q.o.d.) for six times or once daily (q.d.) for 6 days. The part 2 study was a randomized, open-label, 3-stage crossover study. Twelve post-menopausal women received 90 mg vaginal progesterone gel (Test 8%) or 90 mg Crinone (Reference 8%) following single dose and multiple doses (q.o.d. or q.d.). Plasma concentrations of progesterone were measured up to 72 h by using a validated liquid chromatography tandem-mass spectrometry method. The primary pharmacokinetic parameters, maximum plasma concentration (Cmax) and area under the plasma concentration-time curve (AUC) from time zero to last measurable concentration (AUC0-t) and extrapolated to infinity (AUC0-∞) were compared by an analysis of variance using log-transformed data. Results: Totally 24 subjects were enrolled in and completed the study. Following single dose, The geometric mean Cmax values for Test 4%, Test 8%, and Crinone 8% were 6.35, 10.34, 10.45 ng/mL, and their geometric mean AUC0-t (AUC0-∞) were 113.73 (118.00), 169.39 (173.98), and 190.07 (201.13) ng⋅h/mL, respectively. The mean T1/2 values of progesterone were 11.00, 10.92, and 11.40 h, respectively. For 8% test formulation vs. reference, the 90% CIs of the least squares mean test/reference ratios of Cmax, AUC0-t, and AUC0-∞ were 78.32-124.85%, 54.31-146.24%, and 53.64-137.75, respectively. The most frequent adverse events were increased vaginal secretions, most of which were of mild intensity and considered related to treatment. Conclusion: Results with single and multiple doses of vaginal progesterone gel suggest similar pharmacokinetics properties between test formulations and Crinone 8%. Overall, vaginal progesterone gel was well tolerated.

16.
Oncologist ; 21(11): 1294-1295d, 2016 11.
Article in English | MEDLINE | ID: mdl-27789778

ABSTRACT

LESSONS LEARNED: This phase I study evaluated the maximum tolerated dose, dose-limiting toxicities, safety, pharmacokinetics, and efficacy of icotinib with a starting dose of 250 mg in pretreated, advanced non-small cell lung cancer patients. We observed a maximum tolerated dose of 500 mg with a favorable pharmacokinetics profile and antitumor activity.These findings provide clinicians with evidence for application of higher-dose icotinib. BACKGROUND: Icotinib, an oral epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, has shown favorable tolerability and antitumor activity at 100-200 mg in previous studies without reaching the maximum tolerated dose (MTD). In July 2011, icotinib was approved by the China Food and Drug Administration at a dose of 125 mg three times daily for the treatment of patients with locally advanced or metastatic non-small cell lung cancer (NSCLC) after failure of at least one platinum-based chemotherapy regimen. This study investigated the MTD, tolerability, and pharmacokinetics of higher-dose icotinib in patients with advanced NSCLC. METHODS: Twenty-six patients with advanced NSCLC were treated at doses of 250-625 mg three times daily The EGFR mutation test was not mandatory in this study. RESULTS: Twenty-four (92.3%) of 26 patients experienced at least one adverse event (AE); rash (61.5%), diarrhea (23.1%), and oral ulceration (11.5%) were most frequent AEs. Dose-limiting toxicities were seen in 2 of 6 patients in the 625-mg group, and the MTD was established at 500 mg. Icotinib was rapidly absorbed and eliminated. The amount of time that the drug was present at the maximum concentration in serum (Tmax) ranged from 1 to 3 hours (1.5-4 hours) after multiple doses. The t1/2 was similar after single- and multiple-dose administration (7.11 and 6.39 hours, respectively). A nonlinear relationship was observed between dose and drug exposure. Responses were seen in 6 (23.1%) patients, and 8 (30.8%) patients had stable disease. CONCLUSION: This study demonstrated that higher-dose icotinib was well-tolerated, with a MTD of 500 mg. Favorable antitumor activity and pharmacokinetic profile were observed in patients with heavily pretreated, advanced NSCLC.


Subject(s)
Antineoplastic Agents/adverse effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Crown Ethers/adverse effects , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Quinazolines/adverse effects , Adult , Aged , Crown Ethers/pharmacokinetics , ErbB Receptors/genetics , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Mutation , Quinazolines/pharmacokinetics
17.
J Chromatogr Sci ; 53(6): 1013-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25404561

ABSTRACT

An economical, reproducible and automated online solid-phase extraction coupled with liquid chromatography-tandem mass spectrometry method was developed to quantify methylprednisolone in human plasma. The method was validated in terms of selectivity, precision/accuracy, process efficiency, stability, cartridge reproducibility and carryover studies. Sample pretreatment was performed by protein precipitation and elimination using methanol followed by water dilution. Then, the mixture was passed onto the HySphere C8 EC-SE online solid-phase extraction cartridge followed by the separation of the analytes on an Agilent Eclipse XDB column. Electrospray ionization in positive ion mode and multiple reaction monitoring were used to monitor the ion transitions at m/z 375.4/160.8 for methylprednisolone, and m/z 361.2/147.0 for prednisolone. The calibration curve ranged from 5.25 to 525 ng/mL. Meanwhile both the intra-day and inter-day precision values (relative standard deviation) were within 4.45%. The method which turns out to be less laborious, faster and lower consumable cost per sample has already been successfully applied to a pharmacokinetic study in which the oral administration of 16 mg methylprednisolone was conducted in Chinese volunteers.


Subject(s)
Chromatography, Liquid/methods , Methylprednisolone/blood , Solid Phase Extraction/methods , Tandem Mass Spectrometry/methods , Humans , Linear Models , Methylprednisolone/chemistry , Methylprednisolone/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Solid Phase Extraction/economics , Solid Phase Extraction/instrumentation
18.
Pulm Med ; 2014: 581738, 2014.
Article in English | MEDLINE | ID: mdl-25120928

ABSTRACT

This paper indicated that inactivated Bordetella pertussis (iBp) can enhance the lung airway hyperreactivity of the rats sensitized and challenged with OVA. The mechanisms were involved in the upregulation of cAMP-PDE activity and PDE4A, PDE4D, and PDE3 gene expression in the lungs. But only PDE4 activity was different between the OVA and OVA+iBp groups, and PDE4D expression was significantly increased in iBp rats alone. So, our data suggested that cosensitization with OVA and iBp affects lung airway reactivity by modulating the lung cAMP-PDE activity and PDE4D gene expression.


Subject(s)
Lung/enzymology , Pertussis Vaccine/pharmacology , Phosphoric Diester Hydrolases/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Bordetella pertussis/immunology , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Gene Expression Regulation/drug effects , Lung/immunology , Male , Ovalbumin/immunology , Pertussis Vaccine/immunology , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects , Up-Regulation/immunology , Vaccines, Inactivated
19.
Adv Pharmacol Sci ; 2014: 981624, 2014.
Article in English | MEDLINE | ID: mdl-24864138

ABSTRACT

A simple, robust LC-MS/MS assay for quantifying cefuroxime in human plasma was developed. Cefuroxime and tazobactam, as internal standard (IS), were extracted from human plasma by methanol to precipitate protein. Separation was achieved on a Zorbax SB-Aq (4.6 × 250 mm, 5 µ m) column under isocratic conditions. The calibration curve was linear in the concentration range of 0.0525-21.0 µ g/mL (r = 0.9998). The accuracy was higher than 90.92%, while the intra- and interday precision were less than 6.26%. The extraction procedure provides recovery ranged from 89.44% to 92.32%, for both analyte and IS. Finally, the method was successfully applied to a bioequivalence study of a single 500 mg dose of cefuroxime axetil in 22 healthy Chinese male subjects under fasting condition. Bioequivalence was determined by calculating 90% Cls for the ratios of C max, AUC0-t , and AUC0-∞ values for the test and reference products, using logarithmic transformed data. The 90% Cls for the ratios of C max (91.4%~104.2%), AUC0-t (97.4%~110.9%), and AUC0-∞ (97.6%~111.1%) values were within the predetermined range. It was concluded that the two formulations (test for capsule, reference for tablet) analyzed were bioequivalent in terms of rate and extent of absorption and the method met the principle of quick and easy clinical analysis.

20.
Int J Clin Pharmacol Ther ; 51(10): 816-22, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23735180

ABSTRACT

OBJECTIVES: Although sirolimus tablets and oral solutions have been used in clinical practice, no study has been reported on the pharmacokinetics and bioavailability of a single-dose of sirolimus tablets in healthy Chinese volunteers. The purpose of this study was to compare the bioavailability and pharmacokinetic (PK) properties of two different 1-mg sirolimus tablets in healthy Chinese male volunteers and evaluate whether a generic tablet of sirolimus meets the criteria for bioequivalence from the State Food and Drug Administration (SFDA) of China when compared with a reference product. MATERIALS AND METHODS: A total of 24 healthy Chinese volunteers was eligible for this 6 mg single dose, randomized-sequence, open-label, 2-period crossover study. Blood samples were collected before dosing and at 0.25, 0.50, 0.75, 1.0, 1.5, 2, 3, 4, 8, 12, 24, 48, 72, 120, 168, 216, and 264 hours after dosing. Whole blood sirolimus concentration was analyzed by a validated liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. Pharmacokinetic properties of sirolimus were assessed using noncompartmental analysis. RESULTS: The mean (range) Cmax values of the test and the reference were 15.25 (8.48 - 24.40) and 13.43 (7.90 - 22.90) ng/ml; AUC0-t values were 475.65 (293.33 - 1049.86) and 451.96 (221.52 - 809.11) ng/h/ml. The medians (range) tmax values were 2.0 (1.0 - 8.0) and 2.0 (1.0 - 8.0) hours, respectively. The 90% confidence intervals (CIs) for the ratios of Cmax, AUC0-264, and AUC0-∞ were 103.7% to 124.4%, 97.5% to 113.6%, and 98.0% to 114.8%, respectively. CONCLUSION: In this single-dose crossover study the test sirolimus tablets met the criteria for bioequivalence in terms of both rate and extent. Each sirolimus formulation was well tolerated during the study.


Subject(s)
Sirolimus/pharmacokinetics , Adult , Area Under Curve , Asian People , Biological Availability , Chemistry, Pharmaceutical , Cross-Over Studies , Drugs, Generic/pharmacokinetics , Healthy Volunteers , Humans , Male , Sirolimus/administration & dosage , Sirolimus/blood , Tablets/pharmacokinetics , Therapeutic Equivalency , United States , United States Food and Drug Administration , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...