Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 592
Filter
1.
Drug Des Devel Ther ; 18: 1515-1528, 2024.
Article in English | MEDLINE | ID: mdl-38716369

ABSTRACT

Purpose: Estrogen deficiency is the main reason of postmenopausal osteoporosis. Eldecalcitol (ED-71) is a new active vitamin D analogue clinically used in the treatment of postmenopausal osteoporosis. We aimed to investigate whether EphrinB2-EphB4 and RANKL/RANK/OPG signaling cooperate in mediating the process of osteoporosis by ED-71. Methods: In vivo, the ovariectomized (OVX) rats were administered orally with 30 ng/kg ED-71 once a day for 8 weeks. HE staining, Masson staining and Immunofluorescence staining were used to evaluate bone mass, bone formation, osteoclastogenesis associated factors and the expression of EphrinB2, EphB4, RANKL and OPG. In vitro, H2O2 stimulation was used to simulate the cell environment in osteoporosis. Immunofluorescence, quantitative real time PCR (qRT-PCR), enzyme-linked immunosorbent assay (ELISA) and Western Blot were applied to detect the expression of EphrinB2, EphB4, RANKL and OPG. In osteoblasts, EphB4 was knocked down by EphB4 small-interfering RNA (siRNA) transfection. LY294002 (PI3K inhibitor) or ARQ092 (AKT inhibitor) was used to block PI3K/AKT pathway. An indirect co-culture system of osteoblasts and osteoclasts was established. The mRNA and protein expression of osteoclastogenes is associated factors were tested by qRT-PCR and Western Blot. Results: ED-71 increased bone mass and decreased the number of osteoclasts in OVX rats. Moreover, ED-71 promoted the expression of EphrinB2, EphB4, and decreased the RANKL/OPG ratio in osteoblasts. Osteoclastogenesis was restrained when osteoclasts were indirectly co-cultured with ED-71-treated osteoblasts. After silencing of EphB4 expression in osteoblasts, ED-71 inhibited the expression of P-PI3K and P-AKT and increased the ratio of RANKL/OPG. This reversed the inhibitory effect of ED-71 on osteoclastogenes. Therefore, in ED-71-inhibited osteoclastogenes, EphB4 is a key factor affecting the secretion of RANKL and OPG by osteoblasts. EphB4 suppressed the RANKL/OPG ratio through activating PI3K/AKT signaling in osteoblasts. Conclusion: ED-71 inhibits osteoclastogenesis through EphrinB2-EphB4-RANKL/OPG axis, improving bone mass in ovariectomized rats. PI3K/AKT pathway is involved this process.


Subject(s)
Ephrin-B2 , Osteoprotegerin , Ovariectomy , RANK Ligand , Rats, Sprague-Dawley , Receptor, EphB4 , Animals , Rats , RANK Ligand/metabolism , RANK Ligand/antagonists & inhibitors , Female , Receptor, EphB4/metabolism , Receptor, EphB4/antagonists & inhibitors , Ephrin-B2/metabolism , Ephrin-B2/antagonists & inhibitors , Osteoprotegerin/metabolism , Vitamin D/pharmacology , Vitamin D/analogs & derivatives , Osteogenesis/drug effects , Cells, Cultured , Osteoclasts/drug effects , Osteoclasts/metabolism , Signal Transduction/drug effects , Bone Density/drug effects
2.
J Med Chem ; 67(9): 7585-7602, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38630440

ABSTRACT

An efficient protocol for the synthesis of ß-trifluoroethoxydimethyl selenides was achieved under mild reaction conditions, and 39 compounds were prepared. All compounds were evaluated for their abilities to inhibit RANKL-induced osteoclastogenesis, compound 4aa exhibited the most potent activity. Further investigations revealed that 4aa could inhibit F-actin ring generation, bone resorption, and osteoclast-specific gene expression in vitro. Western blot analyses demonstrated that compound 4aa abrogated the RANKL-induced mitogen-activated protein kinase and NF-kB-signaling pathways. In addition, 4aa also displayed a notable impact on the osteoblastogenesis of MC3T3-E1 preosteoblasts. In vivo experiments revealed that compound 4aa significantly ameliorated bone loss in an ovariectomized (OVX) mice model. Furthermore, the surface plasmon resonance experiment results revealed that 4aa probably bound to RANKL. Collectively, the above-mentioned findings suggested that compound 4aa as a potential RANKL inhibitor averted OVX-triggered osteoporosis by regulating the inhibition of osteoclast differentiation and stimulation of osteoblast differentiation.


Subject(s)
Drug Design , Osteoclasts , Osteoporosis , RANK Ligand , Animals , Mice , Osteoporosis/drug therapy , RANK Ligand/metabolism , RANK Ligand/antagonists & inhibitors , Female , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Cell Differentiation/drug effects , Ovariectomy , Organoselenium Compounds/pharmacology , Organoselenium Compounds/chemical synthesis , Organoselenium Compounds/chemistry , Structure-Activity Relationship , Osteogenesis/drug effects , Bone Resorption/drug therapy , NF-kappa B/metabolism , NF-kappa B/antagonists & inhibitors , Mice, Inbred C57BL
3.
J Bone Miner Res ; 39(4): 484-497, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38477789

ABSTRACT

Rebound bone loss following denosumab discontinuation is an important clinical challenge. Current treatment strategies to prevent this fail to suppress the rise and overshoot in osteoclast-mediated bone resorption. In this study, we use a murine model of denosumab treatment and discontinuation to show the temporal changes in osteoclast formation and activity during RANKL inhibition and withdrawal. We show that the cellular processes that drive the formation of osteoclasts and subsequent bone resorption following withdrawal of RANKL inhibition precede the rebound bone loss. Furthermore, a rise in serum TRAP and RANKL levels is detected before markers of bone turnover used in current clinical practice. These mechanistic advances may provide insight into a more defined window of opportunity to intervene with sequential therapy following denosumab discontinuation.


Stopping denosumab, a medication commonly used to improve bone mass by blocking formation of bone resorbing osteoclasts, leads to a rebound loss in the bone which was gained during treatment. Current strategies to prevent this bone loss fail in most cases as they are unable to prevent the rise and overshoot in bone resorption by osteoclasts. Thie stems from an incomplete understanding of how osteoclasts behave during denosumab treatment and after treatment is discontinued. We use a mouse model of this phenomenon to show how osteoclast formation and activity changes throughout this process. We show that increases in the processes that drive the formation of osteoclasts can be detected in the circulation before bone loss occurs. These findings could therefore provide insight into a targeted 'window of opportunity' to intervene and prevent the rebound bone loss following stopping denosumab in patients.


Subject(s)
Bone Resorption , Denosumab , Osteoclasts , RANK Ligand , Animals , Osteoclasts/metabolism , Osteoclasts/drug effects , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , Denosumab/pharmacology , Mice , Bone Resorption/pathology , Bone Resorption/drug therapy , Bone Resorption/blood , Time Factors , Tartrate-Resistant Acid Phosphatase/metabolism , Female , Mice, Inbred C57BL , Biomarkers/metabolism , Biomarkers/blood
4.
J Cell Biochem ; 125(2): e30518, 2024 02.
Article in English | MEDLINE | ID: mdl-38224182

ABSTRACT

Excessive bone-resorbing osteoclast activity during bone remodeling is a major feature of bone diseases, such as osteoporosis. Therefore, the inhibition of osteoclast formation and bone resorption can be an effective therapeutic target for various bone diseases. Gryllus biomaculatus (GB) has recently been approved as an alternative food source because of its high nutritional value and environmental sustainability. Traditionally, GB has been known to have various pharmacological properties, including antipyretic and blood pressure-lowering activity, and it has recently been reported to have various biological activities, including protective effects against inflammation, oxidative stress, insulin resistance, and alcohol-induced liver injury. However, the effect of GB on osteoclast differentiation and bone metabolism has not yet been demonstrated. In this study, we confirmed the inhibitory effect of GB extract (GBE) on the receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation. To determine the effect of GBE on RANKL-induced osteoclast differentiation and function, we performed TRAP and F-actin staining, as well as a bone-resorbing assay. The intracellular mechanisms of GBE responsible for the regulation of osteoclastogenesis were revealed by Western blot analysis and quantitative real-time polymerase chain reaction. We investigated the relationship between GBE and expression of osteoclast-specific molecules to further elucidate the underlying mechanisms. It was found that GBE significantly suppressed osteoclastogenesis by decreasing the phosphorylation of Akt, p38, JNK, and ERK, as well as Btk-PLCγ2 signaling, in pathways involved in early osteoclastogenesis as well as through the subsequent suppression of c-Fos, NFATc1, and osteoclastogenesis-specific marker genes. Additionally, GBE inhibited the formation of F-actin ring-positive osteoclasts and bone resorption activity of mature osteoclasts. Our findings suggest that GBE is a potential functional food and therapeutic candidate for bone diseases involving osteoclasts.


Subject(s)
Bone Resorption , Osteoclasts , RANK Ligand , Humans , Actins/metabolism , Bone Resorption/drug therapy , Cell Differentiation , Ligands , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism
5.
Cancer Immunol Res ; 12(4): 453-461, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38276989

ABSTRACT

Denosumab is a fully human mAb that binds receptor activator of NFκB ligand (RANKL). It is routinely administered to patients with cancer to reduce the incidence of new bone metastasis. RANK-RANKL interactions regulate bone turnover by controlling osteoclast recruitment, development, and activity. However, these interactions also can regulate immune cells including dendritic cells and medullary thymic epithelial cells. Inhibition of the latter results in reduced thymic negative selection of T cells and could enhance the generation of tumor-specific T cells. We examined whether administering denosumab could modify modulate circulating immune cells in patients with cancer. Blood was collected from 23 patients with prostate cancer and 3 patients with renal cell carcinoma, all of whom had advanced disease and were receiving denosumab, prior to and during denosumab treatment. Using high-dimensional mass cytometry, we found that denosumab treatment by itself induced modest effects on circulating immune cell frequency and activation. We also found minimal changes in the circulating T-cell repertoire and the frequency of new thymic emigrants with denosumab treatment. However, when we stratified patients by whether they were receiving chemotherapy and/or steroids, patients receiving these concomitant treatments showed significantly greater immune modulation, including an increase in the frequency of natural killer cells early and classical monocytes later. We also saw broad induction of CTLA-4 and TIM3 expression in circulating lymphocytes and some monocyte populations. These findings suggest that denosumab treatment by itself has modest immunomodulatory effects, but when combined with conventional cancer treatments, can lead to the induction of immunologic checkpoints. See related Spotlight by Nasrollahi and Davar, p. 383.


Subject(s)
Bone Neoplasms , Denosumab , Humans , Male , Bone Neoplasms/drug therapy , Bone Neoplasms/prevention & control , Bone Neoplasms/secondary , Denosumab/therapeutic use , Kidney Neoplasms/drug therapy , RANK Ligand/antagonists & inhibitors , Prostatic Neoplasms/drug therapy
6.
Head Neck ; 46(1): E1-E5, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37823398

ABSTRACT

BACKGROUND: McCune-Albright syndrome is a rare disorder characterized by polyostotic fibrous dysplasia (FD), café-au-lait skin pigmentation, and endocrine dysfunction. Extensive FD in the craniofacial region can present significant challenges in terms of disease control and carries a high risk of permanent visual impairment. METHODS: We present a case of medically and surgically resistant FD that required nine optic nerve decompressions. RESULTS: The condition was ultimately controlled with the use of the denosumab agent. CONCLUSION: The case highlights the importance and potential efficacy of denosumab in resistant FD management, particularly in cases involving sensitive organs.


Subject(s)
Craniofacial Fibrous Dysplasia , Fibrous Dysplasia, Polyostotic , Humans , Bone and Bones , Craniofacial Fibrous Dysplasia/drug therapy , Decompression, Surgical , Denosumab , Fibrous Dysplasia, Polyostotic/diagnostic imaging , Fibrous Dysplasia, Polyostotic/drug therapy , Fibrous Dysplasia, Polyostotic/surgery , RANK Ligand/antagonists & inhibitors
7.
Int J Mol Sci ; 24(14)2023 Jul 10.
Article in English | MEDLINE | ID: mdl-37511048

ABSTRACT

Receptor activator of nuclear factor-κB ligand (RANKL) has been actively pursued as a therapeutic target for osteoporosis, given that RANKL is the master mediator of bone resorption as it promotes osteoclast differentiation, activity and survival. We employed a structure-based virtual screening approach comprising two stages of experimental evaluation and identified 11 commercially available compounds that displayed dose-dependent inhibition of osteoclastogenesis. Their inhibitory effects were quantified through TRAP activity at the low micromolar range (IC50 < 5 µΜ), but more importantly, 3 compounds displayed very low toxicity (LC50 > 100 µΜ). We also assessed the potential of an N-(1-aryl-1H-indol-5-yl)aryl-sulfonamide scaffold that was based on the structure of a hit compound, through synthesis of 30 derivatives. Their evaluation revealed 4 additional hits that inhibited osteoclastogenesis at low micromolar concentrations; however, cellular toxicity concerns preclude their further development. Taken together with the structure-activity relationships provided by the hit compounds, our study revealed potent inhibitors of RANKL-induced osteoclastogenesis of high therapeutic index, which bear diverse scaffolds that can be employed in hit-to-lead optimization for the development of therapeutics against osteolytic diseases.


Subject(s)
Bone Resorption , Osteogenesis , RANK Ligand , Humans , Bone Resorption/drug therapy , Cell Differentiation , I-kappa B Proteins , NF-kappa B/pharmacology , NFATC Transcription Factors , Osteoclasts , Osteogenesis/drug effects , RANK Ligand/antagonists & inhibitors , Structure-Activity Relationship
8.
Expert Opin Biol Ther ; 23(8): 705-715, 2023.
Article in English | MEDLINE | ID: mdl-36892190

ABSTRACT

BACKGROUND: MW031 is a biosimilar candidate of denosumab (Prolia®). This study aimed to compare the pharmacokinetics, pharmacodynamics, safety and immunogenicity of MW031 to denosumab in healthy Chinese participants. RESEARCH DESIGN AND METHODS: In this single-center, randomized, double-blind, parallel-controlled, single-dose trial, participants were given 60 mg MW031 (N = 58) or denosumab (N = 61) by subcutaneous injection and observed for 140 days. The primary endpoint was the bioequivalence of PK parameters (Cmax, AUC0-∞), and secondary endpoints including PD parameter, safety, and immunogenicity. RESULTS: A comparison of main PK parameters showed that the geometric mean ratios (GMR) (90% confidence intervals [CIs]) of AUC0-∞ and Cmax for MW031 over denosumab were 105.48% (98.96%, 112.43%) and 98.58% (92.78%, 104.75%), respectively. The inter-CV values of AUC0-∞ and Cmax for MW031 ranged from 19.9% to 23.1%. PD parameter (sCTX) in the MW031 and denosumab groups were similar, and the positivity rates of immunogenicity were 0% in both groups. This study also showed similar safety profiles in both groups, and there were no drug-related, high-incidence and previously unreported adverse reactions. CONCLUSION: This trial confirmed similar pharmacokinetic profiles of MW031 and denosumab in healthy male participants, and pharmacodynamic profile, immunogenicity and safety were comparable for both drugs. TRIAL REGISTRATION: NCT04798313; CTR20201149.


Subject(s)
Antibodies, Monoclonal, Humanized , Biosimilar Pharmaceuticals , Denosumab , Humans , Male , Area Under Curve , Biosimilar Pharmaceuticals/adverse effects , Biosimilar Pharmaceuticals/metabolism , Biosimilar Pharmaceuticals/pharmacokinetics , Denosumab/adverse effects , Denosumab/immunology , Denosumab/pharmacokinetics , Double-Blind Method , East Asian People , Healthy Volunteers , Therapeutic Equivalency , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , RANK Ligand/antagonists & inhibitors , RANK Ligand/immunology , Injections, Subcutaneous
10.
Drug Des Devel Ther ; 15: 4741-4757, 2021.
Article in English | MEDLINE | ID: mdl-34848946

ABSTRACT

BACKGROUND: Osteolytic diseases such as osteoporosis are featured with accelerated osteoclast differentiation and strong bone resorption. Considering the complications and other limitations of current drug treatments, it is necessary to develop a safer and more reliable drug to deal with osteoclast-related diseases. Saikosaponin D (SSD) is the active extract of Bupleurum, which has anti-inflammation, anti-tumor and liver protection functions. However, the role of SSD in regulating the differentiation and function of osteoclasts is not clear. PURPOSE: To explore whether SSD could prevent osteoclast differentiation and bone resorption induced by M-CSF and RANKL, and further evaluate the potential therapeutic properties of SSD in LPS-induced inflammatory bone loss mouse models. METHODS: BMMs were cultured in complete medium stimulated by RANKL with different concentrations of SSD. TRAP staining, bone resorption determination, qRT-PCR, immunofluorescence and Western blotting were performed. A mouse model of LPS-induced calvarial bone loss was established and treated with different doses of SSD. The excised calvaria bones were used for TRAP staining, micro-CT scan and histological analysis. RESULTS: SSD inhibited the formation and bone resorption of osteoclasts induced by RANKL in vitro. SSD suppressed LPS-induced inflammatory bone loss in vivo. CONCLUSION: SSD inhibited osteoclastogenesis and LPS-induced osteolysis in mice both which served as a new potential agent for the treatment of osteoclast-related conditions.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bone Resorption/drug therapy , Lipopolysaccharides/antagonists & inhibitors , Oleanolic Acid/analogs & derivatives , RANK Ligand/antagonists & inhibitors , Receptor Activator of Nuclear Factor-kappa B/antagonists & inhibitors , Saponins/pharmacology , Animals , Bone Resorption/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Oleanolic Acid/pharmacology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism
11.
J Nat Prod ; 84(10): 2738-2743, 2021 10 22.
Article in English | MEDLINE | ID: mdl-34612636

ABSTRACT

A simple methylenedioxy dibromoindole alkaloid, amakusamine (1), was isolated from a marine sponge of the genus Psammocinia, and its structure was determined from spectroscopic data, time-dependent density-functional theory calculations, and synthesis. Compound 1 inhibited the receptor activator of nuclear factor-κB ligand (RANKL)-induced formation of multinuclear osteoclasts with an IC50 value of 10.5 µM in RAW264 cells. The structure-activity relationship of 1 was also investigated with synthetic derivatives.


Subject(s)
Alkaloids/pharmacology , Osteoclasts/drug effects , Porifera/chemistry , RANK Ligand/antagonists & inhibitors , Animals , Japan , Mice , Molecular Structure , RAW 264.7 Cells , Structure-Activity Relationship
12.
Bioorg Chem ; 116: 105361, 2021 11.
Article in English | MEDLINE | ID: mdl-34562672

ABSTRACT

The OPG/RANKL/RANK pathway is a promising target for the design of therapeutic agents used in the treatment of osteoporosis. E09241 with an N-methylpyridine-chlorofuranformamide structural skeleton was previously identified to decrease bone loss and thus protect against osteoporosis in ovariectomized rats through increasing osteoprotegerin (OPG) expression. In this study, 36 derivatives of E09241 (3a) were prepared. The synthesis, up-regulation of OPG activities, SAR (structure-activity relationship), and cytotoxicity of these compounds are presented. Compounds with good up-regulating OPG activities could inhibit RANKL (the receptor activator of nuclear factor-kappa B ligand)-induced osteoclastogenesis in RAW264.7 cells. Particularly, compounds 3c and 3i1 significantly reduced NFATc1 and MMP-9 protein expression through inhibition of the NF-κB and MAPK pathways in RANKL induced RAW264.7 cells. In addition, compounds 3c and 3v significantly promoted osteoblast differentiation in MC3T3-E1 cells in osteogenic medium, and compounds 3c, 3v, and 3i1 obviously increased OPG protein expression and secretion in MC3T3-E1 cells. Furthermore, the pharmacokinetic profiles, acute toxicity, and hERG K+ channel effects of compounds 3a, 3c, 3e, 3v, and 3i1 were investigated. Taken together, these results indicate that N-methylpyridine-chlorofuranformamide analog 3i1 could serve as a promising lead for the development of new agents for treating osteoporosis.


Subject(s)
Formamides/pharmacology , Furans/pharmacology , Osteoprotegerin/metabolism , Pyridines/pharmacology , RANK Ligand/antagonists & inhibitors , 3T3 Cells , Animals , Dose-Response Relationship, Drug , Formamides/chemistry , Furans/chemistry , Mice , Molecular Structure , Osteogenesis/drug effects , Pyridines/chemistry , RANK Ligand/metabolism , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
13.
Cell Death Dis ; 12(9): 825, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34475393

ABSTRACT

Osteoporosis-related fractures, such as femoral neck and vertebral fractures, are common in aged people, resulting in increased disability rate and health-care costs. Thus, it is of great importance to clarify the mechanism of osteoclast-related osteoporosis and find effective ways to avoid its complication. In this study, gene expression profile analysis and real-time polymerase chain reaction revealed that DUSP6 expression was suppressed in human and mice osteoporosis cases. In vitro experiments confirmed that DUSP6 overexpression prevented osteoclastogenesis, whereas inhibition of DUSP6 by small interference RNA or with a chemical inhibitor, (E/Z)-BCI, had the opposite effect. (E/Z)-BCl significantly accelerated the bone loss process in vivo by enhancing osteoclastogenesis. Bioinformatics analyses and in vitro experiments indicated that miR-181a was an upstream regulator of DUSP6. Moreover, miR-181a positively induced the differentiation and negatively regulated the apoptosis of osteoclasts via DUSP6. Furthermore, downstream signals by ERK2 and SMAD2 were also found to be involved in this process. Evaluation of ERK2-deficiency bone marrow-derived macrophages confirmed the role of ERK2 signaling in the DUSP6-mediated osteoclastogenesis. Additionally, immunoprecipitation assays confirmed that DUSP6 directly modified the phosphorylation status of SMAD2 and the subsequent nuclear transportation of NFATC1 to regulate osteoclast differentiation. Altogether, this study demonstrated for the first time the role of miRNA-181a/DUSP6 in the progression of osteoporosis via the ERK2 and SMAD2 signaling pathway. Hence, DUSP6 may represent a novel target for the treatment of osteoclast-related diseases in the future.


Subject(s)
Cell Differentiation , Dual Specificity Phosphatase 6/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Osteoclasts/pathology , Osteoporosis/pathology , Signal Transduction , Smad2 Protein/metabolism , Animals , Bone Resorption/complications , Bone Resorption/pathology , Bone and Bones/drug effects , Bone and Bones/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/genetics , Dual Specificity Phosphatase 6/metabolism , Humans , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteogenesis/genetics , Osteoporosis/complications , Osteoporosis/enzymology , Osteoporosis/genetics , RANK Ligand/antagonists & inhibitors , RANK Ligand/pharmacology , Signal Transduction/drug effects , Tartrate-Resistant Acid Phosphatase/metabolism
14.
Cells ; 10(8)2021 08 04.
Article in English | MEDLINE | ID: mdl-34440747

ABSTRACT

The receptor activator of the nuclear factor-κB ligand (RANKL)/RANK signaling pathway was identified in the late 1990s and is the key mediator of bone remodeling. Targeting RANKL with the antibody denosumab is part of the standard of care for bone loss diseases, including bone metastases (BM). Over the last decade, evidence has implicated RANKL/RANK pathway in hormone and HER2-driven breast carcinogenesis and in the acquisition of molecular and phenotypic traits associated with breast cancer (BCa) aggressiveness and poor prognosis. This marked a new era in the research of the therapeutic use of RANKL inhibition in BCa. RANKL/RANK pathway is also an important immune mediator, with anti-RANKL therapy recently linked to improved response to immunotherapy in melanoma, non-small cell lung cancer (NSCLC), and renal cell carcinoma (RCC). This review summarizes and discusses the pre-clinical and clinical evidence of the relevance of the RANKL/RANK pathway in cancer biology and therapeutics, focusing on bone metastatic disease, BCa onset and progression, and immune modulation.


Subject(s)
Breast Neoplasms/pathology , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Denosumab/therapeutic use , Drug Repositioning , Female , Humans , RANK Ligand/antagonists & inhibitors , Signal Transduction , Zoledronic Acid/therapeutic use
15.
Cancer Lett ; 520: 374-384, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34416336

ABSTRACT

Normal breast fibroblasts (NBFs) support and maintain the architecture of the organ, and can also suppress tumorigenesis. However, the mechanisms involved are not fully understood. We have shown here that NBFs suppress breast carcinogenesis through secretion of osteoprotegerin (OPG), a soluble decoy receptor for the Receptor Activator of NF-κB ligand (RANKL). Indeed, NBFs and human recombinant OPG (rOPG), suppressed breast cancer cells proliferation and motility through inhibition of the epithelial-to-mesenchymal transition (EMT) process both in vitro and in vivo. Additionally, rOPG inhibited the IL-6/STAT3 and NF-κB pathways as well as the OPG gene, which turned out to be STAT3-regulated. This was confirmed using denosumab, a RANKL-targeted antibody, which also inhibited NF-κB, down-regulated OPG and repressed EMT in breast cancer cells grown in 2D and 3D. Importantly, both rOPG and denosumab targeted cancer stem cells (CSCs). This was mediated through inhibition of the CSC-related pathway ß-catenin. Moreover, rOPG reduced tumor growth and inhibited breast CSC biomarkers in orthotopic humanized breast tumors. Therefore, normal mammary fibroblasts can suppress carcinogenesis through OPG, which constitutes great potential as preventive and/or therapeutic molecule for breast carcinomas.


Subject(s)
Breast Neoplasms/drug therapy , Osteoprotegerin/genetics , RANK Ligand/genetics , Recombinant Proteins/genetics , beta Catenin/genetics , Antibodies/pharmacology , Anticarcinogenic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Denosumab/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Female , Fibroblasts/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/genetics , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Osteoprotegerin/immunology , Osteoprotegerin/pharmacology , RANK Ligand/antagonists & inhibitors , Recombinant Proteins/pharmacology , STAT3 Transcription Factor/genetics
16.
Sci Rep ; 11(1): 12186, 2021 06 09.
Article in English | MEDLINE | ID: mdl-34108600

ABSTRACT

Prostate cancer (PCa) morbidity in the majority of patients is due to metastatic events, which are a clinical obstacle. Therefore, a better understanding of the mechanism underlying metastasis is imperative if we are to develop novel therapeutic strategies. Receptor activator of nuclear factor kappa-B (NF-κB) ligand (RANKL) regulates bone remodelling. Thus, agents that suppress RANKL signalling may be useful pharmacological treatments. Here, we used preclinical experimental models to investigate whether an inactive form of RANKL affects bone metastasis in RANKL-induced PCa. RANKL was associated with epithelial-mesenchymal transition (EMT) and expression of metastasis-related genes in PC3 cells. Therefore, we proposed a strategy to induce anti-cytokine antibodies using mutant RANKL as an immunogen. RANKL promoted migration and invasion of PC3 cells through EMT, and induced a significant increase in binding of ß-catenin to TCF-4, an EMT-induced transcription factor in PCa cells, via mitogen-activated protein kinase and ß-catenin/TCF-4 signalling. Thus, RANKL increased EMT and the metastatic properties of PC3 cells, suggesting a role as a therapeutic target to prevent PCa metastasis. Treatment with mutant RANKL reduced EMT and metastasis of PC3 PCa cells in an experimental metastasis model. Thus, mutant RANKL could serve as a potential vaccine to prevent and treat metastatic PCa.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bone Neoplasms/prevention & control , Epithelial-Mesenchymal Transition , Prostatic Neoplasms/drug therapy , RANK Ligand/antagonists & inhibitors , Animals , Apoptosis , Bone Neoplasms/immunology , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Movement , Cell Proliferation , Humans , Immunization , Male , Mice , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RANK Ligand/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Int Immunopharmacol ; 97: 107720, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33945918

ABSTRACT

Presently, postmenopausal osteoporosis mainly caused by excessive activation of in vivo osteoclasts has become a global public health burden. Natural compounds have gradually become the potential drugs for the treatment of postmenopausal osteoporosis. Aloperine is a new alkaloid extracted from the leaves and seeds of sophora bean. The current studies have proved that aloperine has many biological activities, including anti-inflammatory, antiviral and anticancer activities. This study shows that aloperine can inhibit activity and formation of osteoclast mediated by RANKL in a dose-dependent manner without affecting the activity of bone marrow macrophages (BMM). In addition, it is found that aloperine can inhibit the expression of osteoclast specific marker genes, including nuclear factor of activated T cells cytoplasmic 1 (NFATc1), tartrate resistant acid phosphatase (TRAcP), matrix metallopeptidase 9 (MMP9), cathepsin K (Ctsk), V-ATPase d2 and calcitonin receptor. The in vitro experiment of aloperine proved that aloperine can inhibit the degradation of IκBα and the phosphorylation of P65, ERK and JNK. Additionally, aloperine improves bone loss in ovariectomized (OVX) mice by inhibiting osteoclast activity. This project proved that aloperine can affect the formation of osteoclasts by inhibiting RANKL signaling channel, and it is indicated that aloperine has the potential to be developed as a new drug for the prevention and treatment of postmenopausal osteoporosis.


Subject(s)
Osteogenesis/drug effects , Osteoporosis, Postmenopausal/drug therapy , Quinolizidines/pharmacology , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Femur/drug effects , Femur/pathology , Humans , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Osteoclasts/drug effects , Osteoporosis, Postmenopausal/diagnosis , Osteoporosis, Postmenopausal/etiology , Osteoporosis, Postmenopausal/pathology , Ovariectomy , Quinolizidines/therapeutic use , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , X-Ray Microtomography
18.
Nat Commun ; 12(1): 2450, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33893301

ABSTRACT

Infertile men have few treatment options. Here, we demonstrate that the transmembrane receptor activator of NF-kB ligand (RANKL) signaling system is active in mouse and human testis. RANKL is highly expressed in Sertoli cells and signals through RANK, expressed in most germ cells, whereas the RANKL-inhibitor osteoprotegerin (OPG) is expressed in germ and peritubular cells. OPG treatment increases wild-type mouse sperm counts, and mice with global or Sertoli-specific genetic suppression of Rankl have increased male fertility and sperm counts. Moreover, RANKL levels in seminal fluid are high and distinguishes normal from infertile men with higher specificity than total sperm count. In infertile men, one dose of Denosumab decreases RANKL seminal fluid concentration and increases serum Inhibin-B and anti-Müllerian-hormone levels, but semen quality only in a subgroup. This translational study suggests that RANKL is a regulator of male reproductive function, however, predictive biomarkers for treatment-outcome requires further investigation in placebo-controlled studies.


Subject(s)
Fertility/physiology , RANK Ligand/metabolism , Semen Analysis/methods , Sertoli Cells/metabolism , Spermatozoa/metabolism , Animals , Anti-Mullerian Hormone/blood , Anti-Mullerian Hormone/metabolism , Denosumab/pharmacology , Fertility/drug effects , Humans , Inhibins/antagonists & inhibitors , Inhibins/blood , Inhibins/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Osteoprotegerin/pharmacology , RANK Ligand/antagonists & inhibitors , RANK Ligand/genetics , Semen/drug effects , Semen/metabolism , Sertoli Cells/drug effects , Sperm Count , Spermatozoa/cytology , Spermatozoa/drug effects
19.
Int J Mol Sci ; 22(5)2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33669069

ABSTRACT

Osteoclasts, bone-specified multinucleated cells produced by monocyte/macrophage, are involved in numerous bone destructive diseases such as arthritis, osteoporosis, and inflammation-induced bone loss. The osteoclast differentiation mechanism suggests a possible strategy to treat bone diseases. In this regard, we recently examined the in vivo impact of kalkitoxin (KT), a marine product obtained from the marine cyanobacterium Moorena producens (previously Lyngbya majuscula), on the macrophage colony-stimulating factor (M-CSF) and on the receptor activator of nuclear factor κB ligand (RANKL)-stimulated in vitro osteoclastogenesis and inflammation-mediated bone loss. We have now examined the molecular mechanism of KT in greater detail. KT decreased RANKL-induced bone marrow-derived macrophages (BMMs) tartrate-resistant acid phosphatase (TRAP)-multinucleated cells at a late stage. Likewise, KT suppressed RANKL-induced pit area and actin ring formation in BMM cells. Additionally, KT inhibited several RANKL-induced genes such as cathepsin K, matrix metalloproteinase (MMP-9), TRAP, and dendritic cell-specific transmembrane protein (DC-STAMP). In line with these results, RANKL stimulated both genes and protein expression of c-Fos and nuclear factor of activated T cells (NFATc1), and this was also suppressed by KT. Moreover, KT markedly decreased RANKL-induced p-ERK1/2 and p-JNK pathways at different time points. As a result, KT prevented inflammatory bone loss in mice, such as bone mineral density (BMD) and osteoclast differentiation markers. These experiments demonstrated that KT markedly inhibited osteoclast formation and inflammatory bone loss through NFATc1 and mitogen-activated protein kinase (MAPK) signaling pathways. Therefore, KT may have potential as a treatment for destructive bone diseases.


Subject(s)
Bone Resorption/drug therapy , Lipids/therapeutic use , MAP Kinase Signaling System/drug effects , NFATC Transcription Factors/metabolism , Osteoclasts/drug effects , Osteogenesis/drug effects , Thiazoles/therapeutic use , Actins/genetics , Actins/metabolism , Animals , Bone Density/drug effects , Bone Resorption/metabolism , Cathepsin K/genetics , Cathepsin K/metabolism , Cell Survival , Inflammation/drug therapy , Inflammation/metabolism , Janus Kinases/metabolism , Lipids/pharmacology , Lipopolysaccharides/toxicity , Lyngbya/chemistry , MAP Kinase Signaling System/genetics , Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred ICR , NFATC Transcription Factors/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Osteoclasts/metabolism , Osteogenesis/genetics , Phosphorylation , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , RANK Ligand/pharmacology , Tartrate-Resistant Acid Phosphatase/genetics , Tartrate-Resistant Acid Phosphatase/metabolism , Thiazoles/pharmacology
20.
Am J Respir Cell Mol Biol ; 64(5): 617-628, 2021 05.
Article in English | MEDLINE | ID: mdl-33689672

ABSTRACT

Skeletal muscle dysfunction is one of the important comorbidities of chronic obstructive pulmonary disease (COPD); however, the underlying mechanisms remain largely unknown. RANKL (receptor activator of nuclear factor κB ligand), a key mediator in osteoclast differentiation, was also found to play a role in skeletal muscle pathogenesis. Whether RANKL is involved in COPD-related skeletal muscle dysfunction is as-of-yet unknown. We examined the expression of RANKL/RANK in skeletal muscles from mice exposed to cigarette smoke (CS) for 24 weeks. Grip strength and exercise capacity as well as muscular morphology were evaluated in CS-exposed mice with or without anti-RANKL treatment. The expressions of protein synthesis- or muscle growth-related molecules (IGF-1, myogenin, and myostatin), muscle-specific ubiquitin E3 ligases (MuRF1 and atrogin-1), and the NF-κb inflammatory pathway were also evaluated in skeletal muscles. The effect of CS extract on RANKL/RANK expression and that of exogenous RANKL on the ubiquitin-proteasome pathway in C2C12 myotubes were investigated in vitro. Long-term CS exposure induced skeletal muscle dysfunction and atrophy together with upregulation of RANKL/RANK expression in a well-established mouse model of COPD. RANKL neutralization prevented skeletal muscle dysfunction and atrophy. RANKL inhibition decreased expressions of myostatin and MuRF1/Atrogin1 and suppressed the NF-κb pathway in skeletal muscles from CS-exposed mice. In in vitro experiments with C2C12 myotubes, CS extract induced expression of RANKL/RANK, and exogenous RANKL induced activation of the ubiquitin-proteasome pathway and NF-κb pathway via RANK. Our results revealed an important role of the RANKL/RANK pathway in muscle atrophy induced by CS exposure, suggesting that RANKL may be a potential therapeutic target in COPD-related skeletal muscle dysfunction.


Subject(s)
Muscular Atrophy/genetics , NF-kappa B/genetics , Pulmonary Disease, Chronic Obstructive/genetics , RANK Ligand/genetics , Receptor Activator of Nuclear Factor-kappa B/genetics , Animals , Antibodies, Neutralizing/pharmacology , Cell Line , Cigarette Smoking/adverse effects , Complex Mixtures/antagonists & inhibitors , Complex Mixtures/pharmacology , Disease Models, Animal , Female , Gene Expression Regulation , Hand Strength/physiology , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle Strength/drug effects , Muscle Strength/genetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Atrophy/prevention & control , Myogenin/genetics , Myogenin/metabolism , Myostatin/genetics , Myostatin/metabolism , NF-kappa B/metabolism , Pulmonary Disease, Chronic Obstructive/chemically induced , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/prevention & control , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...