Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Drug Des Devel Ther ; 15: 4741-4757, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34848946

RESUMO

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.


Assuntos
Anti-Inflamatórios/farmacologia , Reabsorção Óssea/tratamento farmacológico , Lipopolissacarídeos/antagonistas & inibidores , Ácido Oleanólico/análogos & derivados , Ligante RANK/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Saponinas/farmacologia , Animais , Reabsorção Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ácido Oleanólico/farmacologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo
2.
Drug Deliv ; 28(1): 620-633, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33779441

RESUMO

Minocycline hydrochloride (MINO) has been one of the most frequently used antibiotics in the treatment of periodontitis due to its antibacterial activity and osteogenesis effects; however, high levels of MINO administered during the treatment halt the formation of new bone. Therefore, the purpose of the present study was to prepare a MINO-microsphere/sucrose acetate isobutyrate (SAIB) hybrid depot to reduce the burst release of MINO and ensure antibacterial and osteogenesis effects of MINO in the treatment of periodontitis. Uniform microspheres, approximately 5 µm size, with a slightly rough surface and different MINO loading (10, 12, and 14%) were prepared, and the microspheres were added into SAIB, after which the burst release significantly decreased from 66.18 to 2.92%, from 71.82 to 3.82%, and from 73.35 to 4.45%, respectively, and the release from all the MINO-microspheres/SAIB hybrid depots lasted for 77 days. In addition, cytotoxicity test showed that the MINO-microsphere with 12% drug loading promoted the proliferation of osteoblasts the most and was subsequently used in vivo experiments. Moreover, in the model of ligatured-induced periodontitis in SD rats, the MINO-microsphere/SAIB hybrid depot not only significantly increased the alveolar bone height and bone volume but also reduced the inflammation of the periodontal tissue. Additionally, it also inhibited the expression of the receptor activator of nuclear factor-kappa B ligand (RANKL) and promoted the expression of osteoprotegerin (OPG).. These results indicated that the MINO-microsphere/SAIB hybrid depot might be promising in the treatment of periodontitis.


Assuntos
Antibacterianos/farmacologia , Implantes de Medicamento/farmacologia , Microesferas , Minociclina/farmacologia , Periodontite/tratamento farmacológico , Sacarose/análogos & derivados , Animais , Antibacterianos/administração & dosagem , Química Farmacêutica , Preparações de Ação Retardada , Implantes de Medicamento/administração & dosagem , Implantes de Medicamento/química , Liberação Controlada de Fármacos , Minociclina/administração & dosagem , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteoprotegerina/biossíntese , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Ratos , Ratos Sprague-Dawley , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Sacarose/química
3.
Br J Haematol ; 193(6): 1034-1043, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33249579

RESUMO

Bone-modifying therapies are essential in the treatment of patients with multiple myeloma. Zoledronic acid is preferred over other bisphosphonates due to its superiority in reducing the incidence of skeletal-related events and improving survival. The anti-receptor activator of nuclear factor-κΒ ligand (RANKL)-targeted agent denosumab has shown its non-inferiority compared to bisphosphonates in preventing skeletal-related events among newly diagnosed patients with myeloma bone disease. Denosumab may confer a survival benefit in patients eligible for autologous transplantation. Denosumab may present a safer profile for patients with renal impairment. Discontinuation of bone-directed therapies can be considered for patients with deep responses and after an adequate time period on treatment; however, a rebound effect may become evident especially in the case of denosumab. Three-monthly infusions of zoledronic acid or at-home denosumab administration should be considered during the coronavirus disease 2019 (COVID-19) pandemic. Measures to prevent hypocalcaemia, renal toxicity and osteonecrosis of the jaw are important for all bone-modifying agents.


Assuntos
Conservadores da Densidade Óssea/efeitos adversos , Denosumab/efeitos adversos , Difosfonatos/efeitos adversos , Mieloma Múltiplo/tratamento farmacológico , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , COVID-19/complicações , Denosumab/uso terapêutico , Difosfonatos/uso terapêutico , Humanos , Hipercalcemia/complicações , Hipercalcemia/tratamento farmacológico , Mieloma Múltiplo/complicações , Osteólise/complicações , Osteólise/tratamento farmacológico , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Insuficiência Renal/complicações , Insuficiência Renal/tratamento farmacológico , Ácido Zoledrônico/uso terapêutico
4.
Eur J Pharmacol ; 879: 173129, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32339516

RESUMO

The main aims of this study were: (1) to investigate whether a blockade of the interaction between the receptor activator of nuclear factor-κB (NF-ĸB) ligand (RANKL) and its receptor RANK may have potential as a novel therapeutic strategy for allergic asthma; (2) to compare the efficacies of the blockade of RANKL/RANK interaction as well as the blockade of NF-κB inhibitor kinase (IKK) and of NF-κB translocation to the nucleus, also in comparison with glucocorticosteroid treatment, in terms of the development of a mouse model of allergic airway inflammation (AAI) and accompanying immune response. The blockade of each of the targets fully prevented the development of AAI. All the tested therapeutic strategies seemed to have a certain advantage over glucocorticosteroids with regard to counteracting the development of AAI. Prevention of the activation and clonal expansion of CD4+ effector T (Teff) cells in the mediastinal lymph nodes (MLNs) constitutes a fundamental event underlying the anti-asthmatic effect induced by the blockade of IKK, NF-κB translocation or of RANKL/RANK interaction. The results indicate that attenuation of the CD11b+CD103-CD11chigh dendritic cell response in the MLNs is an initial but not the main mechanism responsible for this effect. In turn, the direct anti-proliferative action on CD4+ Teff cells seems to constitute the chief mechanism responsible for the anti-asthmatic effect of all the tested therapeutic strategies. A clinical implication is that local inhibition of RANKL/RANK interaction achieved via inhalatory administration of a RANKL antagonist can be considered as a novel therapeutic strategy in treatment of allergic asthma.


Assuntos
Asma/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Ligante RANK/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Animais , Asma/imunologia , Asma/patologia , Líquido da Lavagem Broncoalveolar/citologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/patologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Camundongos Endogâmicos BALB C , NF-kappa B/imunologia , Ovalbumina , Ligante RANK/imunologia , Receptor Ativador de Fator Nuclear kappa-B/imunologia , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
5.
Curr Mol Pharmacol ; 13(3): 224-232, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31951177

RESUMO

OBJECTIVE: To explore the effect of miRNA-143 on osteoclast formation and provide new ideas for the treatment of osteoporosis. METHODS: Mice macrophage lines RAW264.7 cells after transfection were divided into four groups: control group, RANKL group, RANKL combined with miR-143 mimics group and RANKL combined with miR-NC group. TARCP staining was used to observe the effect of miR-143 on osteoclast formation. The expression of RANK, TRAF6 and NFATc-1 in the upstream of RANKL pathway was detected by real-time quantitative PCR (RT qPCR) and Western blotting (WB). The binding of miR-143 to TNFRSF11A was detected by double Luciferase Reporter Analysis. The effect of miR-143 on the expression of NF-κB (p65, I-κB-α) signal pathway in osteoclasts was detected. The effects of I-BET151 on the expression of osteoclast-specific genes TRACP, MMP 9, CtsK and c-Src were detected. RESULTS: The positive level of osteoclasts in RANKL group and RANKL combined with miR-NC group was significantly higher than that of RANKL combined with miR-143 mimics group and control group (P < 0.05). The expression levels of RANK, TRAF6, NFATc-1, TRACP, MMP-9, CtsK and c-Src in RANKL group and RANKL combined with miR-NC group were significantly higher than those of RANKL combined with miR-143 mimics group and control group (P < 0.05). The expression levels of I-κB-α were significantly lower than that of RANKL combined with miR-143 mimics group and control group (P<0.05). CONCLUSION: MiR-143 can inhibit the expression of RANK, TRAF6 and downstream NFATc-1 in the RANKL pathway, thereby inhibiting the RANK/RANKL pathway. MiR-143 can inhibit the signal pathway of NF-κB (p65, I-κB-α). MiR-143 inhibits the expression of osteoclast-specific genes TRACP, MMP 9, CtsK and c-Src. That is to say, miR-143 inhibits osteoclast formation by targeting RANK, NF- κB and MAPK signaling pathways.


Assuntos
MicroRNAs/genética , Osteoclastos/efeitos dos fármacos , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Inibidor de NF-kappaB alfa/biossíntese , Inibidor de NF-kappaB alfa/genética , Fatores de Transcrição NFATC/biossíntese , Fatores de Transcrição NFATC/genética , Osteoclastos/metabolismo , Ligante RANK/genética , Células RAW 264.7 , RNA Mensageiro/biossíntese , Receptor Ativador de Fator Nuclear kappa-B/biossíntese , Receptor Ativador de Fator Nuclear kappa-B/genética , Proteínas Recombinantes/metabolismo , Fator 6 Associado a Receptor de TNF/biossíntese , Fator 6 Associado a Receptor de TNF/genética , Fator de Transcrição RelA/biossíntese , Fator de Transcrição RelA/genética
6.
J Cell Mol Med ; 24(1): 785-798, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31725199

RESUMO

Bone homeostasis is delicately orchestrated by osteoblasts and osteoclasts. Various pathological bone loss situations result from the overactivated osteoclastogenesis. Receptor activator of nuclear factor κB ligand (RANKL)-activated NF-κB and MAPK pathways is vital for osteoclastogenesis. Here, we for the first time explored the effects of l-tetrahydropalmatine (l-THP), an active alkaloid derived from corydalis, on the formation and function of osteoclasts in vitro and in vivo. In RAW264.7 cells and bone marrow monocytes cells (BMMCs), l-THP inhibited osteoclastic differentiation at the early stage, down-regulated transcription level of osteoclastogenesis-related genes and impaired osteoclasts functions. Mechanically, Western blot showed that l-THP inhibited the phosphorylation of P50, P65, IκB, ERK, JNK and P38, and the electrophoretic mobility shift assay (EMSA) revealed that DNA binding activity of NF-κB was suppressed, ultimately inhibiting the expression of nuclear factor of activated T cells (NFATc1). Besides, Co-immunoprecipitation indicated that l-THP blocked the interactions of RANK and TNF receptor associated factor 6 (TRAF6) at an upstream site. In vivo, l-THP significantly inhibited ovariectomy-induced bone loss and osteoclastogenesis in mice. Collectively, our study demonstrated that l-THP suppressed osteoclastogenesis by blocking RANK-TRAF6 interactions and inhibiting NF-κB and MAPK pathways. l-THP is a promising agent for treating osteoclastogenesis-related diseases such as post-menopausal osteoporosis.


Assuntos
Alcaloides de Berberina/farmacologia , Reabsorção Óssea/tratamento farmacológico , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Osteogênese , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Fator 6 Associado a Receptor de TNF/antagonistas & inibidores , Animais , Antiarrítmicos/farmacologia , Diferenciação Celular , Feminino , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/metabolismo
7.
Breast Cancer Res ; 21(1): 132, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31796128

RESUMO

BACKGROUND: ERBB-2 is overexpressed in about 20% of breast cancers (BCs), indicating poor prognosis. The receptor activator of nuclear factor-κB (RANK) pathway is implicated in ERBB-2 (+) BC. The purpose of this study was to elucidate the underlying molecular mechanism of this interaction and the beneficial impact of dual targeting of RANK and ERBB-2 pathways. METHODS: We used SKBR3, MCF7, MDA-MB-453, and BT-474 human BC cell lines. We examined RANK and RANKL expression using RT-PCR, Western blot, and immunofluorescence. The evaluation of RANK expression in a cohort of BC patients was performed using immunohistochemistry. The interaction between RANK and ERBB family members was detected using proximity ligation assay (PLA), which enables the visualization of interacting proteins. We used inhibitors of both pathways [trastuzumab (T), pertuzumab (P), denosumab (D)]. NF-κB pathway activation was studied using Western blot. Cell growth and viability was evaluated using XTT, flow cytometry, and clonogenic assay. For cell migration evaluation, scratch assay was performed. Data were analyzed by one-way ANOVA. RESULTS: Cell lines express RANK and RANKL. RANK immunostaining was also detected in human BC tissue samples. RANK receptor dimerizes with ERBB family members. RANK/ERBB-2 dimer number seems to be associated with ERBB-2 expression (SKBR3, 5.4; BT-474, 8.2; MCF7, 0.7; MDA-MB-453, 0.3). RANK/ERBB-2 dimers were decreased in the presence of the inhibitors D, T, and P, while they were increased after RANKL (R) treatment in SKBR3 (m, 5.4; D, 1.2; T, 1.9; DT, 0.6; TP, 1; DTP, 0.4; R, 11.8) and BT-474 (m, 8.2; D, 3.1; T, 4.3; DT, 0.7; TP, 3.4; DTP, 3.2; R, 11.6). Combination targeting of SKBR3 further decreased NF-κB pathway activation compared to single targeting. In SKBR3, RANKL and ERBB-2 blockage resulted in reduced cell proliferation, increased apoptosis, and lower metastatic potential compared to mock cells (m) and reversed values in RANKL presence. The combination treatment of SKBR3 with D, T, and P had an advantage in functional traits compared to single targeting. Denosumab suppressed NF-κB signaling and diminished proliferation rate in MDA-MB-453 cells. MCF7 did not correspond to inhibitors. CONCLUSIONS: The results indicate a novel physical and molecular association between ERBB-2 and RANK pathways that affects ERBB-2 (+) BC growth. We also present data suggesting that the combination of anti-ERBB-2 agents and RANKL inhibitors have a potential direct anti-tumor effect and should be further tested in certain BC patients.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Neoplasias da Mama/metabolismo , Receptores ErbB/antagonistas & inibidores , Terapia de Alvo Molecular , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Células Cultivadas , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Expressão Gênica , Humanos , Imuno-Histoquímica , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais/efeitos dos fármacos
8.
Crit Rev Oncol Hematol ; 137: 1-8, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31014505

RESUMO

Bone-modifying agents like bisphosphonates and receptor activator of nuclear factor kappaß ligand (RANK-L) inhibitors are used as supportive treatments in breast cancer patients with bone metastases to prevent skeletal-related events (SREs). Due to missing head-to-head comparisons, a network meta-analysis was performed to provide a hierarchy of these therapeutic options. Through a systematic literature search, 21 randomized controlled trials (RCTs) that fulfilled the inclusion criteria were identified. To prevent SREs, the ranking through P-scores showed denosumab (RR: 0.62; 95%CI: 0.50-0.76), zoledronic acid (RR: 0.72; 95%CI: 0.61-0.84) and pamidronate (RR: 0.76; 95%CI: 0.67-0.85) to be significantly superior to placebo. Due to insufficient or heterogeneous data, overall survival, quality of life, pain response and adverse events were not able to be analyzed within the network. Although data were sparse on adverse events, the risk of significant adverse events appeared low. The results of this review can therefore be used to formulate clinical studies more precisely in order to standardise and focus on patient-relevant outcomes.


Assuntos
Conservadores da Densidade Óssea/administração & dosagem , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Neoplasias da Mama/tratamento farmacológico , Difosfonatos/administração & dosagem , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Adjuvantes Imunológicos/administração & dosagem , Neoplasias da Mama/patologia , Feminino , Humanos , Metanálise em Rede , Qualidade de Vida , Ensaios Clínicos Controlados Aleatórios como Assunto
9.
Crit Rev Oncol Hematol ; 133: 85-91, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30661662

RESUMO

Binding between the receptor activator of nuclear factor-kB (RANK) and its ligand (RANKL) triggers recruitment of TNF receptor associated factor (TRAF) adaptor proteins and activation of downstream pathways. RANK/RANKL signaling is controlled by a decoy receptor called osteoprotegerin (OPG) which interacts with RANKL. Additional networks regulating RANK/RANKL signaling are active in a context specific manner. RANK/RANKL signaling is essential for the differentiation of bone-resorbing osteoclasts, and is deregulated in pathological processes such as postmenopausal osteoporosis or cancer induced bone destruction. Cells expressing RANK and RANKL are commonly found in the tumor microenvironment. The RANKL/RANK pathway is often overexpressed in tumors of the breast, prostate, endometrium, cervix, stomach, oesophagus and bladder, thyroid and correlated with poor prognosis. RANK signaling plays an important role in the innate and adaptive immune response as it generates regulatory T (Treg) cells and increases production of cytokines. RANK expression induces chemoresistance in vitro through the activation of multiple signal transduction pathways. RANKL blockade improves the efficacy of anti-CTLA-4 monoclonal antibodies against solid tumors and experimental metastases. As RANK inhibition enhances the immune response there is an increasing interest in combining it with immune therapy in an attempt to sensitize immune resistant tumors to immune therapies. Several studies are ongoing to assess this concept. The role of RANK/RANKL inhibition should be further pursued as an immunomodulatory strategy in combination with other treatment modalities.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Denosumab/administração & dosagem , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/administração & dosagem , Ligante RANK/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Animais , Antineoplásicos Imunológicos/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Denosumab/farmacologia , Regulação para Baixo/efeitos dos fármacos , Sinergismo Farmacológico , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Inibidores de Proteínas Quinases/farmacologia , Ligante RANK/imunologia , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/imunologia , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento
10.
Tissue Eng Part A ; 25(1-2): 34-43, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29652606

RESUMO

Osteoporosis, which is characterized by low bone mineral density and susceptibility to fracture, is caused by increased osteoclastic activity. Receptor activator of nuclear factor kappa B ligand (RANKL)/RANK signaling plays an important role in osteoclast differentiation and activation. The current treatment strategies for osteoporosis do not directly address this underlying cause and generates undesired side effects. This led to emergence of controlled delivery systems to increase drug bioavailability and efficacy specifically at the bone tissue. With better understanding of molecular pathology of bone, the use of small interfering RNA (siRNA) to inhibit translation of abnormal gene expression in cells is becoming a promising approach. In this study, we report a siRNA delivery system consisting of PEI:RANK siRNA complex entrapped in nanosized poly(lactic acid-co-glycolic acid) (PLGA) capsules intended to be used in the treatment of osteoporosis. The nanosize will enable the nanoparticles to be administered by intravenous injection. The RANK siRNA was complexed with polyethylenimine (PEI) and loaded into biodegradable PLGA nanocapsules (NCs). The PEI:RANK siRNA loaded nanocapsules significantly reduced (47%) RANK mRNA levels. The differentiation of osteoclast precursors to mature osteoclasts was significantly suppressed (∼54%). The reduction in the osteoclastic activity of the differentiated osteoclasts (55%) was found to be statistically significant. The siRNA delivery system developed in the study is planned to be tested i.v. in mouse and has the potential to be used as a novel alternative approach for the systemic treatment of osteoporosis.


Assuntos
Nanocápsulas/química , Osteoporose/tratamento farmacológico , Polietilenoimina , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , RNA Interferente Pequeno , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Animais , Camundongos , Osteoporose/genética , Osteoporose/metabolismo , Osteoporose/patologia , Polietilenoimina/química , Polietilenoimina/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/farmacologia , Células RAW 264.7 , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo
11.
Nat Rev Clin Oncol ; 15(11): 676-693, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30232468

RESUMO

Recognizing that the transformative effects of immunotherapy are currently limited to a minority of patients with cancer, research efforts are increasingly focused on expanding and enhancing clinical responses by combining immunotherapies; the repurposing of existing drugs is an attractive approach, given their well-characterized safety and pharmacokinetic profiles. Receptor activator of nuclear factor-κB (RANK) and the RANK ligand (RANKL) were initially described in the context of T cell-dendritic cell interactions; however, the discovery of an obligate role of RANK signalling in osteoclastogenesis led to the development of the anti-RANKL antibody denosumab for antiresorptive indications, including bone metastases. Randomized clinical trials and post-marketing surveillance studies have established the acceptable safety profile of denosumab. More recently, several case reports involving patients with advanced-stage melanoma have described remarkable responses following concurrent treatment with denosumab and immune-checkpoint inhibitors. Randomized trials assessing similar combinations in patients with melanoma or renal cell carcinoma are now underway. Herein, we discuss the hallmark clinical trials of denosumab in light of possible immunological effects of this agent. We highlight the role of immune cells as sources of RANK and RANKL in the tumour microenvironment and review data on RANKL inhibition in mouse models of cancer. Finally, we describe hypothetical immune-related mechanisms of action, which could be assessed in clinical trials of immune-checkpoint inhibitors and denosumab in patients with cancer.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Melanoma/tratamento farmacológico , Ligante RANK/imunologia , Receptor Ativador de Fator Nuclear kappa-B/imunologia , Carcinoma de Células Renais/imunologia , Denosumab/uso terapêutico , Humanos , Melanoma/imunologia , Osteogênese/imunologia , Ligante RANK/antagonistas & inibidores , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Microambiente Tumoral/imunologia
12.
Biochem Biophys Res Commun ; 503(3): 2075-2082, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30107914

RESUMO

Adiponectin (APN) has been shown to play a key role in regulating bone mineral density (BMD). Nevertheless, the effects of APN on receptor activator of NF-κB ligand (RANKL)-induced osteoclast formation and mechanism of regulation are not entirely clear. The study, therefore, aimed to evaluate the effect of APN on osteoclastogenesis. Our results showed that APN inhibits osteoclastogenesis and resorption function in vitro by suppressing nuclear factor-κB (NF-κB) and p38 signaling pathways, which is essential for osteoclast formation. Moreover, APN blocked the formation of F-actin rings and attenuated osteoclast-mediated bone resorptive function. Therefore, we concluded that APN may provide a potential treatment for osteoclast-related diseases, such as osteoporosis.


Assuntos
Adiponectina/farmacologia , NF-kappa B/antagonistas & inibidores , Osteogênese/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Adiponectina/administração & dosagem , Animais , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Camundongos , NF-kappa B/metabolismo , Osteoclastos/efeitos dos fármacos , Células RAW 264.7 , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Praxis (Bern 1994) ; 107(12): 649-654, 2018 Jun.
Artigo em Francês | MEDLINE | ID: mdl-29871576

RESUMO

Multiple Vertebral Fractures after Denosumab Discontinuation: How to Avoid Them? Abstract. Denosumab is a monoclonal antibody raised against the RANK ligand that inhibits the maturation and activity of osteoclasts. It decreases bone resorption, increases bone density and reduces fracture risk. However, after its discontinuation, a significant rebound effect appears that lasts about two years. It results in increased markers of bone remodeling, a loss of bone density that may be greater than gain, and an increased risk of multiple vertebral fractures. These fractures occur at a frequency of 1 to 10 %. Due to this high risk, denosumab should be a second-line treatment limited to very specific indications. At denosumab discontinuation, in order to limit the rebound effect, the current recommendation is to prescribe a strong bisphosphonate (alendronate, zoledronate) and regularly monitor the bone resorption markers.


Assuntos
Denosumab/efeitos adversos , Fraturas Múltiplas/induzido quimicamente , Fraturas da Coluna Vertebral/induzido quimicamente , Síndrome de Abstinência a Substâncias/etiologia , Densidade Óssea/efeitos dos fármacos , Remodelação Óssea/efeitos dos fármacos , Reabsorção Óssea/induzido quimicamente , Denosumab/uso terapêutico , Fraturas Múltiplas/diagnóstico , Fraturas Múltiplas/prevenção & controle , Humanos , Imageamento por Ressonância Magnética , Osteoclastos/efeitos dos fármacos , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Fatores de Risco , Fraturas da Coluna Vertebral/diagnóstico , Fraturas da Coluna Vertebral/prevenção & controle , Síndrome de Abstinência a Substâncias/diagnóstico , Síndrome de Abstinência a Substâncias/prevenção & controle
14.
Curr Top Med Chem ; 18(8): 661-673, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29875003

RESUMO

The overexpression of Tumor Necrosis Factor (TNF) is directly related to the development of several autoimmune diseases, such as rheumatoid and psoriatic arthritis, inflammatory bowel disease, Crohn's disease, refractory asthma, and multiple sclerosis. Receptor Activator of Nuclear Factor Kappa- B Ligand (RANKL) belongs to the TNF family and is the primary mediator of osteoclast-induced bone resorption through interaction with its receptor RANK. The function of RANKL is physiologically inhibited by the action of osteoprotegerin (OPG), which is a decoy receptor that binds to RANKL and prevents the process of osteoclastogenesis. Malfunction among RANK/RANKL/OPG can also result in bone loss diseases, including postmenopausal osteoporosis, rheumatoid arthritis, bone metastasis and multiple myeloma. To disrupt the unwanted functions of TNF and RANKL, current attempts focus on blocking TNF and RANKL binding to their receptors. In this review, we present the research efforts toward the development of low-molecular-weight pharmaceuticals that directly block the detrimental actions of TNF and RANKL.


Assuntos
Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Inibidores do Fator de Necrose Tumoral , Animais , Humanos , Ligantes , Modelos Moleculares , Peso Molecular , Ligação Proteica/efeitos dos fármacos , Receptor Ativador de Fator Nuclear kappa-B/química , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/química , Fatores de Necrose Tumoral/química
15.
Bioorg Chem ; 78: 94-102, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29550534

RESUMO

A total of twenty-two novel coumarin triazole hybrids (4a-4k and 6a-6k) were synthesized from orcinol in good to excellent yields of 70-94%. The structures of all the synthesized compounds were elucidated by spectroscopic techniques such as 1H NMR, 13C NMR, and HRMS. The anti-inflammatory potential of synthesized compounds was investigated against the proinflammatory cytokine, TNF-α on U937 cell line and compounds 4d, 4j, and 6j were found to exhibit promising anti-inflammatory activity. These three compounds were further screened against TNF-α on LPS-stimulated RAW 264.7 cells, which confirm their anti-inflammatory potential. Furthermore, the above said active compounds were tested for their inhibitory effect on RANKL-induced osteoclastogenesis in RAW 264.7 cells by using tartrate resistant acid phosphatase (TRAP) staining assay at 10 µM. Molecular mechanism studies demonstrated that compound 4d exhibited dose dependent inhibition of RANKL-induced osteoclastogenesis by suppression of the NF-kB pathway. Thus, compound 4d is a promising candidate for further optimization to develop as a potent anti-osteoporotic agent.


Assuntos
Cumarínicos/farmacologia , NF-kappa B/antagonistas & inibidores , Osteogênese/efeitos dos fármacos , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Resorcinóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Triazóis/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Cumarínicos/síntese química , Cumarínicos/química , Relação Dose-Resposta a Droga , Humanos , Camundongos , Estrutura Molecular , NF-kappa B/metabolismo , Células RAW 264.7 , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Resorcinóis/química , Relação Estrutura-Atividade , Triazóis/síntese química , Triazóis/química , Células U937
16.
Trends Cell Biol ; 28(3): 213-223, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29241686

RESUMO

The tumor necrosis factor (TNF) receptor RANK (TNFRSF11A) and its ligand RANKL (TNFSF11) regulate osteoclast development and bone metabolism. They also control stem cell expansion and proliferation of mammary epithelial cells via the sex hormone progesterone. As such, RANKL and RANK have been implicated in the onset of hormone-induced breast cancer. Recently, RANK/RANKL were identified as crucial regulators for BRCA1 mutation-driven breast cancer. Current prevention strategies for BRCA1 mutation carriers are associated with wide-ranging risks; therefore, the search for alternative, non-invasive strategies is of paramount importance. We summarize here the functions of the RANKL/RANK pathway in mammalian physiology and focus on its recently uncovered role in breast cancer. We propose that anti-RANKL therapy should be pursued as a preventative strategy for breast cancer.


Assuntos
Proteína BRCA1/genética , Neoplasias da Mama/genética , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Células Epiteliais/metabolismo , Feminino , Humanos , Mutação , Células-Tronco Neoplásicas/metabolismo , Ligante RANK/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores
17.
Genes Dev ; 31(20): 2099-2112, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-29118048

RESUMO

Lung cancer is the leading cause of cancer deaths. Besides smoking, epidemiological studies have linked female sex hormones to lung cancer in women; however, the underlying mechanisms remain unclear. Here we report that the receptor activator of nuclear factor-kB (RANK), the key regulator of osteoclastogenesis, is frequently expressed in primary lung tumors, an active RANK pathway correlates with decreased survival, and pharmacologic RANK inhibition reduces tumor growth in patient-derived lung cancer xenografts. Clonal genetic inactivation of KRasG12D in mouse lung epithelial cells markedly impairs the progression of KRasG12D -driven lung cancer, resulting in a significant survival advantage. Mechanistically, RANK rewires energy homeostasis in human and murine lung cancer cells and promotes expansion of lung cancer stem-like cells, which is blocked by inhibiting mitochondrial respiration. Our data also indicate survival differences in KRasG12D -driven lung cancer between male and female mice, and we show that female sex hormones can promote lung cancer progression via the RANK pathway. These data uncover a direct role for RANK in lung cancer and may explain why female sex hormones accelerate lung cancer development. Inhibition of RANK using the approved drug denosumab may be a therapeutic drug candidate for primary lung cancer.


Assuntos
Neoplasias Pulmonares/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/fisiologia , Células Epiteliais Alveolares/metabolismo , Animais , Respiração Celular , Células Cultivadas , Metabolismo Energético , Feminino , Hormônios Esteroides Gonadais/fisiologia , Homeostase , Humanos , Pulmão/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Masculino , Camundongos , Mitocôndrias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Mucosa Respiratória/metabolismo
18.
ChemMedChem ; 12(20): 1697-1702, 2017 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-28885764

RESUMO

Receptor activator of NF-κB (RANK), a member of the TNF-receptor superfamily, plays an important role in bone resorption and stimulates immune and epithelial cell activation. Denosumab, a human monoclonal antibody that blocks the RANK ligand (RANKL), is approved for the treatment of osteoporosis and bone metastasis. However, a small molecule that inhibits the RANK-RANKL interaction would be beneficial to decrease cost and to facilitate treatments with orally available therapeutic agents. Herein we report the discovery of the first nonpeptidic inhibitors of RANK-RANKL interactions. In screening a chemical library by competitive ELISA, the porphyrin verteporfin was identified as a hit. Derivatives were screened, and the chlorin-macrocycle-containing pheophorbide A and purpurin 18 were found to bind recombinant RANKL, to inhibit RANK-RANKL interactions in the ELISA, and to suppress the RANKL-dependent activation of model cells and the differentiation of RANK-expressing precursors into osteoclasts. This discovery of a family of small molecules that inhibit RANK activation presents an initial basis for further development of nonpeptidic therapeutic agents targeting the interaction between RANK and RANKL.


Assuntos
Porfirinas/farmacologia , Ligante RANK/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Animais , Sobrevivência Celular , Humanos , Células Jurkat , Camundongos , Estrutura Molecular , Osteogênese/efeitos dos fármacos , Porfirinas/química , Ligação Proteica , Ligante RANK/química , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/química , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Relação Estrutura-Atividade
19.
Inflammation ; 40(4): 1177-1190, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28474155

RESUMO

RANKL and RANK are potential contributors of inflammatory cascade in human and animal model of arthritis. The current study aims to investigate the effect of N-(2-hydroxyphenyl)acetamide (NA-2) on regulation of RANKL pathway in collagen-induced arthritis (CIA) model in rats. CIA was induced using bovine type II collagen in female Wistar rats. The clinical parameters, level of pro-inflammatory and oxidative stress markers were measured to determine the progression of the disease. The mRNA level of RANKL and RANK and downstream mediators of inflammation i.e. c-fos, c-jun, NF-κB and Akt were analysed in spleen tissue using real-time PCR. Immunohistochemical analysis of iNOS, pAkt and c-Fos was also done in spleen tissue. Treatment with NA-2 and indomethacin showed increase in body weight and significant reduction in paw volume and arthritic score (p < 0.0001). Marked reduction in the level of oxidative stress markers, NO, PO and GSH (p < 0.0001), and pro-inflammatory markers, IL-1ß (p < 0.0001) and TNF-α (p < 0.01), was also observed. Likewise, NA-2 and indomethacin treatment also significantly suppressed the mRNA expression of RANKL, RANK, c-fos, c-jun, NF-κB (p < 0.0001) and Akt (p < 0.01) and protein expression of iNOS, pAkt and c-Fos (p < 0.0001) compared to the arthritic control group. Our findings suggest that NA-2 is an antiarthritic agent acting in a pleiotropic manner in CIA rats by not only reducing the clinical signs of arthritis, inflammatory cytokines and free radical production but also attenuating the RANK/RANKL signaling pathway.


Assuntos
Acetanilidas/farmacologia , Artrite/metabolismo , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Animais , Artrite/induzido quimicamente , Artrite/tratamento farmacológico , Biomarcadores/análise , Colágeno , Feminino , Mediadores da Inflamação/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ligante RANK/antagonistas & inibidores , Ratos , Ratos Wistar , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Baço/química
20.
Diabetes Obes Metab ; 18 Suppl 1: 71-7, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27615134

RESUMO

ß-Cell dysfunction in type 1 and type 2 diabetes is accompanied by a progressive loss of ß-cells, and an understanding of the cellular mechanism(s) that regulate ß-cell mass will enable approaches to enhance hormone secretion. It is becoming increasingly recognized that enhancement of human ß-cell proliferation is one potential approach to restore ß-cell mass to prevent and/or cure type 1 and type 2 diabetes. While several reports describe the factor(s) that enhance ß-cell replication in animal models or cell lines, promoting effective human ß-cell proliferation continues to be a challenge in the field. In this review, we discuss recent studies reporting successful human ß-cell proliferation including WS6, an IkB kinase and EBP1 inhibitor; harmine and 5-IT, both DYRK1A inhibitors; GNF7156 and GNF4877, GSK-3ß and DYRK1A inhibitors; osteoprotegrin and Denosmab, receptor activator of NF-kB (RANK) inhibitors; and SerpinB1, a protease inhibitor. These studies provide important examples of proteins and pathways that may prove useful for designing therapeutic strategies to counter the different forms of human diabetes.


Assuntos
Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus/prevenção & controle , Células Secretoras de Insulina/efeitos dos fármacos , Inibidores da Monoaminoxidase/farmacologia , Inibidores de Serina Proteinase/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Animais , Denosumab/farmacologia , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/metabolismo , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Harmina/farmacologia , Humanos , Quinase I-kappa B/antagonistas & inibidores , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Osteoprotegerina/farmacologia , Compostos de Fenilureia/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas de Ligação a RNA/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Serpinas/farmacologia , Triptaminas/farmacologia , Quinases Dyrk
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA