Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 360
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 299(2): 102889, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36634847

RESUMEN

Osteoporosis is a chronic skeletal condition characterized by low bone mass and deteriorated microarchitecture of bone tissue and puts tens of millions of people at high risk of fractures. New therapeutic agents like i-bodies, a class of next-generation single-domain antibodies, are needed to overcome some limitations of conventional treatments. An i-body is a human immunoglobulin scaffold with two long binding loops that mimic the shape and position of those found in shark antibodies, the variable new antigen receptors of sharks. Its small size (∼12 kDa) and long binding loops provide access to drug targets, which are considered undruggable by traditional monoclonal antibodies. Here, we have successfully identified a human receptor activator of nuclear factor-κB ligand (RANKL) i-body, ADR3, which demonstrates a high binding affinity to human RANKL (hRANKL) with no adverse effect on the survival or proliferation of bone marrow-derived macrophages. Differential scanning fluorimetry suggested that ADR3 is stable and able to tolerate a wide range of physical environments (including both temperature and pH). In addition, in vitro studies showed a dose-dependent inhibitory effect of ADR3 on osteoclast differentiation, podosome belt formation, and bone resorption activity. Further investigation on the mechanism of action of ADR3 revealed that it can inhibit hRANKL-mediated signaling pathways, supporting the in vitro functional observations. These clues collectively indicate that hRANKL antagonist ADR3 attenuates osteoclast differentiation and bone resorption, with the potential to serve as a novel therapeutic to protect against bone loss.


Asunto(s)
Resorción Ósea , Osteoclastos , Ligando RANK , Anticuerpos de Dominio Único , Humanos , Resorción Ósea/genética , Resorción Ósea/metabolismo , Diferenciación Celular/genética , Macrófagos/citología , Macrófagos/metabolismo , Osteoclastos/citología , Ligando RANK/metabolismo , Transducción de Señal , Anticuerpos de Dominio Único/metabolismo
2.
J Cell Physiol ; 239(5): e31214, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38358001

RESUMEN

Alleviating bone loss is an essential way to prevent osteoporotic fractures. Proper exercise improves bone density without the side effects of long-term medications, but the mechanism is unclear. Our study explored the role of Antxr1/LncRNA H19/Wnt/ß-catenin axis in the process of exercise-mediated alleviation of bone loss. Here we discovered that moderate-intensity treadmill exercise alleviates bone loss caused by ovariectomy and ameliorates bone strength accompanied by an increased lncRNA H19 expression. Concomitantly, Antxr1, a mechanosensitive protein was found downregulated by exercise but upregulated by ovariectomy. Interestingly, knockdown expression of Antxr1 increased lncRNA H19 expression and Wnt/ß-catenin signaling pathway in bone marrow mesenchymal stem cells, whereas overexpression of Antxr1 decreased lncRNA H19 expression and Wnt/ß-catenin signaling pathway. Hence, our study demonstrates the regulation of Antxr1/LncRNA H19/Wnt/ß-catenin axis in the process of mechanical strain-induced osteogenic differentiation, which provides further mechanistic insight into the role of mechanical regulation in bone metabolism.


Asunto(s)
Proteínas de Microfilamentos , Osteogénesis , ARN Largo no Codificante , Receptores de Superficie Celular , Estrés Mecánico , Vía de Señalización Wnt , beta Catenina , Animales , Femenino , Ratones , beta Catenina/metabolismo , beta Catenina/genética , Densidad Ósea/genética , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/genética , Osteoporosis/genética , Osteoporosis/metabolismo , Osteoporosis/patología , Ovariectomía/efectos adversos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Vía de Señalización Wnt/genética , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Receptores de Superficie Celular/metabolismo
3.
Environ Res ; 242: 117820, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38048867

RESUMEN

Engineered nanomaterials (ENMs) are inevitably released into the environment with the exponential application of nanotechnology. Parts of ENMs eventually accumulate in the soil environment leading to potential adverse effects on soil ecology, crop production, and human health. Therefore, the safety application of ENMs on soil has been widely discussed in recent years. More detailed safety information and potential soil environmental risks are urgently needed. However, most of the studies on the environmental effects of metal-based ENMs have been limited to single-species experiments, ecosystem processes, or abiotic processes. The present review formulated the source and the behaviors of the ENMs in soil, and the potential effects of single and co-exposure ENMs on soil microorganisms, soil fauna, and plants were introduced. The toxicity mechanism of ENMs to soil organisms was also reviewed including oxidative stress, the release of toxic metal ions, and physical contact. Soil properties affect the transport, transformation, and toxicity of ENMs. Toxic mechanisms of ENMs include oxidative stress, ion release, and physical contact. Joint toxic effects occur through adsorption, photodegradation, and loading. Besides, future research should focus on the toxic effects of ENMs at the food chain levels, the effects of ENMs on plant whole-lifecycle, and the co-exposure and long-term toxicity effects. A fast and accurate toxicity evaluation system and model method are urgently needed to solve the current difficulties. It is of great significance for the sustainable development of ENMs to provide the theoretical basis for the ecological risk assessment and environmental management of ENMs.


Asunto(s)
Ecosistema , Nanoestructuras , Humanos , Suelo , Nanoestructuras/toxicidad , Nanotecnología , Plantas
4.
Cell Mol Life Sci ; 80(6): 171, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37261512

RESUMEN

Imbalance of bone homeostasis induces bone degenerative diseases such as osteoporosis. Hedgehog (Hh) signaling plays critical roles in regulating the development of limb and joint. However, its unique role in bone homeostasis remained largely unknown. Here, we found that canonical Hh signaling pathway was gradually augmented during osteoclast differentiation. Genetic inactivation of Hh signaling in osteoclasts, using Ctsk-Cre;Smof/f conditional knockout mice, disrupted both osteoclast formation and subsequent osteoclast-osteoblast coupling. Concordantly, either Hh signaling inhibitors or Smo/Gli2 knockdown stunted in vitro osteoclast formation. Mechanistically, Hh signaling positively regulated osteoclast differentiation via transactivation of Traf6 and stabilization of TRAF6 protein. Then, we identified connective tissue growth factor (CTGF) as an Hh-regulatory bone formation-stimulating factor derived from osteoclasts, whose loss played a causative role in osteopenia seen in CKO mice. In line with this, recombinant CTGF exerted mitigating effects against ovariectomy induced bone loss, supporting a potential extension of local rCTGF treatment to osteoporotic diseases. Collectively, our findings firstly demonstrate that Hh signaling, which dictates osteoclast differentiation and osteoclast-osteoblast coupling by regulating TRAF6 and CTGF, is crucial for maintaining bone homeostasis, shedding mechanistic and therapeutic insights into the realm of osteoporosis.


Asunto(s)
Enfermedades Óseas Metabólicas , Resorción Ósea , Osteoporosis , Femenino , Ratones , Animales , Osteoclastos/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Transducción de Señal , Osteoporosis/genética , Osteoporosis/metabolismo , Homeostasis , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/metabolismo , Diferenciación Celular , Resorción Ósea/metabolismo
5.
Phytother Res ; 38(4): 1971-1989, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38358727

RESUMEN

BACKGROUND AND AIM: Osteoporosis, a systemic metabolic bone disease, is characterized by the decline of bone mass and quality due to excessive osteoclast activity. Currently, drug-targeting osteoclasts show promising therapy for osteoporosis. In this study, we investigated the effect of cichoric acid (CA) on receptor activator of nuclear kappa-B ligand (RANKL)-induced osteoclastogenesis and the bone loss induced by ovariectomy in mice. EXPERIMENTAL PROCEDURE: Molecular docking technologies were employed to examine the interaction between CA and RANKL. CCK8 assay was used to evaluate the cell viability under CA treatment. TRAcP staining, podosome belt staining, and bone resorption assays were used to test the effect of CA on osteoclastogenesis and osteoclast function. Further, an OVX-induced osteoporosis mice model was employed to identify the effect of CA on bone loss using micro-CT scanning and histological examination. To investigate underlying mechanisms, network pharmacology was applied to predict the downstream signaling pathways, which were verified by Western blot and immunofluorescence staining. KEY RESULTS: The molecular docking analysis revealed that CA exhibited a specific binding affinity to RANKL, engaging multiple binding sites. CA inhibited RANKL-induced osteoclastogenesis and bone resorption without cytotoxic effects. Mechanistically, CA suppressed RANKL-induced intracellular reactive oxygen species, nuclear factor-kappa B, and mitogen-activated protein kinase pathways, followed by abrogated nuclear factor activated T-cells 1 activity. Consistent with this finding, CA attenuated post-ovariectomy-induced osteoporosis by ameliorating osteoclastogenesis. CONCLUSIONS AND IMPLICATIONS: CA inhibited osteoclast activity and bone loss by targeting RANKL. CA might represent a promising candidate for treating osteoclast-related diseases, such as osteoporosis.


Asunto(s)
Resorción Ósea , Ácidos Cafeicos , Osteoporosis , Succinatos , Animales , Femenino , Humanos , Ratones , Resorción Ósea/prevención & control , Diferenciación Celular , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , FN-kappa B/metabolismo , Osteoclastos , Osteogénesis , Osteoporosis/patología , Ovariectomía/efectos adversos , Ligando RANK/metabolismo
6.
J Environ Manage ; 354: 120429, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38387344

RESUMEN

During the COVID-19 pandemic, an abundance of plastic face masks has been consumed and disposed of in the environment. In addition, substantial amounts of plastic mulch film have been used in intensive agriculture with low recovery. Butyl benzyl phthalate (BBP) and TiO2 nanomaterials (nTiO2) are widely applied in plastic products, leading to the inevitable release of BBP and nTiO2 into the soil system. However, the impact of co-exposure of BBP and nTiO2 at low concentrations on earthworms remains understudied. In the present study, transcriptomics was applied to reveal the effects of individual BBP and nTiO2 exposures at a concentration of 1 mg kg-1, along with the combined exposure of BBP and nTiO2 (1 mg kg-1 BBP + 1 mg kg-1 nTiO2 (anatase)) on Metaphire guillelmi. The result showed that BBP and nTiO2 exposures have the potential to induce neurodegeneration through glutamate accumulation, tau protein, and oxidative stress in the endoplasmic reticulum and mitochondria, as well as metabolism dysfunction. The present study contributes to our understanding of the toxic mechanisms of emerging contaminants at environmentally relevant levels and prompts consideration of the management of BBP and nTiO2 within the soil ecosystems.


Asunto(s)
Nanoestructuras , Oligoquetos , Ácidos Ftálicos , Animales , Humanos , Oligoquetos/genética , Ecosistema , Pandemias , Titanio , Suelo , Perfilación de la Expresión Génica
7.
Trends Biochem Sci ; 44(9): 733-736, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31279651

RESUMEN

Heterogeneous nuclear ribonucleoprotein K (hnRNPK) is an RNA-binding protein that regulates multiple biological processes, including paraspeckles formation and cellular signal transduction. Recently, hnRNPK has been shown to interact with SINE-derived nuclear RNA localization (SIRLOIN)-containing RNAs, and orchestrate nuclear enrichment and cellular functions of long noncoding RNAs (lncRNAs). hnRNPK-lncRNAs interaction is potentially implicated in various pathogenic disorders including tumorigenesis, and Kabuki-like, Au-Kline, and Okamoto syndromes.


Asunto(s)
Fisura del Paladar/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Hidronefrosis/metabolismo , Discapacidad Intelectual/metabolismo , Hipotonía Muscular/metabolismo , Síndromes Paraneoplásicos/metabolismo , ARN Largo no Codificante/metabolismo , Anomalías Múltiples/metabolismo , Cara/anomalías , Facies , Enfermedades Hematológicas/metabolismo , Humanos , Enfermedades Vestibulares/metabolismo
8.
J Cell Physiol ; 238(7): 1478-1491, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37269472

RESUMEN

Osteohematology is an emerging research field that studies the crosstalk between hematopoietic and bone stromal cells, to elucidate the mechanisms of hematological and skeletal malignancies and diseases. The Notch is an evolutionary conserved developmental signaling pathway, with critical roles in embryonic development by controlling cell proliferation and differentiation. However, the Notch pathway is also critically involved in cancer initiation and progression, such as osteosarcoma, leukemia, and multiple myeloma. The Notch-mediated malignant cells dysregulate bone and bone marrow cells in the tumour microenvironment, resulting in disorders ranging from osteoporosis to bone marrow dysfunction. To date, the complex interplay of Notch signaling molecules in hematopoietic and bone stromal cells is still poorly understood. In this mini-review, we summarize the crosstalk between cells in bone and bone marrow and their influence under the Notch signaling pathway in physiological conditions and in tumour microenvironment.


Asunto(s)
Receptores Notch , Transducción de Señal , Humanos , Médula Ósea/metabolismo , Leucemia/patología , Mieloma Múltiple/patología , Receptores Notch/genética , Receptores Notch/metabolismo , Microambiente Tumoral
9.
J Cell Physiol ; 238(8): 1823-1835, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37334837

RESUMEN

This study was designed to determine whether the use of acetylcholinesterase inhibitors (AChEIs), a group of drugs that stimulate acetylcholine receptors and are used to treat Alzheimer's disease (AD), is associated with osteoporosis protection and inhibition of osteoclast differentiation and function. Firstly, we examined the effects of AChEIs on RANKL-induced osteoclast differentiation and function with osteoclastogenesis and bone resorption assays. Next, we investigated the impacts of AChEIs on RANKL-induced nuclear factor κB and NFATc1 activation and expression of osteoclast marker proteins CA-2, CTSK and NFATc1, and dissected the MAPK signaling in osteoclasts in vitro by using luciferase assay and Western blot. Finally, we assessed the in vivo efficacy of AChEIs using an ovariectomy-induced osteoporosis mouse model, which was analyzed using microcomputed tomography, in vivo osteoclast and osteoblast parameters were assessed using histomorphometry. We found that Donepezil and Rivastigmine inhibited RANKL-induced osteoclastogenesis and impaired osteoclastic bone resorption. Moreover, AChEIs reduced the RANKL-induced transcription of Nfatc1, and expression of osteoclast marker genes to varying degrees (mainly Donepezil and Rivastigmine but not Galantamine). Furthermore, AChEIs variably inhibited RANKL-induced MAPK signaling accompanied by downregulation of AChE transcription. Finally, AChEIs protected against OVX-induced bone loss mainly by inhibiting osteoclast activity. Taken together, AChEIs (mainly Donepezil and Rivastigmine) exerted a positive effect on bone protection by inhibiting osteoclast function through MAPK and NFATc1 signaling pathways through downregulating AChE. Our findings have important clinical implications that elderly patients with dementia who are at risk of developing osteoporosis may potentially benefit from therapy with the AChEI drugs. Our study may influence drug choice in those patients with both AD and osteoporosis.


Asunto(s)
Resorción Ósea , Osteoporosis , Ratones , Animales , Femenino , Humanos , Osteogénesis , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/uso terapéutico , Acetilcolinesterasa , Rivastigmina/farmacología , Rivastigmina/uso terapéutico , Donepezilo/farmacología , Donepezilo/uso terapéutico , Microtomografía por Rayos X , Resorción Ósea/genética , Osteoclastos/metabolismo , Factores de Transcripción , FN-kappa B/metabolismo , Osteoporosis/etiología , Ligando RANK/metabolismo , Factores de Transcripción NFATC/metabolismo , Diferenciación Celular , Ovariectomía/efectos adversos
10.
J Transl Med ; 21(1): 839, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37993937

RESUMEN

BACKGROUND: Activated osteoclasts cause excessive bone resorption, and disrupt bone homeostasis, leading to osteoporosis. The extracellular signal-regulated kinase (ERK) signaling is the classical pathway related to osteoclast differentiation, and mitochondrial reactive oxygen species are closely associated with the differentiation of osteoclasts. Myrislignan (MRL), a natural product derived from nutmeg, has multiple pharmacological activities; however, its therapeutic effect on osteoporosis is unclear. Here, we investigated whether MRL could inhibit osteoclastogenesis and bone mass loss in an ovariectomy mouse model by suppressing mitochondrial function and ERK signaling. METHODS: Tartrate-resistant and phosphatase (TRAP) and bone resorption assays were performed to observe the effect of MRL on osteoclastogenesis of bone marrow macrophages. MitoSOX RED and tetramethyl rhodamine methyl ester (TMRM) staining was performed to evaluate the inhibitory effect of MRL on mitochondria. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) assay was performed to detect whether MRL suppressed the expression of osteoclast-specific genes. The impact of MRL on the protein involved in the mitogen-activated protein kinase (MAPK) and nuclear factor-κB pathways was evaluated using western blotting. In addition, a specific ERK agonist LM22B-10, was used to revalidate the inhibitory effect of MRL on ERK. Finally, we established an ovariectomy mouse model to assess the therapeutic effect of MRL on osteoporosis in vivo. RESULTS: MRL inhibited osteoclast differentiation and the associated bone resorption, by significantly decreasing osteoclastic gene expression. Mechanistically, MRL inhibited the phosphorylation of ERK by suppressing the mitochondrial function, thereby downregulating the nuclear factor of activated T cells 1 (NFATc1) signaling. LM22B-10 treatment further verified the targeted inhibition effect of MRL on ERK. Microscopic computed tomographic and histologic analyses of the tibial tissue sections indicated that ovariectomized mice had lower bone mass and higher expression of ERK compared with normal controls. However, MRL treatment significantly reversed these effects, indicating the anti-osteoporosis effect of MRL. CONCLUSION: We report for the first time that MRL inhibits ERK signaling by suppressing mitochondrial function, thereby ameliorating ovariectomy-induced osteoporosis. Our findings can provide a basis for the development of a novel therapeutic strategy for osteoporosis.


Asunto(s)
Resorción Ósea , Osteoporosis , Humanos , Femenino , Ratones , Animales , Osteogénesis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Osteoclastos , Resorción Ósea/patología , Osteoporosis/tratamiento farmacológico , Osteoporosis/etiología , Osteoporosis/metabolismo , FN-kappa B/metabolismo , Diferenciación Celular , Ovariectomía , Ligando RANK/metabolismo
11.
J Cell Physiol ; 237(1): 480-488, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34550600

RESUMEN

Leukocyte cell-derived chemotaxin-2 (LECT2 or LECT-2), also called chondromodulin II (ChM-II or CHM2) plays a versatile role in various tissues. It was first identified as a chemotactic factor to promote the migration of neutrophils. It was also reported as a hepatokine to regulate glucose metabolism, obesity, and nonalcoholic fatty liver disease. As a secreted factor, LECT2 binds to several cell surface receptors CD209a, Tie1, and Met to regulate inflammatory reaction, fibrogenesis, vascular invasion, and tumor metastasis in various cell types. As an intracellular molecule, it is associated with LECT2-mediated amyloidosis, in which LECT2 misfolding results in insoluble fibrils in multiple tissues such as the kidney, liver, and lung. Recently, LECT2 was found to be associated with the development of rheumatoid arthritis and osteoarthritis, involving the dysregulation of osteoclasts, mesenchymal stem cells, osteoblasts, chondrocytes, and endothelial cells in the bone microenvironment. LECT2 is implicated in the development of cancers, such as hepatocellular carcinoma via MET-mediated PTP1B/Raf1/ERK signaling pathways and is proposed as a biomarker. The mechanisms by which LECT2 regulates diverse pathogenic conditions in various tissues remain to be fully elucidated. Further research to understand the role of LECT2 in a tissue tropism-dependent manner would facilitate the development of LECT2 as a biomarker for diagnosis and therapeutic target.


Asunto(s)
Artritis , Neoplasias , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Artritis/genética , Artritis/metabolismo , Biomarcadores/metabolismo , Células Endoteliales/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Estructura Molecular , Neoplasias/genética , Neoplasias/metabolismo , Microambiente Tumoral
12.
J Cell Physiol ; 237(3): 1711-1719, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34893976

RESUMEN

Siglec-15, a Siglec family member and type-1 transmembrane protein, is expressed mainly in human macrophages and dendritic cells. It is comprised of a lysine-containing transmembrane domain, two extracellular immunoglobulin (Ig)-like domains and a short cytoplasmic domain. Siglec-15 is highly conserved in vertebrates and acts as an immunoreceptor. It exerts diverse functions on osteoclast physiology as well as the tumor microenvironment. Siglec-15 interacts with adapter protein DAP12 - Syk signaling pathway to regulate the RANKL/RANK-mediated PI3K, AKT, and ERK signaling pathways during osteoclast formation in vitro. Consistently, the lack of the Siglec-15 gene in mice leads to impaired osteoclast activity and osteopetrosis in vivo. In addition, Siglec-15 is expressed by tumor-associated macrophages (TAMs) and regulates the tumor microenvironment by activating the SYK/MAPK signaling pathway. Interestingly, Siglec-15 shares sequence homology to programmed death-ligand 1 (PD-L1) and has a potential immune-regulatory role in cancer immunology. Thus, Siglec-15 might also represent an alternative target for the treatment of cancers that do not respond to anti-PD-L1/PD-1 immunotherapy. Understanding the role of Siglec-15 in osteoclastogenesis and the tumor microenvironment will help us to develop new treatments for bone disorders and cancer.


Asunto(s)
Inmunoglobulinas , Neoplasias , Animales , Biología , Inmunoglobulinas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Estructura Molecular , Neoplasias/metabolismo , Osteoclastos/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Microambiente Tumoral/genética
13.
J Cell Physiol ; 237(3): 1790-1803, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34796915

RESUMEN

Excessive activity of osteoclasts contributes to skeletal diseases such as osteoporosis and osteolysis. However, current drugs targeting osteoclast have various deficiencies, placing natural compounds as substitutions of great potential. Roburic acid (RA) is a triterpenoid exacted from Radix Gentianae Macrophyllae, which exhibits inhibitory effects on inflammation and oxidation. By employing an in vitro osteoclastogenesis model, this study investigates the effects and mechanisms of RA on intracellular signaling induced by receptor activator of nuclear factor-κB ligand (RANKL). As expected, RA at a concentration scope from 1 to 10 µM dampened the osteoclast differentiation of bone marrow macrophages (BMMs) but without cell toxicity. Interestingly, RA showed no effect on osteoblastogenesis in vitro. Furthermore, RA mitigated F-actin ring formation, hydroxyapatite resorption, and gene expression in osteoclasts. Mechanistically, RA suppressed TNF receptor-associated factor 6 (TRAF6), the crucial adaptor protein following RANKL-RANK binding. On the one hand, RA downregulated the nuclear factor-κB (NF-κB) activity, extracellular regulated protein kinases (ERK) phosphorylation, and calcium oscillations. On the other hand, RA upregulated the antioxidative response element (ARE) response and the protein expression of heme oxygenase (HO)-1. These upstream alterations eventually led to the suppression of the nuclear factor of activated T cells 1 (NFATc1) activity and the expression of proteins involved in osteoclastogenesis and bone resorption. Furthermore, by using an ovariectomized (OVX) mice model, RA was found to have therapeutic effects against bone loss. On account of these findings, RA could be used to restrain osteoclasts for treating osteoporosis and other osteolytic diseases.


Asunto(s)
Resorción Ósea , Osteoporosis , Animales , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/metabolismo , Señalización del Calcio , Diferenciación Celular , Femenino , Humanos , Ratones , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Osteoclastos/metabolismo , Osteogénesis , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Ovariectomía , Ligando RANK/metabolismo , Ligando RANK/farmacología
14.
Anal Chem ; 94(40): 13889-13896, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36189785

RESUMEN

Subcellular partitioning of therapeutic agents is highly relevant to their interactions with target molecules and drug efficacy, but studying subcellular partitioning is an enormous challenge. Here, we describe the application of nanoscale secondary ion mass spectrometry (NanoSIMS) analysis to define the subcellular pharmacokinetics of a cytotoxic chemotherapy drug, arsenic trioxide (ATO). We reasoned that defining the partitioning of ATO would yield valuable insights into the mechanisms underlying ATO efficacy. NanoSIMS imaging made it possible to define the intracellular fate of ATO in a label-free manner─and with high resolution and high sensitivity. Our studies of ATO-treated cells revealed that arsenic accumulates in the nucleolus. After prolonged ATO exposure, ∼40 nm arsenic- and sulfur-rich protein aggregates appeared in the cell nucleolus, nucleus, and membrane-free compartments in the cytoplasm, and our studies suggested that the partitioning of nanoscale aggregates could be relevant to cell survival. All-trans retinoic acid increased intracellular ATO levels and accelerated the nanoscale aggregate formation in the nucleolus. This study yielded fresh insights into the subcellular pharmacokinetics of an important cancer therapeutic agent and the potential impact of drug partitioning and pharmacokinetics on drug activity.


Asunto(s)
Antineoplásicos , Arsénico , Arsenicales , Leucemia Promielocítica Aguda , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Arsénico/farmacología , Trióxido de Arsénico/farmacología , Trióxido de Arsénico/uso terapéutico , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Óxidos , Agregado de Proteínas , Azufre , Tretinoina/farmacología , Tretinoina/uso terapéutico
15.
Pharmacol Res ; 184: 106400, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35988868

RESUMEN

BACKGROUND AND OBJECTIVE: Bone loss occurs in several inflammatory diseases because of chronic persistent inflammation that activates osteoclasts (OCs) to increase bone resorption. Currently available antiresorptive drugs have severe side effects or contraindications. Herein, we explored the effects and mechanism of Alpinetin (Alp) on receptor activator of nuclear factor κB ligand (RANKL)-mediated OCs differentiation, function, and in inflammatory osteolysis of mice. METHOD: Primary mouse bone marrow-derived macrophages (BMMs) induced by RANKL and macrophage colony-stimulating factor (M-CSF) were utilized to test the impact of Alp on OCs differentiation, function, and intracellular reactive oxygen species (ROS) production, respectively. Expression of oxidant stress relevant factors and OCs specific genes were assessed via real-time quantitative PCR. Further, oxidative stress-related factors, NF-κB, MAPK, PI3K/AKT/GSK3-ß, and NFATc1 pathways were examined via Western blot. Finally, LPS-induced mouse calvarial osteolysis was used to investigate the effect of Alp on inflammatory osteolysis in vivo. RESULT: Alp suppressed OCs differentiation and resorption function, and down-regulated the ROS production. Alp inhibited IL-1ß, TNF-α and osteoclast-specific gene transcription. It also blocked the gene and protein expression of Nox1 and Keap1, but enhanced Nrf2, CAT, and HO-1 protein levels. Additionally, Alp suppressed the phosphorylation of PI3K and P38, and restrained the expression of osteoclast-specific gene Nfatc1 and its auto-amplification, hence minimizing LPS-induced osteolysis in mice. CONCLUSION: Alp is a novel candidate or therapeutics for the osteoclast-associated inflammatory osteolytic ailment.


Asunto(s)
Conservadores de la Densidad Ósea , Osteólisis , Animales , Conservadores de la Densidad Ósea/farmacología , Diferenciación Celular , Flavanonas , Glucógeno Sintasa Quinasa 3/metabolismo , Inflamación/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Lipopolisacáridos/farmacología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Osteoclastos , Osteólisis/inducido químicamente , Osteólisis/tratamiento farmacológico , Oxidantes/metabolismo , Oxidantes/farmacología , Oxidantes/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ligando RANK/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
16.
Biochem Genet ; 60(3): 843-867, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34689290

RESUMEN

Nuclear enriched abundant transcript 1 (NEAT1), consisting of two kinds of lncRNAs of 3.7 kB NEAT1-1 and 23 kB NEAT1-2, can be highly expressed in organs and tissues such as the ovary, prostate, colon, and pancreas, and is involved in paraspeckle formation and mRNA editing and gene expression. Therefore, NEAT1 is a potential biomarker for the treatment of a variety of diseases, which may be caused by two factors (isoforms of NEAT1 and NEAT1 sponging miRNA as ceRNA). However, there is still much confusion about the mechanism and downstream effector between the abnormal expression of NEAT1 and various diseases. This review summarizes recent research progress on NEAT1 in cancer and other pathologies and provides a more reliable theoretical basis for the treatment of related diseases.


Asunto(s)
MicroARNs , Neoplasias , ARN Largo no Codificante , Femenino , Humanos , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
17.
J Cell Physiol ; 236(9): 6090-6100, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33559242

RESUMEN

Ankylosing spondylitis (AS) is inflammatory arthritis predominantly affecting the spine, which is involved in the disorders of both immune and skeletal systems. The exact pathogenesis of AS is not fully understood. Osteoimmunology is a new subject of study in inflammatory arthritis, in particular the pathogenic events involved in the cross-regulation of both skeletal and immune systems. In this review, we discuss osteoimmunological and pathological changes of AS in the spine that are characterized by altered osteogenesis and osteolytic bone destruction, accompanied by the changes of the immune system. It was revealed that bone cells like mesenchymal stem cells, osteoblast, and osteoclast in crossing talking with immune cells such as T cells, B cells coregulate to the pathogenesis of AS. Further, an array of cytokines and molecules expressed by both skeletal and immune systems contribute to these complex interplays. Understanding the cellular and molecular mechanisms underlying the pathogenesis of AS will lay a foundation for the exploration of the potential new treatment to AS.


Asunto(s)
Columna Vertebral/inmunología , Columna Vertebral/patología , Espondilitis Anquilosante/inmunología , Espondilitis Anquilosante/patología , Animales , Humanos , Inmunomodulación , Inmunoterapia , Modelos Biológicos , Espondilitis Anquilosante/terapia
18.
J Cell Physiol ; 236(6): 4207-4215, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33241559

RESUMEN

SNX10 is a member of the phox homology domain-containing family of phosphoinositide-binding proteins. Intracellularly, SNX10 localizes to endosomes where it mediates intracellular trafficking, endosome organization, and protein localization to the centrosome and cilium. It is highly expressed in bone and the gut where it participates in bone mineral and calcium homeostasis through the regulation of osteoclastic bone resorption and gastric acid secretion, respectively. Not surprisingly, patients harboring mutations in SNX10 mutation manifest a phenotype of autosomal recessive osteopetrosis or malignant infantile osteopetrosis, which is clinically characterized by dense bones with increased cortical bone into the medullary space with bone marrow occlusion or depletion, bone marrow failure, and anemia. Accordingly, SNX10 mutant osteoclasts exhibit impaired bone resorptive capacity. Beyond the skeleton, there is emerging evidence implicating SNX10 in cancer development, metabolic disorders, inflammation, and chaperone-mediated autophagy. Understanding the structural basis through which SNX10 exerts its diverse biological functions in both cell and tissue-specific manners may therefore inform new therapeutic opportunities toward the treatment and management of SNX10-related diseases.


Asunto(s)
Endosomas/metabolismo , Neoplasias/metabolismo , Osteopetrosis/metabolismo , Nexinas de Clasificación/metabolismo , Animales , Endosomas/genética , Endosomas/patología , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Mutación , Neoplasias/genética , Neoplasias/patología , Osteopetrosis/genética , Osteopetrosis/patología , Conformación Proteica , Transporte de Proteínas , Nexinas de Clasificación/química , Nexinas de Clasificación/genética , Relación Estructura-Actividad
19.
J Cell Physiol ; 236(3): 1606-1615, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32697358

RESUMEN

Osteosarcoma (OS) is a primary malignant bone tumour which usually occurs in children and adolescents. OS is primarily a result of chromosomal aberrations, a combination of acquired genetic changes and, hereditary, resulting in the dysregulation of cellular functions. The Hippo signalling pathway regulates cell and tissue growth by modulating cell proliferation, differentiation, and migration in developing organs. Mammalian STE20-like 1/2 (MST1/2) protein kinases are activated by neurofibromatosis type 2, Ras association domain family member 2, kidney and brain protein, or other factors. Interactions between MST1/2 and salvador family WW domain-containing protein 1 activate large tumour suppressor kinase 1/2 proteins, which in turn phosphorylate the downstream Yes-associated protein 1/transcriptional coactivator with PDZ-binding motif (YAP/TAZ). Moreover, dysregulation of this pathway can lead to aberrant cell growth, resulting in tumorigenesis. Interestingly, small molecules targeting the Hippo signalling pathways, through affecting YAP/TAZ cellular localisation and their interaction with members of the TEA/ATTS domain family of transcriptional enhancers are being developed and hold promise for the treatment of OS. This review discusses the existing knowledge about the involvement of the Hippo signalling cascade in OS and highlights several small molecule inhibitors as potential novel therapeutics.


Asunto(s)
Osteosarcoma/enzimología , Osteosarcoma/terapia , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Animales , Modelos Biológicos , Terapia Molecular Dirigida , Osteosarcoma/patología
20.
J Cell Physiol ; 236(1): 41-48, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32572962

RESUMEN

Microfibrillar-associated proteins (MFAPs) are extracellular matrix glycoproteins, which play a role in microfibril assembly, elastinogenesis, and tissue homeostasis. MFAPs consist of five subfamily members, including MFAP1, MFAP2, MFAP3, MFAP4, and MFAP5. Among these, MFAP2 and MFAP5 are most closely related, and exhibit very limited amino acid sequence homology with MFAP1, MFAP3, and MFAP4. Gene expression profiling analysis reveals that MFAP2, MFAP5, and MFAP4 are specifically expressed in osteoblastic like cells, whereas MFAP1 and MFAP3 are more ubiquitously expressed, indicative of their diverse role in the tropism of tissues. Molecular structural analysis shows that each MFAP family member has distinct features, and functional evidence reveals discrete purposes of individual MFAPs. Animal studies indicate that MFAP2-deficient mice exhibit progressive osteopenia with elevated receptor activator of NF-κB ligand (RANKL) expression, whereas MFAP5-deficient mice are neutropenic, and MFAP4-deficient mice displayed emphysema-like pathology and the impaired formation of neointimal hyperplasia. Emerging data also suggest that MFAPs are involved in cancer progression and fat metabolism. Further understanding of tissue-specific pathophysiology of MFAPs might offer potential novel therapeutic targets for related diseases, such as skeletal and metabolic disorders, and cancers.


Asunto(s)
Enfermedades Metabólicas/genética , Neoplasias/genética , Factores de Empalme de ARN/genética , Secuencia de Aminoácidos , Animales , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Humanos , Hiperplasia/genética , Neointima/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA