Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Intervalo de año de publicación
1.
BMC Med Genomics ; 17(1): 159, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38867275

RESUMEN

BACKGROUND: Association between glucose and inflammatory bowel disease (IBD) was found in previous observational studies and in cohort studies. However, it is not clear whether these associations reflect causality. Thus, this study investigated whether there is such a causal relation between elevated glucose and IBD, Crohn's disease (CD) and ulcerative colitis (UC). METHODS: We performed a two-sample Mendelian Randomization (MR) with the independent genetic instruments identified from the largest available genome-wide association study (GWAS) for IBD (5,673 cases; 213,119 controls) and its main subtypes, CD and UC. Summarized data for glucose which included 200,622 cases and glycemic traits including HbA1c and type 2 diabetes(T2DM) were obtained from different GWAS studies. Primary and secondary analyses were conducted by preferentially using the radial inverse-variance weighted (IVW) approach. A number of other meta-analysis approach and sensitivity analyses were carried out to assess the robustness of the results. RESULTS: We did not find a causal effect of genetically predicted glucose on IBD as a whole (OR 0.858; 95% CI 0.649-1.135; P = 0.286). In subtype analyses glucose was also suggestively not associated with Crohn's disease (OR 0.22; 95% CI 0.04-1.00; P = 0.05) and ulcerative colitis (OR 0.940; 95% CI 0.628-1.407; P = 0.762). In the other direction, IBD and its subtypes were not related to glucose and glycemic traits. CONCLUSIONS: This MR study is not providing any evidence for a causal relationship between genetically predicted elevated glucose and IBD as well as it's subtypes UC and CD. Regarding the other direction, no causal associations could be found. Future studies with robust genetic instruments are needed to confirm this conclusion.


Asunto(s)
Estudio de Asociación del Genoma Completo , Enfermedades Inflamatorias del Intestino , Análisis de la Aleatorización Mendeliana , Humanos , Enfermedades Inflamatorias del Intestino/genética , Glucemia , Polimorfismo de Nucleótido Simple , Enfermedad de Crohn/genética , Colitis Ulcerosa/genética , Diabetes Mellitus Tipo 2/genética , Predisposición Genética a la Enfermedad
2.
Biochim Biophys Acta Mol Basis Dis ; 1869(7): 166795, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37385514

RESUMEN

The senescence of bone marrow mesenchymal stem cells (BMSCs) is the basis of senile osteoporosis (SOP). Targeting BMSCs senescence is of paramount importance for developing anti-osteoporotic strategy. In this study, we found that protein tyrosine phosphatase 1B (PTP1B), an enzyme responsible for tyrosine dephosphorylation, was significantly upregulated in BMSCs and femurs with advancing chronological age. Therefore, the potential role of PTP1B in BMSCs senescence and senile osteoporosis was studied. Firstly, significantly upregulated PTP1B expression along with impaired osteogenic differentiation capacity was observed in D-galactose (D-gal)-induced BMSCs and naturally-aged BMSCs. Furthermore, PTP1B silencing could effectively alleviate senescence, improve mitochondrial dysfunction, and restore osteogenic differentiation in aged BMSCs, which was attributable to enhanced mitophagy mediated by PKM2/AMPK pathway. In addition, hydroxychloroquine (HCQ), an autophagy inhibitor, significantly reversed the protective effects from PTP1B knockdown. In SOP animal model, transplantation of LVsh-PTP1B-transfected D-gal-induced BMSCs harvested double protective effects, including increased bone formation and reduced osteoclastogenesis. Similarly, HCQ treatment remarkably suppressed osteogenesis of LVsh-PTP1B-transfected D-gal-induced BMSCs in vivo. Taken together, our data demonstrated that PTP1B silencing protects against BMSCs senescence and mitigates SOP via activating AMPK-mediated mitophagy. Targeting PTP1B may represent a promising interventional strategy to attenuate SOP.


Asunto(s)
Células Madre Mesenquimatosas , Osteoporosis , Animales , Osteogénesis , Proteínas Quinasas Activadas por AMP/metabolismo , Mitofagia , Monoéster Fosfórico Hidrolasas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteoporosis/genética , Osteoporosis/metabolismo
3.
J Nanobiotechnology ; 21(1): 125, 2023 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-37041523

RESUMEN

Osteoporosis (OP) is a metabolic bone disease characterized by decreased bone mass and increased bone fragility. The imbalance of bone homeostasis modulated by osteoclasts and osteoblasts is the most crucial pathological change in osteoporosis. As a novel treatment strategy, nanomedicine has been applied in drug delivery and targeted therapy due to its high efficiency, precision, and fewer side effects. Gold nanospheres (GNS), as a common kind of gold nanoparticles (GNPs), possess significant antimicrobial and anti-inflammatory activity, which have been applied for the treatment of eye diseases and rheumatoid arthritis. However, the effect of GNS on osteoporosis remains elusive. In this study, we found that GNS significantly prevented ovariectomy (OVX)-induced osteoporosis in a gut microbiota-dependent manner. 16S rDNA gene sequencing demonstrated GNS markedly altered the gut microbial diversity and flora composition. In addition, GNS reduced the abundance of TMAO-related metabolites in OVX mice. Low TMAO levels might alleviate the bone loss phenomenon by reducing the inflammation response. Therefore, we investigated the alteration of cytokine profiles in OVX mice. GNS inhibited the release of pro-osteoclastogenic or proinflammatory cytokines including tumor necrosis factor α (TNF-α), interleukin (IL)-6, and granulocyte colony-stimulating factor (G-CSF) in the serum. In conclusion, GNS suppressed estrogen deficiency-induced bone loss by regulating the destroyed homeostasis of gut microbiota so as to reduce its relevant TMAO metabolism and restrain the release of proinflammatory cytokines. These results demonstrated the protective effects of GNS on osteoporosis as a gut microbiota modulator and offered novel insights into the regulation of the "gut-bone" axis.


Asunto(s)
Microbioma Gastrointestinal , Nanopartículas del Metal , Nanosferas , Osteoporosis , Femenino , Ratones , Animales , Oro/farmacología , Citocinas , Interleucina-6
4.
Acta Biomater ; 164: 458-473, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37072065

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease causing severe symptoms that are difficult to treat. Nano-drug delivery system is recognized as a promising strategy for management of RA. However, how to thoroughly release payloads from nanoformulations and synergistic therapy of RA needs to be further investigated. To address this issue, a pH and reactive oxygen species (ROS) dual-responsive, methylprednisolone (MPS)-loaded and arginine-glycine-aspartic acid (RGD)-modified nanoparticles (NPs) was fabricated using phytochemical and ROS-responsive moiety co-modified α-cyclodextrin (α-CD) as a carrier. In vitro and in vivo experiments verified that the pH/ROS dual-responsive nanomedicine could be efficiently internalized by activated macrophages and synovial cells, and the released MPS could promote transformation of M1-type macrophages into M2 phenotype, thereby down-regulating pro-inflammatory cytokines. In vivo experiments demonstrated that the pH/ROS dual-responsive nanomedicine was remarkably accumulated in the inflamed joints of mice with collagen-induced arthritis (CIA). The accumulated nanomedicine could obviously relieve joint swelling and cartilage destruction without obvious adverse effects. Importantly, the expression of interleukin-6 and tumor necrosis factor-α in the joints of CIA mice were significantly inhibited by the pH/ROS dual-responsive nanomedicine in comparison with free drug and non-targeted counterparts. In addition, the expression of the NF-κB signaling pathway molecule P65 was also significantly decreased by nanomedicine-treatment. Our results reveal that MPS-loaded pH/ROS dual-responsive NPs can effectively alleviate joint destruction via down-regulation of the NF-κB signaling pathway. STATEMENT OF SIGNIFICANCE: Nanomedicine is recognized as an attractive method for the targeting treatment of rheumatoid arthritis (RA). To thorough release of payloads from nanoformulations and synergistic therapy of RA, herein, a phytochemical and ROS-responsive moiety co-modified α-cyclodextrin was used as a pH/ROS dual-responsive carrier to encapsulate methylprednisolone to manage RA. The fabricated nanomedicine can effectively release its payloads under pH and/or ROS microenvironment, and the released drugs dramatically promote transformation of M1-type macrophages into M2 phenotype to reduce the release of pro-inflammatory cytokines. The prepared nanomedicine also obviously decreased the NF-κB signaling pathway molecule P65 expression in the joints, thereby down-regulating pro-inflammatory cytokines expression to alleviate joint swelling and cartilage destruction. We provided a candidate for the targeting treatment of RA.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Nanopartículas , alfa-Ciclodextrinas , Ratones , Animales , FN-kappa B/metabolismo , Glucocorticoides/farmacología , Especies Reactivas de Oxígeno , alfa-Ciclodextrinas/farmacología , alfa-Ciclodextrinas/uso terapéutico , Transducción de Señal , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/patología , Citocinas/farmacología , Nanopartículas/uso terapéutico , Nanopartículas/química , Metilprednisolona , Concentración de Iones de Hidrógeno
5.
Bioact Mater ; 6(12): 4697-4706, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34095626

RESUMEN

Antiresorptive drugs are widely used for treatment of osteoporosis and cancer bone metastasis, which function mainly through an overall inhibition of osteoclast. However, not all osteoclasts are "bone eaters"; preosteoclasts (pOCs) play anabolic roles in bone formation and angiogenesis through coupling with osteoblasts and secreting platelet derived growth factor-BB (PDGF-BB). In this study, a bone-targeted pH-responsive nanomaterial was designed for selectively eliminating mature osteoclasts (mOCs) without affecting pOCs. Biocompatible cerium nano-system (CNS) was guided to the acidic extracellular microenvironment created by mOCs and gained oxidative enzymatic activity. Oxidative CNS decreased the viability of mOCs through accumulating intracellular reactive oxygen species and enhancing calcium oscillation. Non-acid secreting anabolic pOCs were thus preserved and kept producing PDGF-BB, which lead to mesenchymal stem cell osteogenesis and endothelial progenitor cell angiogenesis via PI3K-Akt activated focal adhesion kinase. In treating osteoporotic ovariectomized mice, CNS showed better protective effects compare with the current first line antiresorptive drug due to the better anabolic effects marked by higher level of bone formation and vascularization. We provided a novel anabolic therapeutic strategy in treating bone disorders with excessive bone resorption.

6.
Acta Biomater ; 126: 183-198, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33711525

RESUMEN

Selective cell retention (SCR) has been widely used as a bone tissue engineering technique for the real-time fabrication of bone grafts. The greater the number of mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs) retained in the scaffold, the better the osteoinductive and angiogenic properties of the scaffold's microenvironment. Improved bioscaffold properties in turn lead to improved bone graft survival, bone regeneration, and angiogenesis. Laminin plays a key role in cell-matrix adhesion, cell proliferation, and differentiation. We designed a collagen-binding domain (CBD) containing the core functional amino acid sequences of laminin α4 (CBD-LN peptide) to supplement the functional surface of a collagen-based decalcified bone matrix (DBM) scaffold. This scaffold promoted MSCs and EPCs early cell adhesion through up-regulating the expression of integrin α5ß1 and integrin αvß3 respectively, thus accelerated the following cell spreading, proliferation, and differentiation. Interestingly, it promoted the retention of MSCs (CD90+/CD105+ cells) and EPCs (CD31+ cells) in the scaffold following the use of clinical SCR technology. Furthermore, the DBM/CBD-LN scaffold induced the formation of type H vessels through the activation of the HIF-1α signaling pathway. The DBM/CBD-LN scaffold displayed rapid bone formation and angiogenesis in vivo, suggesting that it might be used as a new biomaterial in bone tissue engineering. STATEMENT OF SIGNIFICANCE: Selective cell retention technology (SCR) has been utilized in clinical settings to manufacture bioactive bone grafts. Specifically, demineralized bone matrix (DBM) is a widely-used SCR clinical biomaterial but it displays poor adhesion performance and angiogenic activity. In this work, we designed a collagen-binding domain (CBD) containing the core functional amino acid sequences of laminin α4 to supplement the functional surface of a collagen-based DBM scaffold. This bioscaffold promoted SCR-mediated MSCs and EPCs early cell adhesion, thus accelerated the following cell spreading, proliferation, and differentiation. Our results indicate this bioscaffold greatly induced osteogenesis and angiogenesis in vivo. In general, this bioscaffold has a good prospect for SCR application and may provide highly bioactive bone implant in clinical environment.


Asunto(s)
Regeneración Ósea , Adhesión Celular , Laminina , Andamios del Tejido , Diferenciación Celular , Células Progenitoras Endoteliales , Humanos , Células Madre Mesenquimatosas , Osteogénesis , Ingeniería de Tejidos
7.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 34(12): 1594-1601, 2020 Dec 15.
Artículo en Chino | MEDLINE | ID: mdl-33319542

RESUMEN

OBJECTIVE: Based on the cell-extracellular matrix adhesion theory in selective cell retention (SCR) technology, demineralized bone matrix (DBM) modified by simplified polypeptide surface was designed to promote both bone regeneration and angiogenesis. METHODS: Functional peptide of α4 chains of laminin protein (LNα4), cyclic RGDfK (cRGD), and collagen-binding domain (CBD) peptides were selected. CBD-LNα4-cRGD peptide was synthesized in solid phase and modified on DBM to construct DBM/CBD-LNα4-cRGD scaffold (DBM/LN). Firstly, scanning electron microscope and laser scanning confocal microscope were used to examine the characteristics and stability of the modified scaffold. Then, the adhesion, proliferation, and tube formation properties of CBD-LNα4-cRGD peptide on endothelial progenitor cells (EPCs) were detected, respectively. Western blot method was used to verify the molecular mechanism affecting EPCs. Finally, 24 10-week-old male C57 mice were used to establish a 2-mm-length defect of femoral bone model. DBM/LN and DBM scaffolds after SCR treatment were used to repair bone defects in DBM/LN group ( n=12) and DBM group ( n=12), respectively. At 8 weeks after operation, the angiogenesis and bone regeneration ability of DBM/LN scaffolds were evaluated by X-ray film, Micro-CT, angiography, histology, and immunofluorescence staining [CD31, endomucin (Emcn), Ki67]. RESULTS: Material related tests showed that the surface of DBM/LN scaffold was rougher than DBM scaffold, but the pore diameter did not change significantly ( t=0.218, P=0.835). After SCR treatment, DBM/LN scaffold was still stable and effective. Compared with DBM scaffold, DBM/LN scaffold could adhere to more EPCs after the surface modification of CBD-LNα4-cRGD ( P<0.05), and the proliferation rate and tube formation ability increased. Western blot analysis showed that the relative expressions of VEGF, phosphorylated FAK (p-FAK), and phosphorylated ERK1/2 (p-ERK1/2) proteins were higher in DBM/LN than in DBM ( P<0.05). In the femoral bone defect model of mice, it was found that mice implanted with DBM/LN scaffold had stronger angiogenesis and bone regeneration capacity ( P<0.05), and the number of CD31 hiEmcn hi cells increased significantly ( P<0.05). CONCLUSION: DBM/LN scaffold can promote the adhesion of EPCs. Importantly, it can significantly promote the generation of H-type vessels and realize the effective coupling between angiogenesis and bone regeneration in bone defect repair.


Asunto(s)
Matriz Ósea , Osteogénesis , Animales , Regeneración Ósea , Laminina , Masculino , Ratones , Péptidos , Andamios del Tejido
8.
Aging (Albany NY) ; 12(12): 11698-11716, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32527985

RESUMEN

Inflammatory osteolysis is a common osteolytic specificity that occurs during infectious orthopaedic surgery and is characterized by an imbalance in bone homeostasis due to excessive osteoclast bone resorption activity. Epothilone B (Epo B) induced α-tubulin polymerization and enhanced microtubule stability, which also played an essential role in anti-inflammatory effect on the regulation of many diseases. However, its effects on skeletal system have rarely been investigated. Our study demonstrated that Epo B inhibited osteoclastogenesis in vitro and prevented inflammatory osteolysis in vivo. Further analysis showed that Epo B also markedly induced mature osteoclasts apoptosis during osteoclastogenesis. Mechanistically, Epo B directly suppressed osteoclastogenesis by the inhibitory regulation of the phosphorylation and activation of PI3K/Akt/STAT3 signaling directly, and the suppressive regulation of the CD9/gp130/STAT3 signaling pathway indirectly. The negative regulatory effect on STAT3 signaling further restrained the translocation of NF-κB p65 and NFATc1 from the cytosol to the nuclei during RANKL stimulation. Additionally, the expression of osteoclast specific genes was also significantly attenuated during osteoclast fusion and differentiation. Taken together, these findings illustrated that Epo B protected against LPS-induced bone destruction through inhibiting osteoclastogenesis via regulating the STAT3 dependent signaling pathway.


Asunto(s)
Epotilonas/farmacología , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteólisis/prevención & control , Factor de Transcripción STAT3/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Modelos Animales de Enfermedad , Epotilonas/uso terapéutico , Femenino , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Fémur/inmunología , Fémur/patología , Humanos , Lipopolisacáridos/inmunología , Ratones , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Osteoclastos/citología , Osteoclastos/fisiología , Osteogénesis/inmunología , Osteólisis/diagnóstico , Osteólisis/inmunología , Osteólisis/patología , Cultivo Primario de Células , Ligando RANK/metabolismo , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Factor de Transcripción ReIA/metabolismo , Microtomografía por Rayos X
9.
Cell Death Dis ; 11(6): 414, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32488049

RESUMEN

Bone infection contributing to inflammatory osteolysis is common in orthopedic surgery. The dynamic balance between bone formation and bone resorption is destroyed due to excessive osteoclast fusion and differentiation, which results in severe bone matrix loss. Many therapeutic approaches that restrain osteoclast formation and function act as efficient ways to prevent inflammatory bone erosion. We have demonstrated for the first time that dendritic cells-derived interferon-λ1 (IFN-λ1) inhibited inflammatory bone destruction in vivo and explored its underlying mechanisms on osteoclast formation in vitro. We found that IFN-λ1 was highly expressed in infectious bone tissue compared with that of non-infectious bone tissue. Additionally, dendritic cells marker genes such as CD80, CD86, and CD1a were higher expressed in infectious bone tissue than that of non-infectious bone tissue. Dendritic cells that were pretreated with LPS showed high expression of IFN-λ1. Moreover, conditioned medium of LPS-pretreated dendritic cells significantly inhibited osteoclast differentiation, as determined by TRAP staining assay. This suppressive effect was reversed by adding an IFN-λ1 monoclonal antibody. It was also investigated whether exogenous IFN-λ1 restrained osteoclastogenesis, bone resorption, F-actin ring formation, osteoclast-specific gene expression, release of pro-inflammatory cytokines, and translocation of p65 and NFATc1 by preventing the NF-κB signaling pathway and NLRP3 inflammasome formation, as well as by inducing the JAK-STAT signaling pathways in vitro. In vivo study indicated that IFN-λ1 prevents lipopolysaccharide (LPS)-induced inflammatory bone destruction by inhibiting excessive osteoclast fusion and bone resorption activity. In conclusion, our findings confirmed that dendritic cells-derived IFN-λ1 could attenuate osteoclast formation and bone resorptive activity in vitro and in vivo. These novel findings pave the way for the use of exogenous IFN-λ1 as a potential therapeutic treatment for excessive osteoclast-related diseases, such as inflammatory osteolysis, by regulating osteoclastogenesis to maintain the dynamic balance between bone formation and bone resorption.


Asunto(s)
Huesos/patología , Células Dendríticas/metabolismo , Inflamación/patología , Interferones/metabolismo , Interleucinas/metabolismo , Osteoclastos/patología , Osteogénesis , Animales , Resorción Ósea/complicaciones , Resorción Ósea/patología , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Fusión Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamasomas/metabolismo , Inflamación/complicaciones , Mediadores de Inflamación/metabolismo , Interferones/farmacología , Interleucinas/farmacología , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Osteólisis/patología , Osteomielitis/complicaciones , Osteomielitis/patología , Ligando RANK/metabolismo , Células RAW 264.7 , Transducción de Señal
10.
Adv Healthc Mater ; 9(13): e2000353, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32424991

RESUMEN

The immunologic response toward chronic inflammation or bone regeneration via the accumulation of M1 or M2 macrophages after injury could determine the fate of biomaterial. Human umbilical cord mesenchymal stem cells (hUCMSCs) have a pivotal immunomodulatory property on directing macrophage behaviors. Herein, for the first time, 3D-printed poly(lactide-co-glycolide) (PLGA) scaffolds modified with hUCMSC-derived extracellular matrix (PLGA-ECM) are prepared by a facile tissue engineering technique with physical decellularization and 2.44 ± 0.29 mg cm-3 proteins immobilized on the PLGA-ECM contain multiple soluble cytokines with a sustainable release profile. The PLGA-ECM not only attenuates the foreign body response, but also improves bone regeneration by increasing the accumulation of M2 macrophages in an improved heterotopic transplantation model of SCID mice. Furthermore, the PLGA-ECM scaffolds with the knockdown of transforming growth factor-ß-induced protein (TGFßI/ßig-H3) demonstrate that M2 macrophage accumulation improved by the PLGA-ECM could be attributed to increasing the migration of M2 macrophages and the repolarization of M1 macrophages to M2 phenotype, which are mediated by multiple integrin signaling pathways involving in integrin ß7, integrin α9, and integrin ß1 in a TGFßI-dependent manner. This study presents an effective surface modification strategy of polymeric scaffolds to initiate tissue regeneration and combat inflammatory response by increasing M2 macrophage accumulation.


Asunto(s)
Matriz Extracelular , Andamios del Tejido , Animales , Regeneración Ósea , Inflamación , Macrófagos , Ratones , Ratones SCID , Factor de Crecimiento Transformador beta
11.
Mol Biol Rep ; 47(6): 4587-4629, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32333246

RESUMEN

Given that the PI3K/AKT pathway has manifested its compelling influence on multiple cellular process, we further review the roles of hyperactivation of PI3K/AKT pathway in various human cancers. We state the abnormalities of PI3K/AKT pathway in different cancers, which are closely related with tumorigenesis, proliferation, growth, apoptosis, invasion, metastasis, epithelial-mesenchymal transition, stem-like phenotype, immune microenvironment and drug resistance of cancer cells. In addition, we investigated the current clinical trials of inhibitors against PI3K/AKT pathway in cancers and found that the clinical efficacy of these inhibitors as monotherapy has so far been limited despite of the promising preclinical activity, which means combinations of targeted therapy may achieve better efficacies in cancers. In short, we hope to feature PI3K/AKT pathway in cancers to the clinic and bring the new promising to patients for targeted therapies.


Asunto(s)
Neoplasias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Apoptosis/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Transformación Celular Neoplásica/genética , Transición Epitelial-Mesenquimal/fisiología , Humanos , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología
12.
ACS Appl Mater Interfaces ; 12(3): 3474-3493, 2020 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-31874023

RESUMEN

Vasculogenesis (de novo formation of vessels) induced by endothelial progenitor cells (EPCs) is requisite for vascularized bone regeneration. However, there exist few available options for promoting vasculogenesis within artificial bone grafts except for exogenous EPC transplantation, which suffers from the source of EPC, safety, cost, and time concerns in clinical applications. This study aimed at endogenous EPC recruitment for vascularized bone regeneration by using a bioinspired EPC-induced graft. The EPC-induced graft was created by immobilizing two bioactive peptides, WKYMVm and YIGSR, on the surface of poly(ε-caprolactone) (PCL)/poliglecaprone (PGC) nanofibrous scaffolds via a polyglycolic acid (PGA)-binding peptide sequence. Remarkable immobilization efficacy of WKYMVm and YIGSR peptides and their sustained release (over 14 days) from scaffolds were observed. In vivo and in vitro studies showed robust recruitment of EPCs, which subsequently contributed to early vasculogenesis and ultimate bone regeneration. The dual-peptide-functionalized nanofibrous scaffolds proposed in this study provide a promising therapeutic strategy for vasculogenesis in bone defect repair.


Asunto(s)
Enfermedades Óseas/terapia , Células Progenitoras Endoteliales/citología , Nanofibras/química , Péptidos/química , Cráneo/anomalías , Cráneo/irrigación sanguínea , Animales , Enfermedades Óseas/fisiopatología , Regeneración Ósea , Adhesión Celular , Proliferación Celular , Células Progenitoras Endoteliales/trasplante , Humanos , Masculino , Neovascularización Patológica , Péptidos/administración & dosificación , Ratas , Ratas Sprague-Dawley , Cráneo/cirugía , Ingeniería de Tejidos , Andamios del Tejido/química
13.
Cell Death Dis ; 10(5): 353, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31040267

RESUMEN

A variety of osteolytic factors have been identified from breast cancer cells leading to osteolysis, but less is known about which factor plays an essential role in the initiation process prior to the overt vicious osteolytic cycle. Here, we present in vitro and in vivo evidences to clarify the role of interleukin-11 (IL-11) as an essential contributor to breast cancer bone metastasis mediated osteolysis. Animal studies showed that bone specific metastatic BoM-1833 cells induce earlier onset of osteolysis and faster tumor growth compared with MCF7 and parental MDA-MB-231 cells in BALB/c-nu/nu nude mice. IL-11 was further screened and identified as the indispensable factor secreted by BoM-1833 cells inducing osteoclastogenesis independently of receptor activator of nuclear factor κB ligand (RANKL). Mechanistic investigation revealed that the JAK1/STAT3 signaling pathway as a downstream effector of IL-11, STAT3 activation further induces the expression of c-Myc, a necessary factor required for osteoclastogenesis. By inhibiting STAT3 phosphorylation, AG-490 was shown effective in reducing osteolysis and tumor growth in the metastatic niche. Overall, our results revealed the essential role and the underlying molecular mechanism of IL-11 in breast cancer bone metastasis mediated osteolysis. STAT3 targeting through AG-490 is a potential therapeutic strategy for mitigating osteolysis and tumor growth of bone metastatic breast cancer.


Asunto(s)
Neoplasias Óseas/patología , Neoplasias de la Mama/patología , Interleucina-11/farmacología , Osteogénesis/efectos de los fármacos , Osteólisis/prevención & control , Ligando RANK/farmacología , Animales , Neoplasias Óseas/mortalidad , Neoplasias Óseas/secundario , Huesos/diagnóstico por imagen , Huesos/patología , Línea Celular Tumoral , Femenino , Humanos , Interleucina-11/metabolismo , Interleucina-11/uso terapéutico , Janus Quinasa 1/metabolismo , Estimación de Kaplan-Meier , Factor Estimulante de Colonias de Macrófagos/farmacología , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Osteólisis/patología , Ligando RANK/uso terapéutico , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Tirfostinos/farmacología , Tirfostinos/uso terapéutico
14.
J Bone Miner Res ; 33(5): 899-908, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29281118

RESUMEN

In this study, for the first time we discovered that the M1/M2 macrophage phenotype ratio is increased in bone marrow of ovariectomized (OVX) osteoporotic C57BL/6 mice. Considering estrogen is the main variable, we assumed that estrogen participated in this alteration. To determine whether and how estrogen contributes to the change of the M1/M2 ratio, we first isolated bone marrow macrophages (BMMs) from mice femur and stimulated the cells with lipopolysaccharide (LPS)/interferon γ (IFN-γ) for M1 polarization and interleukin 4 (IL-4)/IL-13 for M2 polarization. M1 and M2 macrophages were then exposed to RANKL stimulation, we found that M2 macrophage but not M1 macrophage differentiated into functional osteoclast leading to increased M1/M2 ratio. Intriguingly, 17ß-estradiol (E2) pretreatment prevented osteoclastogenesis from M2 macrophages. By constructing shRNA lentivirus interfering the expression of different estrogen receptors in M2 macrophages, we found that estrogen protects M2 macrophage from receptor activator of nuclear factor κB ligand (RANKL) stimulation selectively through estrogen receptor α (ERα) and the downstream blockage of NF-κB p65 nuclear translocation. Animal studies showed that ERα selective agonist 4,4',4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl) trisphenol (PPT) was able to replicate the therapeutic effects of E2 in treating osteoporotic OVX mice. Together, our findings reveal that estrogen deficiency-mediated M2 macrophage osteoclastogenesis leads to increased M1/M2 ratio in OVX mice. Reducing the M1/M2 ratio is a potential therapeutic target in treating postmenopausal osteoporosis. © 2017 American Society for Bone and Mineral Research.


Asunto(s)
Células de la Médula Ósea/metabolismo , Estrógenos/deficiencia , Fémur/metabolismo , Osteoclastos/metabolismo , Osteoporosis/metabolismo , Animales , Células de la Médula Ósea/patología , Receptor alfa de Estrógeno/agonistas , Receptor alfa de Estrógeno/metabolismo , Femenino , Fémur/patología , Ratones , Osteoclastos/patología , Osteoporosis/patología , Ovariectomía , Fenoles/farmacología , Pirazoles/farmacología , Ligando RANK/metabolismo , Factor de Transcripción ReIA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA