Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Cell Physiol ; 237(1): 815-823, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34378805

RESUMEN

Merlin is known as a tumor suppressor, while its role in osteomyelitis remains unclear. This study aimed to investigate the role of Merlin in Staphylococcus aureus-induced osteomyelitis and its underlying mechanisms. S. aureus-induced osteomyelitis mouse model was established in Merlinfl/fl Lyz2cre/+ and Merlinfl/fl Lyz2+/+ mice. Bone marrow-derived macrophages (BMDMs) were isolated and stimulated by lipopolysaccharide (LPS). Bioassays, including quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blot analysis, and enzyme-linked immunosorbent assays, were conducted to determine the levels of target genes or proteins. Immunoprecipitation was applied to determine the interactions between proteins. DCAF1fl/fl mice were further crossed with Lyz2-Cre mice to establish myeloid cell conditional knockout mice (DCAF1fl/fl Lyz2cre/+ ). It was found that the level of Merlin was elevated in patients with osteomyelitis and S. aureus-infected BMDMs. Merlin deficiency in macrophages suppressed the production of inflammatory cytokines and ameliorated the symptoms of osteomyelitis induced by S. aureus. Merlin deficiency in macrophages also suppressed the production of proinflammatory cytokines in BMDMs induced by LPS. The inhibitory effects of Merlin deficiency on the inflammatory response were associated with DDB1-Cul4-associated factor 1 (DCAF1). In summary, Merlin deficiency ameliorates S. aureus-induced osteomyelitis through the regulation of DCAF1.


Asunto(s)
Osteomielitis , Infecciones Estafilocócicas , Animales , Citocinas , Humanos , Lipopolisacáridos/farmacología , Ratones , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Staphylococcus aureus/metabolismo
2.
Immunopharmacol Immunotoxicol ; 44(4): 556-564, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35404181

RESUMEN

INTRODUCTION: 17ß-Estradiol (E2) is an immune-regulatory agent with anti-inflammatory effects. However, it is still unknown whether E2 exerts pharmacological properties against Achilles tendinitis (AT). This study aims to investigate the effects of E2 on AT and its underlying mechanisms. MATERIALS AND METHODS: The established model of Achilles tendinitis was intraperitoneally injected with E2 (10, 20, or 30 µg/kg/d). After 8 weeks, biomechanical properties of the Achilles tendon were determined. Hydroxyproline content and tendon degeneration-related biomarkers were determined. The levels of inflammatory cytokines and apoptotic-related biomarkers in tendon tissues were determined. Furthermore, western blotting was determined to detect the expressions of ER-α and the PI3K/Akt pathway in tendon tissues. RESULTS: E2 relieved AT-related symptoms in a dose-dependent manner. E2 ameliorated tendon degeneration by regulating tendon degeneration-related biomarkers (e.g. collagen types I and III, Decorin (DCN), and tenascin-C). Besides, treatment with E2 suppressed inflammatory cytokines and increased anti-inflammatory cytokines. Treatment with E2 also regulated cell apoptosis in tendon tissues. The underlying mechanism study revealed that treatment with E2 activated ER-α and upregulated the PI3K/Akt pathway. CONCLUSION: The regulatory effects of E2 on inflammation and tendon degeneration in a rat model of AT were associated with the ER-α and the PI3K/Akt signaling pathways.


Asunto(s)
Antiinflamatorios , Estradiol , Tendinopatía , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Citocinas/metabolismo , Estradiol/farmacología , Estradiol/uso terapéutico , Inflamación/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Tendinopatía/tratamiento farmacológico , Tendones/metabolismo
3.
J Cell Physiol ; 236(1): 284-293, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32592173

RESUMEN

Bone marrow mesenchymal stem cells (BMSCs) can be induced to process osteogenic differentiation with appropriate mechanical and/or chemical stimuli. The present study described the successful culture of murine BMSCs under mechanical strain. BMSCs were subjected to 0%, 3%, 8%, 13%, and 18% cyclic tensile strain at 0.5 Hz for 8 hr/day for 3 days. The expression of osteogenic markers and mechanosensitive ion channels was evaluated with real-time reverse transcription-polymerase chain reaction (RT-PCR) and western blot. The expression of alkaline phosphatase (ALP) and matrix mineralization were evaluated with histochemical staining. To investigate the effects of mechanosensitive ion channel expression on cyclic tensile strain-induced osteogenic differentiation, the expression of osteogenic markers was evaluated with real-time RT-PCR in the cells without mechanosensitive ion channel expression. This study revealed a significant augment in osteogenic marker in BMSC strained at 8% compared to other treatments; therefore, an 8% strain was used for further investigations. The ALP expression and matrix mineralization were enhanced in osteogenic induced BMSCs subjected to 8% strain after 7 and 14 days, respectively. Under the same conditions, the osteogenic marker and mechanosensitive ion channel expression were significantly promoted. However, the loss function of mechanosensitive ion channels resulted in the inhibition of osteogenic marker expression. This study demonstrated that strain alone can successfully induce osteogenic differentiation in BMSCs and the expression of mechanosensitive ion channels was involved in the process. The current findings suggest that mechanical stretch could function as efficient stimuli to induce the osteogenic differentiation of BMSCs via the activation of mechanosensitive ion channels.


Asunto(s)
Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Diferenciación Celular/fisiología , Canales Iónicos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Osteogénesis/fisiología , Fosfatasa Alcalina/metabolismo , Animales , Biomarcadores/metabolismo , Células Cultivadas , Ratones
4.
Connect Tissue Res ; 62(5): 485-494, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-32500755

RESUMEN

AIMS: Inflammatory responses to wear debris cause osteolysis that leads to aseptic loosening and hip arthroplasty failure. Wear debris stimulate macrophages and fibroblasts to secret proinflammatory cytokines, including TNF-α and IL-6, which have been specifically implicated in periprosthetic osteolysis and osteoclast differentiation. Naringin has anti-inflammatory effect in macrophages. Moreover, naringin inhibited osteoclastogenesis and wear particles-induced osteolysis. In this study, we examined the potential inhibitory effects of naringin on titanium (Ti) particle-induced proinflammatory cytokines secretion in fibroblasts and the possible underlying molecular mechanisms. MATERIALS AND METHODS: Fibroblasts were isolated from periprosthetic membrane at the time of revision surgery performed due to aseptic loosening after hip arthroplasty and were cultured in the presence or absence of Ti particles, naringin and mitogen-activated protein kinase (MAPK) inhibitors, PD98059 (a selective inhibitor of ERK), SP600125 (a selective inhibitor of JNK), and SB203580 (a selective inhibitor of p38). TNF-α and IL-6 assays were performed using enzyme-linked immunosorbent assay kits. The phosphorylation levels of p38 and nuclear factor kappa B p65 (NF-κB p65) were examined by western blot. RESULTS: Naringin or SB203580 pretreatment significantly suppressed the secretion of TNF-α and IL-6 induced by titanium particles in fibroblasts, while inhibition of ERK or JNK pathways showed no effect on production of TNF-α and IL-6. Moreover, naringin inhibited Ti particle-induced phosphorylation of p38 and p65. CONCLUSIONS: These results indicated that naringin could inhibit Ti particle-induced inflammation in fibroblasts by inhibiting p38 MAPK/NF-κB p65 activity and might be a potential drug for the treatment of inflammatory periprosthetic osteolysis after arthroplasty.


Asunto(s)
Fibroblastos , Fibroblastos/metabolismo , Flavanonas , Humanos , Interleucina-6 , FN-kappa B/metabolismo , Osteoclastos/metabolismo , Osteólisis/inducido químicamente , Osteólisis/tratamiento farmacológico , Titanio/efectos adversos , Factor de Necrosis Tumoral alfa , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/genética
5.
J Cell Mol Med ; 24(1): 686-694, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31691506

RESUMEN

Type 2 diabetes mellitus (T2DM) is the most common diabetes and has numerous complications. Recent studies demonstrated that T2DM compromises bone fracture healing in which miR-222 might be involved. Furthermore, tissue inhibitor of metalloproteinase 3 (TIMP-3) that is the target of miR-222 accelerates fracture healing. Therefore, we assume that miR-222 could inhibit TIMP-3 expression. Eight-week-old rats were operated femoral fracture or sham, following the injection of streptozotocin (STZ) to induce diabetes one week later in fractured rats, and then, new generated tissues were collected for measuring the expression of miR-222 and TIMP-3. Rat mesenchymal stem cells (MSCs) were isolated and treated with miR-222 mimic or inhibitor to analyse osteogenic differentiation. MiR-222 was increased in fractured rats and further induced in diabetic rats. In contrast, TIMP-3 was reduced in fractured and further down-regulated in diabetic rats. Luciferase report assay indicated miR-222 directly binds and mediated TIMP-3. Furthermore, osteogenic differentiation was suppressed by miR-222 mimic and promoted by miR-222 inhibitor. miR-222 is a key regulator that is promoted in STZ-induced diabetic rats, and it binds to TIMP3 to reduce TIMP-3 expression and suppressed MSCs' differentiation.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Fracturas Óseas/terapia , Células Madre Mesenquimatosas/citología , MicroARNs/genética , Osteogénesis , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Animales , Femenino , Curación de Fractura , Fracturas Óseas/etiología , Fracturas Óseas/metabolismo , Fracturas Óseas/patología , Regulación de la Expresión Génica , Ratas , Ratas Sprague-Dawley , Inhibidor Tisular de Metaloproteinasa-3/genética
6.
Int Immunol ; 31(4): 263-273, 2019 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-30779845

RESUMEN

Inflammation plays an important role in osteonecrosis. Obesity, a risk factor for osteonecrosis, leads to a chronic inflammatory status. We hypothesized that inflammation mediated the effects of obesity on osteonecrosis and tested our hypothesis in a mouse model of osteonecrosis. We fed mice with a high-fat diet (HFD) for 12 weeks before osteonecrosis induction by methylprednisolone and examined bone structure and IL-6 expression. Then we investigated the effects of IL-6 deletion in mice with osteonecrosis on the HFD. Next, we isolated bone marrow cells and determined the cell types responsible for HFD-induced IL-6 secretion. Finally, we investigated the roles of macrophages and macrophage-driven IL-6 in HFD-mediated effects on osteonecrosis and osteogenesis of bone marrow stromal cells (BMSCs). The HFD lead to exacerbated destruction of the femoral head in mice with osteonecrosis and increased IL-6 expression in macrophages. Il-6 knockout or macrophage depletion suppressed the effects of the HFD on bone damage. When co-cultured with macrophages isolated from HFD-fed mice with osteonecrosis, BMSCs showed reduced viability and suppressed osteogenic differentiation. Our results suggest that macrophage-driven IL-6 bridges obesity and osteonecrosis and inhibition of IL-6 or depletion of macrophage may represent a therapeutic strategy for obesity-associated osteonecrosis.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Inflamación/metabolismo , Interleucina-6/metabolismo , Macrófagos/inmunología , Obesidad/metabolismo , Osteonecrosis/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Interleucina-6/genética , Masculino , Metilprednisolona , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/inmunología , Osteogénesis , Osteonecrosis/inmunología , Transducción de Señal
7.
Immun Ageing ; 17: 19, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32536957

RESUMEN

BACKGROUND: Whether IL-15 is involved in the development of steroid-induced osteonecrosis of the femoral head (ONFH) is investigated. METHODS: C57BL/6 J and l15-/-mice were injected with methylprednisolone to induce wide type osteonecrosis (WT ON) and IL-15 deficiency osteonecrosis (IL-15-/- ON). Hematoxylin-Eosin (H&E) staining and micro-computed tomography (micro-CT) scanning was used to detect the microstructure. The differentiation and formation of osteoclasts were determined with colony-forming unit-granulocyte macrophages (CFU-GM), colony-forming unit-macrophage/mononuclear (CFU-M) per tibia, and tartrate-resistant acid phosphatase (TRACP or TRAP) positive cells. Serum interleukin (IL)-15, osteocalcin, bone alkaline phosphatase (BAP), bone Gla protein (BGP), and TRACP were assayed with enzyme-linked immunosorbent assay (ELISA). The receptor activator of nuclear factor-κB (RANK), RANK ligand (RANKL), and osteoprotegerin (OPG) in the femoral heads were detected by Western blot. CD34 staining was performed to detect microvascular density. RESULTS: IL-15 secretion was increased in the femoral heads and the serum of steroid-induced ONFH mice. IL-15 deficiency may lead to up-regulated vessel remodeling, improved microstructure, and up-regulated serum osteocalcin, BAP, and BGP secretion. Both the expression of RANKL/RANK/OPG and osteoclast differentiation and formation can be down-regulated by IL-15 deficiency. CONCLUSION: IL-15 deficiency alleviates steroid-induced ONFH by impact osteoclasts via RANKL-RANK-OPG system.

8.
Nanomedicine ; 24: 102116, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31672602

RESUMEN

Few studies reported the application of miRNA in bone regeneration. In this study, the expression of miR133a and miR133b in murine BMSCs was inhibited via antagomiR-133a/b and the osteogenic differentiation in murine BMSCs was evaluated. The RT-PCR, flow cytometry, cell counting kit-8, and annexin V-FITC/PI double staining assays were performed. Double knockdown miR133a and miR133b can promote BMSC osteogenic differentiation. At optimum N/P ration (15:1), the loading efficiency can reach over 90%. CTH-antagomiR-133a/b showed no cytotoxicity to BMSCs and diminished miR133a and miR133b expression in BMSCs. Furthermore, chitosan-based sustained delivery system can facilitate continuous dosing of antagomiR-133a/b, which enhanced calcium deposition and osteogenic specific gene expression in vitro. The new bone formation was enhanced after the sustained delivery system containing CTH-antagomiR-133a/b nanoparticles was used in mouse calvarial bone defect model. Our results demonstrate that CTH nanoparticles could facilitate continuous dosing of antagomiR133a/b, which can promote osteogenic differentiation.


Asunto(s)
Antagomirs , Regeneración Ósea/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/antagonistas & inhibidores , Nanopartículas/química , Osteogénesis/efectos de los fármacos , Animales , Antagomirs/química , Antagomirs/farmacocinética , Antagomirs/farmacología , Células de la Médula Ósea , Regeneración Ósea/genética , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Técnicas de Silenciamiento del Gen , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Osteogénesis/genética
9.
Am J Otolaryngol ; 41(4): 102547, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32474328

RESUMEN

BACKGROUND: N6-Methyladenosine (m6A) is a ubiquitous RNA modification with vital roles in various cancers, but little is known about its role in papillary thyroid carcinoma (PTC), a common endocrine malignancy. METHODS: In this study, an m6A RNA methylation regulator-based biomarker signature was developed for the effective prediction of prognosis in patients with PTC. The gene expression profiles of m6A RNA methylation regulators and the corresponding clinical information was downloaded from The Cancer Genome Atlas (TCGA). Differentially expressed m6A RNA methylation regulators between tumor and normal control samples, and correlation expression levels, clinical parameters, and outcomes were evaluated. And a prognostic signature was built using a PTC cohort from TCGA. RESULTS: The expression level of HNRNPC was remarkably upregulated in tumor samples, while WTAP, RBM15, YTHDC2, YTHDC1, FTO, METTL14, METTL3, ALKBH5, KIAA1429, YTHDF1, and ZC3H13 were significantly downregulated in the cancer specimens compared with those in control samples. A three-gene prognostic signature comprising RBM15, KIAA1429, and FTO could predict overall survival in patients with PTC. In addition, the prognostic signature-based risk score was identified as an independent prognostic indicator for PTC. CONCLUSIONS: We established a robust m6A RNA methylation regulator-based molecular signature for predicting prognosis in patients with PTC with high accuracy; this signature might provide important guidance for therapeutic strategies.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Expresión Génica , Metiltransferasas/genética , Metiltransferasas/metabolismo , Cáncer Papilar Tiroideo/genética , Neoplasias de la Tiroides/genética , Adulto , Anciano , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Femenino , Ribonucleoproteína Heterogénea-Nuclear Grupo C/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo C/metabolismo , Humanos , Masculino , Metilación , Persona de Mediana Edad , Pronóstico , ARN Helicasas/genética , ARN Helicasas/metabolismo , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Tasa de Supervivencia , Cáncer Papilar Tiroideo/mortalidad , Neoplasias de la Tiroides/mortalidad , Regulación hacia Arriba/genética
10.
Am J Otolaryngol ; 41(2): 102370, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31889554

RESUMEN

BACKGROUND: Main surgical treatments for secondary hyperparathyroidism (SHPT) include subtotal parathyroidectomy (sPTX), total parathyroidectomy with autotransplantation (tPTX+AT), and total parathyroidectomy (tPTX); however, determining the best treatment is debatable. We conducted a network meta-analysis (NMA) comparing three treatments in terms of postoperative hypocalcemia (or hypoparathyroidism), postoperative recurrence, and reoperation. METHODS: We searched PubMed, Medline, the Cochrane Library, and Embase for relevant research from inception to July 30, 2019. We performed our Bayesian NMA using R 3.51 software to assess odds ratios (OR) and 95% confidence intervals (CI). Network and forest plots displayed study outputs. Potential publication bias was assessed with funnel plots using software Stata/MP 13.0. RESULTS: Twenty-six articles comprising 5063 patients were included in our NMA, which showed that postoperative hypocalcemia (or hypoparathyroidism) occurred more frequently in tPTX than in sPTX (OR = 3.50, 95% CI 1.10-11.0) or tPTX+AT patients (OR = 1.80, 95% CI 0.66-5.20). Regarding postoperative hypocalcemia (or hypoparathyroidism), there was no significant difference between sPTX and tPTX+AT (OR = 0.53, 95% CI 0.24-1.10). As for recurrence rates, statistically significant differences were observed between sPTX and tPTX (OR = 25.0, 95% CI 5.1-260), tPTX+AT and tPTX (OR = 20.0, 95% CI 4.2-200), and sPTX and tPTX+AT (OR = 1.30, 95% CI 0.65-2.50). Regarding reoperation rates, sPTX experienced higher incidence compared with tPTX+AT (OR = 1.20, 95% CI 0.53-2.70) or tPTX patients (OR = 2.70, 95% CI 1.20-14.00). CONCLUSIONS: TPTX+AT is recommended as the most efficient and safe surgical SHPT treatment with minimal adverse effects. Large-scale randomized controlled trials are recommended to confirm the NMA results.


Asunto(s)
Hiperparatiroidismo Secundario/cirugía , Metaanálisis en Red , Paratiroidectomía/métodos , Humanos , Hipocalcemia , Hipoparatiroidismo , Complicaciones Posoperatorias , Reoperación , Trasplante Autólogo
11.
Eur Arch Otorhinolaryngol ; 277(3): 881-886, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31792653

RESUMEN

PURPOSE: To investigate the risk factors of lymph node posterior to the right recurrent laryngeal nerve (LN-prRLN) metastasis in papillary thyroid carcinoma. METHODS: Clinicopathologic feature data of 427 patients with right or double lobes who underwent surgery between January 2014 to August 2019 in the Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, were retrospectively analyzed. The risk factors of LN-prRLN metastasis were analyzed by the Chi-squared test and multivariate logistic regression. RESULTS: LN-prRLN metastasis was detected in 96 patients. Univariate analysis showed that age, right-side tumor diameter, capsular invasion, comorbid adenoma, and VIa compartment LN metastasis were significantly associated with LN-prRLN metastasis (all P < 0.05). Multivariate logistic regression analysis showed that right-side tumor diameter, capsular invasion, and VIa compartment LN metastasis were independent risk factors of LN-prRLN metastasis (all P ≤ 0.001). The receiver operating characteristic curve showed that the cutoff value of the right tumor diameter for predicting LN-prRLN metastasis was 1.25 cm (sensitivity = 0.5, specificity = 0.819, area under the curve = 0.720, P < 0.001). CONCLUSION: The incidence of LN-prRLN metastasis cannot be ignored, and our findings indicate that prophylactic LN-prRLN dissection should be performed in patients with right-side tumor diameter ≥ 1.25 cm, capsular invasion, and VIa compartment LN metastasis.


Asunto(s)
Carcinoma Papilar , Carcinoma , Neoplasias de la Tiroides , Carcinoma/cirugía , Carcinoma Papilar/cirugía , China/epidemiología , Humanos , Ganglios Linfáticos/cirugía , Metástasis Linfática , Nervio Laríngeo Recurrente , Estudios Retrospectivos , Factores de Riesgo , Cáncer Papilar Tiroideo/cirugía , Neoplasias de la Tiroides/cirugía , Tiroidectomía
12.
Pharmacology ; 102(3-4): 117-125, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29961054

RESUMEN

BACKGROUND/AIMS: Osteoarthritis is the most common degenerative joint disease and causes major pain and disability in adults. It has been reported that mitochondrial dysfunction in chondrocytes was associated with osteoarthritis. Puerarin has multiple effects including restoring mitochondrial function. In this study, the potential effects of puerarin on osteoarthritis and osteoarthritis associated mitochondrial dysfunctions were evaluated. METHODS: Osteoarthritis rats were treated with puerarin and the severity of osteoarthritis and cartilage damages was evaluated. The mitochondrial biogenesis and functions were analyzed by measuring related proteins expression, mitochondrial DNA content, ATP production, and oxygen consumption. The dependence of AMP-activated protein kinase (AMPK) pathway on puerarin-regulated mitochondrial function was analyzed by applying AMPK inhibitor Compound C. RESULTS: Puerarin treatment alleviated mechanical hyperalgesia and cartilage damage in osteoarthritis rats. Puerarin increased mitochondrial biogenesis and attenuated mitochondrial dysfunctions in osteoarthritis rats. AMPK inhibitor Compound C abolished puerarin's effects. CONCLUSION: Puerarin attenuates osteoarthritis by upregulating the AMPK/proliferator-activated receptor-γ coactivator signaling pathway in osteoarthritis rats.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Isoflavonas/farmacología , Osteoartritis/tratamiento farmacológico , Osteoartritis/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Animales , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Células Cultivadas , Interacciones Farmacológicas , Hiperalgesia/tratamiento farmacológico , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Vasodilatadores/farmacología
13.
Small Methods ; : e2401127, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39300860

RESUMEN

The hydrogel platform with intelligent drug delivery system possesses great potential in the treatment of diabetic wounds. Nevertheless, the intelligent elimination of reactive oxygen species (ROS) remains a formidable challenge in facilitating diabetic wound healing. Herein, a hydrogel platform with triple-triggered on-demand release is constructed to intelligently scavenge ROS and modulate the wound microenvironment to accelerate diabetic wound healing through the release of antioxidative factors. Specifically, the gelatin (Gel) is modified with phenylboronic acid (PBA) to obtain a glucose-sensitive Gel derivative (Gel-BA), which is mixed with oxidized dextran (ODex) and the strong antioxidant myricetin (MY) to swiftly generate a hydrogel platform (OGM). Significantly, the smart release of MY from the hybrid hydrogel under inflammatory conditions intelligently eliminates ROS, effectively alleviating oxidative stress and promoting angiogenic reprogramming of the wound immune microenvironments by activating the Nrf2 pathway. In summary, in vitro and in vivo studies reveal that the OGM hydrogel platform significantly promotes cell proliferation, migration, and tube formation and greatly accelerates diabetic wound healing, offering a local-specific triple-response drug release strategy for the treatment of diabetic wound management.

14.
Front Cell Infect Microbiol ; 14: 1325390, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38379772

RESUMEN

The impact of various fatty acid types on adaptive immunity remains uncertain, and their roles remain unelucidated. Stearoyl-CoA desaturase (Scd) is a Δ-9 desaturase, which is a key rate-limiting enzyme for the conversion of saturated fatty acids (SFA) to monounsaturated fatty acids (MUFA) in the fatty acid de novo synthesis. Scd-1 converts stearic acid (SA) and palmitic acid (PA) to oleic acid (OA) and palmitoleic acid (PO), respectively. In this study, through a series of experiments, we showed that Scd-1 and its resulting compound, OA, have a substantial impact on the transformation of CD8+ naïve T cells into effector T cells. Inactivation of Scd-1 triggers the specialization of CD8+ T cells into the Teff subset, enhancing the effector function and mitochondrial metabolism of Teff cells, and OA can partially counteract this. A deeper understanding of lipid metabolism in immune cells and its impact on cell function can lead to new therapeutic approaches for controlling the immune response and improving prognosis.


Asunto(s)
Ácidos Grasos , Estearoil-CoA Desaturasa , Ácidos Grasos/metabolismo , Ácido Oléico/metabolismo
15.
IEEE Trans Neural Netw Learn Syst ; 34(9): 5816-5827, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34890340

RESUMEN

The topic of identification for sparse vector in a distributed way has triggered great interest in the area of adaptive filtering. Grouping components in the sparse vector has been validated to be an efficient way for enhancing identification performance for sparse parameter. The technique of pairwise fused lasso, which can promote similarity between each possible pair of nonnegligible components in the sparse vector, does not require that the nonnegligible components have to be distributed in one or multiple clusters. In other words, the nonnegligible components may be randomly scattered in the unknown sparse vector. In this article, based on the technique of pairwise fused lasso, we propose the novel pairwise fused lasso diffusion least mean-square (PFL-DLMS) algorithm, to identify sparse vector. The objective function we construct consists of three terms, i.e., the mean-square error (MSE) term, the regularizing term promoting the sparsity of all components, and the regularizing term promoting the sparsity of difference between each pair of components in the unknown sparse vector. After investigating mean stability condition of mean-square behavior in theoretical analysis, we propose the strategy of variable regularizing coefficients to overcome the difficulty that the optimal regularizing coefficients are usually unknown. Finally, numerical experiments are conducted to verify the effectiveness of the PFL-DLMS algorithm in identifying and tracking sparse parameter vector.

16.
Jpn J Infect Dis ; 76(4): 240-245, 2023 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-37121674

RESUMEN

Osteomyelitis is the infection and destruction of the bone. To date, there is no universal protocol for its treatment. Receptor-interacting serine/threonine-protein kinase 2 (RIPK2) has been implicated in osteomyelitis development. However, the detailed mechanism remains unknown. Here, 6-8w wild-type or Pellino E3 Ubiquitin Protein Ligase Family Member 3 (Peli3)-deficient mice were injected with Staphylococcus aureus to induce osteomyelitis. RAW264.7 cells or bone marrow-derived macrophages isolated from mice were treated with lipopolysaccharide (LPS). Knocking down Peli3 in RAW264.7 cells increased the expression of inflammatory cytokines (interleukin-1ß, interleukin-6, and tumor necrosis factor-α) after LPS stimulation. Inflammation was also activated in S. aureus-induced Peli3-deficient mice. Moreover, S. aureus-infected Peli3-deficient mice also displayed more severe symptoms of osteomyelitis than S. aureus-infected wild-type mice. Moreover, Peli3 targets and degrades RIPK2 through K48-linked ubiquitination, and negatively modulates osteomyelitis by degrading RIPK2. Our data further expands the current understanding of RIPK2 in osteomyelitis, and suggests that RIPK2 might serve as a novel therapeutic target for treating osteomyelitis.


Asunto(s)
Lipopolisacáridos , Osteomielitis , Animales , Ratones , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Staphylococcus aureus , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
17.
Jpn J Infect Dis ; 76(3): 197-203, 2023 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-36858599

RESUMEN

Receptor-interacting serine/threonine kinase (RIPK) is associated with cellular inflammation and immune regulation. The current study explored the role of RIPK2 in osteomyelitis and the potential upstream targets of RIPK2. A Staphylococcus aureus-induced osteomyelitis mouse model was established using wild-type (WT) and ubiquitin-specific peptidase 8 (USP8)-deficient (USP-/-) mice, and the osteomyelitis-related symptoms were evaluated. Bone marrow-derived macrophages (BMDMs) were isolated from the WT and USP-/- mice. Enzyme-linked immunosorbent assays, quantitative polymerase chain reaction, and immunoblot analysis were used to determine the levels of target biomarkers, which were induced by lipopolysaccharide (LPS), CpG, or PAM3CSK4. USP8 promoted RIPK2-mediated NF-κB activation. USP8 is indispensable for RIPK2-mediated LPS-induced NF-κB activation in BMDMs. USP8 is required for the production of inflammatory cytokines induced by LPS, CpG, or PAM3CSK4 in BMDMs. In addition, USP-/- mice exhibited ameliorated symptoms, including less body weight and cortical bone loss, and reduced bacterial load and reactive bone formation in the S. aureus-induced osteomyelitis mouse model. USP8 is critical in the S. aureus-induced osteomyelitis mouse model by targeting RIPK2 ubiquitination.


Asunto(s)
Enfermedades Transmisibles , Osteomielitis , Ratones , Animales , FN-kappa B , Lipopolisacáridos/farmacología , Staphylococcus aureus , Ubiquitinación , Proteasas Ubiquitina-Específicas/genética , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor
18.
Cell Prolif ; 56(11): e13485, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37186483

RESUMEN

We investigated the role of astragaloside IV (AS-IV) in preventing glucocorticoid-induced avascular necrosis of the femoral head (ANFH) and the underlying molecular mechanisms. Network pharmacology was used to predict the molecular targets of AS-IV. Molecular dynamic simulations were performed to explore the binding mechanism and interaction mode between AS-IV and Akt. Rat models of glucocorticoid-induced ANFH with AS-IV intervention were established, and osteogenesis, angiogenesis, apoptosis and oxidative stress were evaluated before and after blocking the PI3K/Akt pathway with LY294002. The effects of glucocorticoid and AS-IV on bone marrow mesenchymal stem cells and human umbilical vein endothelial cells incubated with and without LY294002 were determined. Downregulated p-Akt expression could be detected in the femoral heads of glucocorticoid-induced ANFH patients and rats. AS-IV increased trabecular bone integrity and vessel density of the femoral head in the model rats. AS-IV increased Akt phosphorylation and upregulated osteogenesis-, angiogenesis-, apoptosis- and oxidative stress-related proteins and mRNA and downregulated Bax, cleaved caspase-3 and cytochrome c levels. AS-IV promoted human umbilical vein endothelial cell migration, proliferation and tube formation ability; bone marrow mesenchymal stem cell proliferation; and osteogenic differentiation under glucocorticoid influence. AS-IV inhibited apoptosis. LY294002 inhibited these effects. AS-IV prevented glucocorticoid-induced ANFH by promoting osteogenesis and angiogenesis via the Akt/Runx2 and Akt/HIF-1α/VEGF pathways, respectively, and suppressing apoptosis and oxidative stress via the Akt/Bad/Bcl-2 and Akt/Nrf2/HO-1 pathways, respectively.


Asunto(s)
Necrosis de la Cabeza Femoral , Glucocorticoides , Humanos , Ratas , Animales , Glucocorticoides/efectos adversos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Osteogénesis , Fosfatidilinositol 3-Quinasas , Necrosis de la Cabeza Femoral/inducido químicamente , Necrosis de la Cabeza Femoral/tratamiento farmacológico , Células Endoteliales de la Vena Umbilical Humana/metabolismo
19.
J Ginseng Res ; 46(4): 526-535, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35818420

RESUMEN

Background: During the pathogenesis of tendinopathy, the chronic inflammation caused by the injury and apoptosis leads to the generation of scars. Ginsenoside Rg1 (Rg1) is extracted from ginseng and has anti-inflammatory effects. Rg1 is a unique phytoestrogen that can activate the estrogen response element. This research aimed to explore whether Rg1 can function in the process of tendon repair through the estrogen receptor. Methods: In this research, the effects of Rg1 were evaluated in tenocytes and in a rat model of Achilles tendinitis (AT). Protein levels were shown by western blotting. qRT-PCR was employed for evaluating mRNA levels. Cell proliferation was evaluated through EdU assay and cell migration was evaluated by transwell assay and scratch test assay. Results: Rg1 up-regulated the expression of matrix-related factors and function of tendon in AT rat model. Rg1 reduced early inflammatory response and apoptosis in the tendon tissue of AT rat model. Rg1 promoted tenocyte migration and proliferation. The effects of Rg1 on tenocytes were inhibited by ICI182780. Rg1 activates the insulin-like growth factor-I receptor (IGF1R) and MAPK signaling pathway. Conclusion: Rg1 promotes injured tendon healing in AT rat model through IGF1R and MAPK signaling pathway activation.

20.
Immune Netw ; 22(3): e25, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35799706

RESUMEN

IL-34 can promote osteoclast differentiation and activation, which may contribute to steroid-induced osteonecrosis of the femoral head (ONFH). Animal model was constructed in both BALB/c and IL-34 deficient mice to detect the relative expression of inflammation cytokines. Micro-CT was utilized to reveal the internal structure. In vitro differentiated osteoclast was induced by culturing bone marrow-derived macrophages with IL-34 conditioned medium or M-CSF. The relative expression of pro-inflammation cytokines, osteoclast marker genes, and relevant pathways molecules was detected with quantitative real-time RT-PCR, ELISA, and Western blot. Up-regulated IL-34 expression could be detected in the serum of ONFH patients and femoral heads of ONFH mice. IL-34 deficient mice showed the resistance to ONFH induction with the up-regulated trabecular number, trabecular thickness, bone value fraction, and down-regulated trabecular separation. On the other hand, inflammatory cytokines, such as TNF-α, IFN-γ, IL-6, IL-12, IL-2, and IL-17A, showed diminished expression in IL-34 deficient ONFH induced mice. IL-34 alone or works in coordination with M-CSF to promote osteoclastogenesis and activate ERK, STAT3, and non-canonical NF-κB pathways. These data demonstrate that IL-34 can promote the differentiation of osteoclast through ERK, STAT3, and non-canonical NF-κB pathways to aggravate steroid-induced ONFH, and IL-34 can be considered as a treatment target.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA