Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
World J Stem Cells ; 15(8): 787-806, 2023 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-37700823

RESUMEN

BACKGROUND: The immunosuppressive capacity of mesenchymal stem cells (MSCs) is dependent on the "license" of several proinflammatory factors to express immunosuppressive factors such as programmed cell death 1 ligand 1 (PD-L1), which determines the clinical therapeutic efficacy of MSCs for inflammatory or immune diseases. In MSCs, interferon-gamma (IFN-γ) is a key inducer of PD-L1 expression, which is synergistically enhanced by tumor necrosis factor-alpha (TNF-α); however, the underlying mechanism is unclear. AIM: To reveal the mechanism of pretreated MSCs express high PD-L1 and explore the application of pretreated MSCs in ulcerative colitis. METHODS: We assessed PD-L1 expression in human umbilical-cord-derived MSCs (hUC-MSCs) induced by IFN-γ and TNF-α, alone or in combination. Additionally, we performed signal pathway inhibitor experiments as well as RNA interference experiments to elucidate the molecular mechanism by which IFN-γ alone or in combination with TNF-α induces PD-L1 expression. Moreover, we used luciferase reporter gene experiments to verify the binding sites of the transcription factors of each signal transduction pathway to the targeted gene promoters. Finally, we evaluated the immunosuppressive capacity of hUC-MSCs treated with IFN-γ and TNF-α in both an in vitro mixed lymphocyte culture assay, and in vivo in mice with dextran sulfate sodium-induced acute colitis. RESULTS: Our results suggest that IFN-γ induction alone upregulates PD-L1 expression in hUC-MSCs while TNF-α alone does not, and that the co-induction of IFN-γ and TNF-α promotes higher expression of PD-L1. IFN-γ induces hUC-MSCs to express PD-L1, in which IFN-γ activates the JAK/STAT1 signaling pathway, up-regulates the expression of the interferon regulatory factor 1 (IRF1) transcription factor, promotes the binding of IRF1 and the PD-L1 gene promoter, and finally promotes PD-L1 mRNA. Although TNF-α alone did not induce PD-L1 expression in hUC-MSCs, the addition of TNF-α significantly enhanced IFN-γ-induced JAK/STAT1/IRF1 activation. TNF-α up-regulated IFN-γ receptor expression through activation of the nuclear factor kappa-B signaling pathway, which significantly enhanced IFN-γ signaling. Finally, co-induced hUC-MSCs have a stronger inhibitory effect on lymphocyte proliferation, and significantly ameliorate weight loss, mucosal damage, inflammatory cell infiltration, and up-regulation of inflammatory factors in colitis mice. CONCLUSION: Overall, our results suggest that IFN-γ and TNF-α enhance both the immunosuppressive ability of hUC-MSCs and their efficacy in ulcerative colitis by synergistically inducing high expression of PD-L1.

2.
Immun Inflamm Dis ; 11(5): e846, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37249292

RESUMEN

OBJECTIVES: To reveal the relationship between the fibulin-2 protein and immune dysfunction after bone trauma. METHODS: Individuals who were admitted to the study were divided into a bone trauma group, a recovered from bone trauma group and a volunteer without bone trauma group based on the reason for admission. Fibulin-2 levels in the three groups were compared. Fibulin-2-knockout (fibulin-2-/- ) mice and wild-type (WT) mice were used to detect susceptibility to infection. Hematoxylin and eosin (HE) staining and immunohistochemical staining were employed to observe pathological changes in each organ from fibulin-2-/- mice and WT mice. RESULTS: In total, 132 patients were enrolled in this study. The fibulin-2 level in the bone trauma group was lower than that in the recovered bone trauma group (3.39 ± 1.41 vs. 4.30 ± 1.38 ng/mL, t = 2.948, p < .05) and also lower than that in the volunteers without bone trauma group (3.39 ± 1.41 vs. 4.73 ± 1.67 ng/mL, t = 4.135, p < .05). Fibulin-2-/- mice are more prone to infection. Compared with those in WT mice, spleen function and thymus function in fibulin-2-/- mice were impaired. Immunohistochemical staining revealed that compared with those in WT mice, significantly fewer CD4+ T cells, CD8+ T cells, and CD19+ B cells were noted in the spleen and thymus of fibulin-2-/- mice. CONCLUSIONS: The plasma fibulin-2 level was lower in patients with bone trauma. Decreased fibulin-2 is associated with immune dysfunction after bone trauma.


Asunto(s)
Huesos , Proteínas de Unión al Calcio , Proteínas de la Matriz Extracelular , Sistema Inmunológico , Animales , Ratones , Proteínas de Unión al Calcio/metabolismo , Proteínas de la Matriz Extracelular/genética , Huesos/lesiones , Sistema Inmunológico/fisiopatología
3.
Exp Mol Med ; 55(2): 443-456, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36797542

RESUMEN

Bone fracture remains a common occurrence, with a population-weighted incidence of approximately 3.21 per 1000. In addition, approximately 2% to 50% of patients with skeletal fractures will develop an infection, one of the causes of disordered bone healing. Dysfunction of bone marrow mesenchymal stem cells (BMSCs) plays a key role in disordered bone repair. However, the specific mechanisms underlying BMSC dysfunction caused by bone infection are largely unknown. In this study, we discovered that Fibulin2 expression was upregulated in infected bone tissues and that BMSCs were the source of infection-induced Fibulin2. Importantly, Fibulin2 knockout accelerated mineralized bone formation during skeletal development and inhibited inflammatory bone resorption. We demonstrated that Fibulin2 suppressed BMSC osteogenic differentiation by binding to Notch2 and inactivating the Notch2 signaling pathway. Moreover, Fibulin2 knockdown restored Notch2 pathway activation and promoted BMSC osteogenesis; these outcomes were abolished by DAPT, a Notch inhibitor. Furthermore, transplanted Fibulin2 knockdown BMSCs displayed better bone repair potential in vivo. Altogether, Fibulin2 is a negative regulator of BMSC osteogenic differentiation that inhibits osteogenesis by inactivating the Notch2 signaling pathway in infected bone.


Asunto(s)
Curación de Fractura , Osteogénesis , Humanos , Huesos , Diferenciación Celular/genética , Células Cultivadas , Curación de Fractura/genética , Osteogénesis/genética , Transducción de Señal , Células de la Médula Ósea/metabolismo , Células Madre/metabolismo
4.
Infect Dis Ther ; 11(3): 1057-1073, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35303288

RESUMEN

INTRODUCTION: Infection remains a major cause of morbidity and mortality in hospital. As uncontrolled early infection may develop into systemic infection and eventually progress to sepsis, it is important to address infection at an early stage. Furthermore, early detection and prompt diagnosis of infection are the basis of clinical intervention. However, as a result of the interference of complex aetiologies, including fever and trauma, problems regarding the sensitivity and specificity of current diagnostic indices remain, such as for C-reactive protein (CRP), procalcitonin (PCT), white blood cells (WBC), neutrophil ratio (NEU%), interleukin-6 (IL-6) and D-dimer. As a result, there is an urgent need to develop new biomarkers to diagnose infection. METHODS: From January to October 2021, consecutive patients in the emergency department (ED) were recruited to investigate the feasibility of fibulin-2 as a diagnostic indicator of early infection. Fibulin-2 concentrations in plasma were determined with enzyme-linked immunosorbent assay (ELISA). The performance of fibulin-2 for predicting infection was analysed by receiver operating characteristic (ROC) curves. RESULTS: We found that the plasma fibulin-2 level was elevated in patients with infection compared with those without infection. ROC curve analysis showed that the area under the curve (AUC) for fibulin-2 was 0.712. For all patients included, the diagnostic ability of fibulin-2 (AUC 0.712) performed as well as CRP (AUC 0.667) and PCT (AUC 0.632), and better than WBC (AUC 0.620), NEU% (AUC 0.619), IL-6 (AUC 0.561) and D-dimer (AUC 0.630). In patients with fever, fibulin-2 performed as well as PCT and better than the other biomarkers in infection diagnosis. In particular, fibulin-2 performed better than all these biomarkers in patients with trauma. CONCLUSION: Fibulin-2 is a novel promising diagnostic biomarker for predicting infection.

5.
J Biomater Appl ; 36(7): 1231-1242, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34723682

RESUMEN

Implant-related infection is a disastrous complication. Surface modification of titanium is considered as an important strategy to prevent implant-related infection. However, there is no recognized surface modification strategy that can be applied in clinic so far. We explored a new strategy of coating. The clindamycin-loaded titanium was constructed by layer-by-layer self-assembly. The release of clindamycin from titanium was detected through high performance liquid chromatography. Different titanium was co-cultured with Staphylococcus aureus for 24 h in vitro, then the effect of different titanium on bacterial colonization and biofilm formation was determined by spread plate method and scanning electron microscopy. Cytotoxicity and cytocompatibility of clindamycin-loaded titanium on MC3T3-E1 cells were measured by CCK8. The antibacterial ability of clindamycin-loaded titanium in vivo was also evaluated using a rat model of osteomyelitis. The number of osteoclasts in bone defect was observed by tartrate-resistant acid phosphatase staining. Bacterial burden of surrounding tissues around the site of infection was calculated by tissue homogenate and colony count. Clindamycin-loaded titanium could release clindamycin slowly within 160 h. It reduced bacterial colonization by three orders of magnitude compare to control (p < .05) and inhibits biofilm formation in vitro. Cells proliferation and adhesion were similar on three titanium surfaces (p > .05). In vivo, clindamycin-loaded titanium improved bone healing, reduced microbial burden, and decreased the number of osteoclasts compared control titanium in the rat model of osteomyelitis. This study demonstrated that clindamycin-loaded titanium exhibited good biocompatibility, and showed antibacterial activity both in vivo and in vitro. It is promising and might have potential for clinical application.


Asunto(s)
Clindamicina , Titanio , Animales , Antibacterianos/química , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Biopelículas , Clindamicina/farmacología , Clindamicina/uso terapéutico , Materiales Biocompatibles Revestidos/química , Ratas , Staphylococcus aureus , Titanio/química
6.
Platelets ; 32(4): 479-491, 2021 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-32396493

RESUMEN

To be faced with an infected bone defect and the need to accelerate bone union while controlling infection is a welcome challenge for orthopedists. Platelet-rich plasma (PRP) has been applied in tissue defects given their composition of growth factors however the weak antibacterial effects have limited the use of PRP in the clinical setting. Therefore, the aim of this study was to explore the feasibility of using PRP in a local antibiotic delivery system (PADS) with the characteristics of promoting wound healing of bone infection. PADS was prepared with the addition of antibiotics or no antibiotics as control after PRP was prepared by a two-step centrifugation procedure. Antibacterial tests showed zones of inhibition produced by antibiotics were not significantly different with antibiotics combined with PRP. HPLC analysis demonstrated that about 60% of the total vancomycin (VAN) and ceftazidime (CAZ) dose were released within 10 min, then the release rate gradually decreased. However, 90% clindamycin was released within 10 min. Interestingly, above 10 times the minimum inhibitory concentration was presented after 72 h. Additionally, ELISA and morphology studies of PADS indicated that loaded antibiotics could reduce the PRP-released growth factor concentration and disturb the structure of platelet-fibrin beams and fibrin network in a dose-dependent manner. Fortunately, the lower dose of antibiotics maintained their anti-microbial effect, meanwhile growth factors released from PADS, the structure of platelet-fibrin beams, fibrin network remained unaffected. In addition, a patient experiencing infected bone defect receiving this PADS treatment achieved union within the 15-month follow-up. Therefore, this novel PADS approach might represent a potential therapy for patients who have sustained infected bone defects.


Asunto(s)
Antibacterianos/uso terapéutico , Plasma Rico en Plaquetas/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Adulto , Antibacterianos/farmacología , Humanos , Masculino
7.
Artículo en Inglés | MEDLINE | ID: mdl-25953563

RESUMEN

A new series of Fe80-x-yCrxMoyP13C7 (x = 10, y = 10; x = 20, y = 5; x = 2 0, y = 10, all in at.%) bulk metallic glasses (BMGs) with the maximum diameter of 6mm have been developed for biomedical implant application by the combination method of fluxing treatment and J-quenching technique. The corrosion performance of the present Fe-based BMGs is investigated in both Hank's solution (pH = 7.4) and artificial saliva solution (pH = 6.3) at 37 °C by electrochemical measurements. The result indicates that the corrosion resistance of the present Fe-based BMGs in the above two simulated body solutions is much better than that of biomedical 316 L stainless steel (316 L SS), and approaching that of Ti6Al4V biomedical alloy (TC4). The concentrations of Fe, Ni and Cr ions released into the Hank's solution and artificial saliva solution from the present Fe-based BMGs after potentiodynamic polarization are significant lower than that released from 316 L SS. The biocompatibility of the present Fe-based BMGs is evaluated through the in vitro test of NIH3T3 cells culture in the present Fe-based BMG extraction media for 1, 3 and 5 days. The result indicates that the present Fe-based BMGs exhibit no cytotoxicity to NIH3T3 cells. And the test result of the cell adhesion and growth on the surface of the samples indicates that the present Fe-based BMGs exhibit the better cell viability compared with 316 L SS and TC4 biomedical alloys. The present Fe-based BMGs, especially Fe55Cr20Mo5P13C7 BMG, exhibit good glass formation ability, the high corrosion resistance and excellent biocompatibility, suggesting their promising potential as biomaterials.


Asunto(s)
Materiales Biocompatibles/química , Nanocompuestos/química , Benzofuranos/química , Materiales Biocompatibles/farmacología , Ensayo de Materiales , Microscopía Electrónica de Transmisión , Staphylococcus epidermidis/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...