Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 731: 150388, 2024 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-39024974

RESUMEN

The poor healing characteristics of diabetic foot ulcers are partially attributed to diabetes-induced pro-inflammatory wounds. Our previous study reported that both miR-146a-5p and miR-200b-3p decrease endothelial inflammation in human aortic endothelial cells and db/db diabetic mice. Although miR-146a-5p has been reported to improve diabetic wound healing, the role of miR-200b-3p is not clear. This study compared the roles of these miRNAs in diabetic wound healing. Two 8-mm full-thickness wounds were created in 12-week-old male db/db mice on the left and right back. After surgery, 100 ng miR-146a-5p, miR-200b-3p, or miR-negative control (NC) was injected in each wound. Full-thickness skin samples were harvested from mice at the 14th day for real-time polymerase chain reaction and immunohistochemistry analyses. At the 14th day, the miR-200b-3p group showed better wound healing and greater granulation tissue thickness than the miR-146a-5p group. The miR-200b-3p group showed a significant decrease of IL-6 and IL-1ß gene expression and a significant increase of Col3α1 gene expression compared to those in the miR-NC group. The miR-200b-3p group had the lowest gene expression of TGF-ß1, followed by the miR-146a-5p and miR-NC groups. Our findings suggest that the miR-200b-3p group had better healing characteristics than the other two groups. Immunohistochemical staining revealed that CD68 immunoreactivity was significantly decreased in both the miR-146a-5p and miR-200b-3p groups compared with that in the miR-NC group. In addition, CD31 immunoreactivity was significantly higher in the miR-200b-3p group than in the miR-146a-5p group. In conclusion, these results suggest that miR-200b-3p is more effective than miR-146a-5p in promoting diabetic wound healing through its anti-inflammatory and pro-angiogenic effects.


Asunto(s)
MicroARNs , Cicatrización de Heridas , MicroARNs/genética , MicroARNs/metabolismo , Animales , Cicatrización de Heridas/genética , Masculino , Ratones , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Pie Diabético/genética , Pie Diabético/metabolismo , Pie Diabético/patología , Neovascularización Fisiológica/genética , Interleucina-6/metabolismo , Interleucina-6/genética , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Antígenos CD/genética , Antígenos CD/metabolismo , Piel/metabolismo , Piel/patología , Inflamación/genética , Inflamación/patología , Inflamación/metabolismo , Ratones Endogámicos C57BL , Molécula CD68
2.
J Nanobiotechnology ; 22(1): 382, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951872

RESUMEN

Reperfusion therapy is critical for saving heart muscle after myocardial infarction, but the process of restoring blood flow can itself exacerbate injury to the myocardium. This phenomenon is known as myocardial ischemia-reperfusion injury (MIRI), which includes oxidative stress, inflammation, and further cell death. microRNA-146a (miR-146a) is known to play a significant role in regulating the immune response and inflammation, and has been studied for its potential impact on the improvement of heart function after myocardial injury. However, the delivery of miR-146a to the heart in a specific and efficient manner remains a challenge as extracellular RNAs are unstable and rapidly degraded. Milk exosomes (MEs) have been proposed as ideal delivery platform for miRNA-based therapy as they can protect miRNAs from RNase degradation. In this study, the effects of miR-146a containing MEs (MEs-miR-146a) on improvement of cardiac function were examined in a rat model of MIRI. To enhance the targeting delivery of MEs-miR-146a to the site of myocardial injury, the ischemic myocardium-targeted peptide IMTP was modified onto the surfaces, and whether the modified MEs-miR-146a could exert a better therapeutic role was examined by echocardiography, myocardial injury indicators and the levels of inflammatory factors. Furthermore, the expressions of miR-146a mediated NF-κB signaling pathway-related proteins were detected by western blotting and qRT-PCR to further elucidate its mechanisms. MiR-146 mimics were successfully loaded into the MEs by electroporation at a square wave 1000 V voltage and 0.1 ms pulse duration. MEs-miR-146a can be up-taken by cardiomyocytes and protected the cells from oxygen glucose deprivation/reperfusion induced damage in vitro. Oral administration of MEs-miR-146a decreased myocardial tissue apoptosis and the expression of inflammatory factors and improved cardiac function after MIRI. The miR-146a level in myocardium tissues was significantly increased after the administration IMTP modified MEs-miR-146a, which was higher than that of the MEs-miR-146a group. In addition, intravenous injection of IMTP modified MEs-miR-146a enhanced the targeting to heart, improved cardiac function, reduced myocardial tissue apoptosis and suppressed inflammation after MIRI, which was more effective than the MEs-miR-146a treatment. Moreover, IMTP modified MEs-miR-146a reduced the protein levels of IRAK1, TRAF6 and p-p65. Therefore, IMTP modified MEs-miR-146a exerted their anti-inflammatory effect by inhibiting the IRAK1/TRAF6/NF-κB signaling pathway. Taken together, our findings suggested miR-146a containing MEs may be a promising strategy for the treatment of MIRI with better outcome after modification with ischemic myocardium-targeted peptide, which was expected to be applied in clinical practice in future.


Asunto(s)
Exosomas , MicroARNs , Daño por Reperfusión Miocárdica , FN-kappa B , Ratas Sprague-Dawley , Transducción de Señal , Animales , MicroARNs/metabolismo , MicroARNs/genética , Daño por Reperfusión Miocárdica/metabolismo , Exosomas/metabolismo , FN-kappa B/metabolismo , Ratas , Masculino , Leche/química , Miocardio/metabolismo , Cardiotónicos/farmacología , Miocitos Cardíacos/metabolismo
3.
Eur Spine J ; 33(5): 2056-2067, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38551688

RESUMEN

OBJECTIVE: This research was to unravel the impact of the lncRNA differentiation antagonizing non-protein coding RNA (DANCR)/microRNA (miR)-146a-5p/mitogen-activated protein kinase 6 (MAPK6) axis on spinal cord injury (SCI). METHODS: SCI mouse models were established and injected with si-DANCR or miR-146a-5p agomir. The recovery of motor function was assessed by Basso Mouse Scale. SCI was pathologically evaluated, and serum inflammatory factors were measured in SCI mice. Mouse spinal cord neurons were injured by H2O2 and transfected, followed by assessment of proliferation and apoptosis. DANCR, miR-146a-5p, and MAPK6 in tissues and cells were detected, as well as their relationship. RESULTS: DANCR increased and miR-146a-5p decreased in SCI. Silencing DANCR or enhancing miR-146a-5p stimulated the proliferation of mouse spinal cord neurons and reduced apoptosis. DANCR was bound to miR-146a-5p to target MAPK6. DANCR affected the proliferation and apoptosis of spinal cord neurons by mediating the miR-146a-5p/MAPK6 axis. Downregulating DANCR or upregulating miR-146a-5p improved inflammation, the destruction of spinal cord tissue structure, and apoptosis in SCI mice. CONCLUSION: DANCR affects spinal cord neuron apoptosis and inflammation of SCI by mediating the miR-146a-5p/MAPK6 axis.


Asunto(s)
Apoptosis , MicroARNs , Neuronas , ARN Largo no Codificante , Traumatismos de la Médula Espinal , Animales , Masculino , Ratones , Inflamación/genética , Inflamación/metabolismo , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Proteína Quinasa 6 Activada por Mitógenos/genética , Proteína Quinasa 6 Activada por Mitógenos/metabolismo , Neuronas/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología
4.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(3): 630-634, 2024 May 20.
Artículo en Zh | MEDLINE | ID: mdl-38948270

RESUMEN

Objective: To investigate the effect of Sanshentongmai (SSTM) mixture on the regulation of oxidative damage to rat cardiomyocytes (H9C2) through microRNA-146a and its mechanism. Methods: H9C2 were cultured in vitro, H2O2 was used as an oxidant to create an oxidative damage model in H9C2 cells. SSTM intervention was administered to the H9C2 cells. Then, the changes in H2O2-induced oxidative damage in H9C2 cells and the expression of microRNA-146a were observed to explore the protective effect of SSTM on H9C2 and its mechanism. H9C2 cells cultured i n vitro were divided into 3 groups, including a control group, a model group of H2O2-induced oxidative damage (referred to hereafter as the model group), and a group given H2O2 modeling plus SSTM intervention at 500 µg/L for 72 h (referred to hereafter as the treatment group). The cell viability was measured by CCK8 assay. In addition, the levels of N-terminal pro-brain natriuretic peptide (Nt-proBNP), nitric oxide (NO), high-sensitivity C-reactive protein (Hs-CRP), and angiotensin were determined by enzyme-linked immunosorbent assay (ELISA). The expression level of microRNA-146a was determined by real-time PCR (RT-PCR). Result: H9C2 cells were pretreated with SSTM at mass concentrations ranging from 200 to 1500 µg/L. Then, CCK8 assay was performed to measure cell viability and the findings showed that the improvement in cell proliferation reached its peak when the mass concentration of SSTM was 500 µg/L, which was subsequently used as the intervention concentration. ELISA was performed to measure the indicators related to heart failure, including Nt-proBNP, NO, Hs-CRP, and angiotensin Ⅱ. Compared with those of the control group, the expressions of Nt-proBNP and angiotensin Ⅱ in the treatment group were up-regulated (P<0.05), while the expression of NO was down-regulated (P<0.05). There was no significant difference in the expression of Hs-CRP between the treatment group and the control group. These findings indicate that SSTM could effectively ameliorate oxidative damage in H9C2 rat cardiomyocytes. Finally, according to the RT-PCR findings for the expression of microRNA-146a in each group, H2O2 treatment at 15 µmol/L could significantly reduce the expression of microRNA-146a, and the expression of microRNA-146a in the treatment group was nearly doubled compared with that in the model group. There was no significant difference between the treatment group and the control group. Conclusion: SSTM can significantly resist the H2O2-induced oxidative damage of H9C2 cells and may play a myocardial protective role by upregulating microRNA-146a.


Asunto(s)
Medicamentos Herbarios Chinos , Peróxido de Hidrógeno , MicroARNs , Miocitos Cardíacos , Estrés Oxidativo , Regulación hacia Arriba , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/citología , Animales , MicroARNs/metabolismo , MicroARNs/genética , Ratas , Estrés Oxidativo/efectos de los fármacos , Peróxido de Hidrógeno/toxicidad , Medicamentos Herbarios Chinos/farmacología , Regulación hacia Arriba/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Línea Celular , Combinación de Medicamentos
5.
Am J Physiol Cell Physiol ; 324(5): C1007-C1016, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36939200

RESUMEN

MicroRNAs play a critical role in bone marrow mesenchymal stem cell (MSC) chondrogenesis and regulate the progression of joint regeneration in osteoarthritis. Our previous research confirmed that miR146a relieves osteoarthritis by modulating cartilage homeostasis. However, few studies have revealed the relationship between miR146a and the chondrogenesis of MSCs, and the exact mechanisms remain unclear. This study aimed to determine the function of miR146a in the chondrogenic differentiation of MSCs and the potential mechanisms involved. MiR146a expression increased during chondrogenesis. MiR146a knockout (KO) led to the increased chondrogenesis of MSCs compared to that in wild-type (WT) MSCs, whereas the overexpression of miR146a by mimics resulted in the decreased chondrogenesis of MSCs, as determined by the mRNA expression of collagen, type II, alpha 1 (COL2A1), aggrecan, cartilage oligomeric matrix protein (COMP), and matrix metallopeptidase 13 (MMP13). Furthermore, cartilage defects could be treated better when injected with spheres induced from miR146aKO MSCs than from WT MSCs, indicating that miR146a inhibits chondrogenesis in vivo. In addition, based on miRNA-mRNA prediction analysis and a dual-luciferase reporter assay, we observed that the deletion of miR146a led to the increased expression of Lsm11 during chondrogenesis and demonstrated that miR146a targeted Lsm11 by binding to its 3'-untranslated region (UTR) and inhibited its translation. The inhibition of Lsm11 by silencing RNA (siRNA) reversed the increased ability of chondrogenesis by knocking out miR146a both in vivo and in vitro, suggesting that miR146a inhibits chondrogenesis by directly inhibiting Lsm11 in MSCs, which may be a novel target for treating osteoarthritis.


Asunto(s)
Células Madre Mesenquimatosas , MicroARNs , Osteoartritis , Humanos , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Condrocitos/metabolismo , Condrogénesis/genética , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Osteoartritis/genética , Osteoartritis/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo
6.
Lasers Med Sci ; 38(1): 129, 2023 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-37243832

RESUMEN

Diabetic wounds are categorized by chronic inflammation, leading to the development of diabetic foot ulcers, which cause amputation and death. Herewith, we examined the effect of photobiomodulation (PBM) plus allogeneic diabetic adipose tissue-derived stem cells (ad-ADS) on stereological parameters and expression levels of interleukin (IL)-1ß and microRNA (miRNA)-146a in the inflammatory (day 4) and proliferation (day 8) stages of wound healing in an ischemic infected (with 2×107 colony-forming units of methicillin-resistant Staphylococcus aureus) delayed healing wound model (IIDHWM) in type I diabetic (TIDM) rats. There were five groups of rats: group 1 control (C); group 2 (CELL) in which rat wounds received 1×106 ad-ADS; group 3 (CL) in which rat wounds received the ad-ADS and were subsequently exposed to PBM(890 nm, 80 Hz, 3.5 J/cm2, in vivo); group 4 (CP) in which the ad-ADS preconditioned by the PBM(630 nm + 810 nm, 0.05 W, 1.2 J/cm2, 3 times) were implanted into rat wounds; group 5 (CLP) in which the PBM preconditioned ad-ADS were implanted into rat wounds, which were then exposed to PBM. On both days, significantly better histological results were seen in all experimental groups except control. Significantly better histological results were observed in the ad-ADS plus PBM treatment correlated to the ad-ADS alone group (p<0.05). Overall, PBM preconditioned ad-ADS followed by PBM of the wound showed the most significant improvement in histological measures correlated to the other experimental groups (p<0.05). On days 4 and 8, IL-1 ß levels of all experimental groups were lower than the control group; however, on day 8, only the CLP group was different (p<0.01). On day 4, miR-146a expression levels were substantially greater in the CLP and CELL groups correlated to the other groups, on day 8 miR-146a in all treatment groups was upper than C (p<0.01). ad-ADS plus PBM, ad-ADS, and PBM all improved the inflammatory phase of wound healing in an IIDHWM in TIDM1 rats by reducing inflammatory cells (neutrophils, macrophages) and IL-1ß, and increasing miRNA-146a. The ad-ADS+PBM combination was better than either ad-ADS or PBM alone, because of the higher proliferative and anti-inflammatory effects of the PBM+ad-ADS regimen.


Asunto(s)
Diabetes Mellitus Experimental , Terapia por Luz de Baja Intensidad , Staphylococcus aureus Resistente a Meticilina , MicroARNs , Ratas , Animales , Diabetes Mellitus Experimental/patología , Ratas Wistar , Cicatrización de Heridas , Células Madre/patología , Inflamación/radioterapia , Terapia por Luz de Baja Intensidad/métodos , MicroARNs/genética
7.
Int J Mol Sci ; 24(5)2023 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-36902041

RESUMEN

Amyotrophic lateral sclerosis (ALS) is characterized by the progressive, irreversible loss of upper and lower motor neurons (UMNs, LMNs). MN axonal dysfunctions are emerging as relevant pathogenic events since the early ALS stages. However, the exact molecular mechanisms leading to MN axon degeneration in ALS still need to be clarified. MicroRNA (miRNA) dysregulation plays a critical role in the pathogenesis of neuromuscular diseases. These molecules represent promising biomarkers for these conditions since their expression in body fluids consistently reflects distinct pathophysiological states. Mir-146a has been reported to modulate the expression of the NFL gene, encoding the light chain of the neurofilament (NFL) protein, a recognized biomarker for ALS. Here, we analyzed miR-146a and Nfl expression in the sciatic nerve of G93A-SOD1 ALS mice during disease progression. The miRNA was also analyzed in the serum of affected mice and human patients, the last stratified relying on the predominant UMN or LMN clinical signs. We revealed a significant miR-146a increase and Nfl expression decrease in G93A-SOD1 peripheral nerve. In the serum of both ALS mice and human patients, the miRNA levels were reduced, discriminating UMN-predominant patients from the LMN ones. Our findings suggest a miR-146a contribution to peripheral axon impairment and its potential role as a diagnostic and prognostic biomarker for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , MicroARNs , Degeneración Nerviosa , Animales , Humanos , Ratones , Esclerosis Amiotrófica Lateral/diagnóstico , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Biomarcadores/sangre , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Ratones Transgénicos , MicroARNs/sangre , MicroARNs/genética , MicroARNs/metabolismo , Degeneración Nerviosa/diagnóstico , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Nervios Periféricos/patología , Superóxido Dismutasa-1/genética , Axones/patología , Proteínas de Neurofilamentos , Diagnóstico Precoz , Progresión de la Enfermedad
8.
Cell Tissue Res ; 390(1): 71-92, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35788900

RESUMEN

Despite advances in the treatment of acute myocardial infarction, due to the non-proliferative nature of adult cardiomyocytes, the injured myocardium is mainly replaced by fibrotic tissue, which ultimately causes heart failure. To prevent heart failure, particularly after myocardial infarction, exosome-based therapy has emerged as one of the most promising strategies to regenerate cardiac function. Exosomes can carry microRNAs in support of neovascularization, anti-inflammatory, and endogenous cardiac regeneration. This study demonstrated that animal rat models' combination treatment with microRNA-126 and microRNA-146a mimics in exosomes is desirable for cardiac regeneration after myocardial infarction. The exosomes isolated from stem cells and loaded with microRNAs were characterized their impacts in cell migration, tube formation, and vascular endothelial growth factor degree. In the following, the usefulness of loaded microRNAs in exosomes and their encapsulation within alginate derivative hydrogel was analyzed in myocardial infarction for an animal model. Exosomes isolated and loaded with microRNAs showed the synergetic impact on cell migration, tube formation, and promoted vascular endothelial growth factor folding. Moreover, microRNAs loaded exosomes and encapsulated them in alginate hydrogel could help in reducing infarct size and improving angiogenesis in myocardial infarction. The angiogenesis markers including CD31 and connexion 43 upregulated for myocardial infarction models treated with alginate-based hydrogels loaded with exosomes and microRNAs-exosomes. Histological analysis indicated that myocardial infarction model rats treated with alginate hydrogel loaded with microRNAs-exosomes possessed lower and higher degrees of fibrosis and collagen fiber, respectively. These findings have important therapeutic implications for a myocardial infarction model through angiogenesis and vascular integrity regulation.


Asunto(s)
Exosomas , Insuficiencia Cardíaca , MicroARNs , Infarto del Miocardio , Alginatos , Animales , Colágeno/metabolismo , Exosomas/metabolismo , Fibrosis , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Hidrogeles , MicroARNs/genética , MicroARNs/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , Ratas , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Immunol Invest ; 51(7): 1975-1993, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35723582

RESUMEN

Systemic lupus erythematosus (SLE)-associated diffuse alveolar hemorrhage (DAH) is a rare but extremely harmful condition. The current study sought to dissect the mechanisms underlying the effects of human umbilical cord mesenchymal stem cell (HUCMSC)-derived exosomes on M2 macrophage polarization in SLE-associated DAH via the microRNA (miR)-146a-5p/NOTCH1 axis. A DAH mouse model was established using pristane. Exosomes were isolated from HUCMSCs transfected or untransfected with the miR-146a-5p antagonist or agonist and their NCs and then injected into DAH mice. Additionally, miR-146a-5p was overexpressed in macrophages. Expression of miR-146a-5p, NOTCH1, M1 macrophage markers, and M2 macrophage markers was measured in mice and macrophages, and inflammatory factor levels were detected. Mouse lung injuries were evaluated, so was the binding of miR-146a-5p to NOTCH1. Rescue experiments were conducted in mice and macrophages using NOTCH1 shRNA and pcDNA3.1-NOTCH1, respectively. NOTCH1 expression was enhanced in DAH mice. HUCMSC-derived exosomes reduced NOTCH1 expression, bleeding, inflammation, and M1 macrophage polarization but elevated M2 macrophage polarization in lung tissues of DAH mice. Mechanistically, NOTCH1 is negatively targeted by miR-146a-5p. miR-146a-5p overexpression diminished M1 marker and inflammatory factor levels but enhanced M2 marker levels in macrophages, which was nullified by NOTCH1 overexpression. HUCMSC-derived exosomes with miR-146a-5p inhibition increased NOTCH1 expression, worsened bleeding and inflammation, and augmented M1 macrophage polarization while decreasing M2 macrophage polarization in lung tissues of DAH mice, which was abrogated by silencing NOTCH1. HUCMSC-derived exosomes diminished NOTCH1 expression to accelerate M2 macrophage polarization via delivery of miR-146a-5p, thus alleviating SLE-associated DAH in mice.


Asunto(s)
Exosomas , Lupus Eritematoso Sistémico , Células Madre Mesenquimatosas , MicroARNs , Animales , Exosomas/metabolismo , Hemorragia/metabolismo , Hemorragia/terapia , Humanos , Inflamación/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , ARN Interferente Pequeño/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Cordón Umbilical/metabolismo
10.
J Clin Lab Anal ; 36(2): e24198, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34952998

RESUMEN

BACKGROUND: MicroRNA (miR)-146a and miR-146b regulate autoimmunity, inflammation, and keratinocytes proliferation to engage in psoriasis pathology. The current study aimed to investigate their correlation with disease risk and clinical features, and the linkage of their longitudinal changes with clinical response to etanercept in psoriasis patients. METHODS: Plasma samples were collected from 84 moderate-to-severe psoriasis patients who underwent etanercept treatment (at baseline (M0), 1 month (M1), 3 months (M3), and 6 months (M6)), 80 disease controls and 80 health controls (both after enrollment); afterward, miR-146a and miR-146b expressions were detected by RT-qPCR. Furthermore, PASI75 and PASI90 responses were assessed in psoriasis patients. RESULTS: Both miR-146a and miR-146b were decreased in psoriasis patients compared with disease controls and health controls (all p < 0.001), which also distinguished psoriasis patients from disease controls and health controls by receiver-operating characteristic analyses. Furthermore, miR-146a positively correlated with miR-146b in psoriasis patients (p < 0.001) and disease controls (p = 0.005) but not in healthy controls (p = 0.062). In psoriasis patients, miR-146a negatively related to psoriatic body surface area (p = 0.011) and PASI score (p = 0.003); miR-146b negatively linked with PASI score (p = 0.020). At M1, M3, and M6 after etanercept treatment, PASI75 response rate was 14.3%, 32.1%, and 69.0%, respectively; PASI90 response rate was 1.2%, 17.9%, and 36.9%, respectively. During etanercept treatment, both miR-146a and miR-146b elevated gradually over time and their longitude increments were associated with PASI75 response (all p < 0.001). CONCLUSION: MiR-146a and miR-146b might serve as indicators for optimizing etanercept application and improving treatment outcomes in psoriasis patients.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Etanercept/uso terapéutico , MicroARNs , Psoriasis/genética , Adulto , Área Bajo la Curva , Estudios de Casos y Controles , Femenino , Humanos , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Psoriasis/tratamiento farmacológico , Psoriasis/fisiopatología
11.
Nanomedicine ; 40: 102498, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34838994

RESUMEN

Acute respiratory distress syndrome (ARDS) is a highly morbid pulmonary disease characterized by hypoxic respiratory failure. Its pathogenesis is characterized by unrestrained oxidative stress and inflammation, with long-term sequelae of pulmonary fibrosis and diminished lung function. Unfortunately, prior therapeutic ARDS trials have failed and therapy is limited to supportive measures. Free radical scavenging cerium oxide nanoparticles (CNP) conjugated to the anti-inflammatory microRNA-146a (miR146a), termed CNP-miR146a, have been shown to prevent acute lung injury in a pre-clinical model. In this study, we evaluated the potential of delayed treatment with CNP-miR146a at three or seven days after injury to rescue the lung from acute injury. We found that intratracheal CNP-miR146a administered three days after injury lowers pulmonary leukocyte infiltration, reduce inflammation and oxidative stress, lower pro-fibrotic gene expression and collagen deposition in the lung, and ultimately improve pulmonary function.


Asunto(s)
Lesión Pulmonar Aguda , Lesión Pulmonar , Nanopartículas , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/patología , Cerio , Humanos , Pulmón/patología , Lesión Pulmonar/patología , Tiempo de Tratamiento
12.
Microb Pathog ; 157: 104962, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34022359

RESUMEN

Cystic echinococcosis, an important zoonotic disease, is caused by Echinococcus granulosus. MicroRNAs are a small group of single-stranded noncoding RNAs, which play an effective role in biological processes. This study aimed at comparing the expression levels of miR-146a and miR-155 in the plasma of patients with hydatidosis and healthy individuals. A group of 20 patients with hydatid cyst formed a study group and 20 healthy individuals with no known chronic diseases formed a control group. Plasma samples were collected from hydatidosis patients as well as sex- and age-matched healthy volunteers. After that, RNA extraction and cDNA synthesis were done and the expression levels of miR-146a and miR-155 were determined by quantitative real-time polymerase chain reaction (PCR) for both groups. The results indicated that the level of miR-146a increased in all patients with hydatidosis compared to the control group. Also, the level of miR-155 increased in all hydatidosis patients, but no correlation was observed in the level of miR-155 between the two groups. The results also revealed that miR-146a and miR-155 upregulation in the plasma leads to the development of novel biomarkers for echinococcosis. One of the reasons for the increase of miRNAs in hydatidosis may be their role in modulating the immune system. These miRNAs are likely to be considered as one of the most important biomarkers in determining the severity of hydatidosis.


Asunto(s)
Equinococosis , MicroARNs , Animales , Biomarcadores , Equinococosis/diagnóstico , Equinococosis/inmunología , Humanos , Inmunidad , MicroARNs/sangre
13.
J Clin Lab Anal ; 35(1): e23571, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32935891

RESUMEN

OBJECTIVE: Dry eye syndrome in which tear fluid quality or abnormality, or kinetic abnormality is caused by various reasons, resulting in decreased tear film stability. In recent years, more and more results from the studies indicate that miRNA alterations are involved in dry eye syndrome. And miRNA-146a-5p is a key regulator to regulate the inflammatory response. In this paper, we demonstrated whether miRNA-146a-5p could cure dry eye syndrome by regulating target genes based on network analysis. METHODS: In current study, we collected the blood of patients with dry eye disease served as a model group; the blood of healthy people was served as control group. The expression of miRNA-146a-5p in the patients was detected by RT-PCR, the genes controlled by miRNA-146a-5p were predicted by TargetScan, miRDB, miRWalk, and PicTar databases, and the genes regulated by miRNA-146a-5p which relative with dry eye disease were selected by drawing Venn diagram. RESULTS: The comparison of the general information between patients and healthy people was no significant difference, and it indicated that the two groups were comparable. The results of databases showed that IRAK1 was one of the target genes regulated by miRNA-146a-5p, and it is related to dry eye disease. The expression of miRNA-146a-5p was negatively related to IRAK1 mRNA and protein, while IRAK1 had a positive correlation with IL-6, TNF-α, and CBP proteins. CONCLUSION: These results emphasized that miRNA-146a-5p could inhibit the expression of IRAK1, IL-6, TNF-α, and CBP to help reduce the inflammatory response in dry eye syndrome.


Asunto(s)
Síndromes de Ojo Seco , MicroARNs , Adulto , Animales , Estudios de Casos y Controles , Células Cultivadas , Biología Computacional , Citocinas/sangre , Citocinas/metabolismo , Síndromes de Ojo Seco/epidemiología , Síndromes de Ojo Seco/genética , Síndromes de Ojo Seco/metabolismo , Femenino , Humanos , Inflamación/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Aparato Lagrimal/citología , Masculino , MicroARNs/sangre , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Ratas
14.
Nanomedicine ; 34: 102388, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33753282

RESUMEN

Acute respiratory distress syndrome (ARDS) is a devastating pulmonary disease with significant in-hospital mortality and is the leading cause of death in COVID-19 patients. Excessive leukocyte recruitment, unregulated inflammation, and resultant fibrosis contribute to poor ARDS outcomes. Nanoparticle technology with cerium oxide nanoparticles (CNP) offers a mechanism by which unstable therapeutics such as the anti-inflammatory microRNA-146a can be locally delivered to the injured lung without systemic uptake. In this study, we evaluated the potential of the radical scavenging CNP conjugated to microRNA-146a (termed CNP-miR146a) in preventing acute lung injury (ALI) following exposure to bleomycin. We have found that intratracheal delivery of CNP-miR146a increases pulmonary levels of miR146a without systemic increases, and prevents ALI by altering leukocyte recruitment, reducing inflammation and oxidative stress, and decreasing collagen deposition, ultimately improving pulmonary biomechanics.


Asunto(s)
Bleomicina/efectos adversos , Cerio , Sistemas de Liberación de Medicamentos , MicroARNs , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Animales , Bleomicina/farmacología , COVID-19/genética , COVID-19/metabolismo , Cerio/química , Cerio/farmacología , Modelos Animales de Enfermedad , Masculino , Ratones , MicroARNs/química , MicroARNs/farmacología , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/genética , Síndrome de Dificultad Respiratoria/metabolismo , SARS-CoV-2/metabolismo , Tratamiento Farmacológico de COVID-19
15.
Int J Mol Sci ; 22(17)2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34502105

RESUMEN

The human brain and central nervous system (CNS) harbor a select sub-group of potentially pathogenic microRNAs (miRNAs), including a well-characterized NF-kB-sensitive Homo sapiens microRNA hsa-miRNA-146a-5p (miRNA-146a). miRNA-146a is significantly over-expressed in progressive and often lethal viral- and prion-mediated and related neurological syndromes associated with progressive inflammatory neurodegeneration. These include ~18 different viral-induced encephalopathies for which data are available, at least ~10 known prion diseases (PrD) of animals and humans, Alzheimer's disease (AD) and other sporadic and progressive age-related neurological disorders. Despite the apparent lack of nucleic acids in prions, both DNA- and RNA-containing viruses along with prions significantly induce miRNA-146a in the infected host, but whether this represents part of the host's adaptive immunity, innate-immune response or a mechanism to enable the invading prion or virus a successful infection is not well understood. Current findings suggest an early and highly interactive role for miRNA-146a: (i) as a major small noncoding RNA (sncRNA) regulator of innate-immune responses and inflammatory signaling in cells of the human brain and CNS; (ii) as a critical component of the complement system and immune-related neurological dysfunction; (iii) as an inducible sncRNA of the brain and CNS that lies at a critical intersection of several important neurobiological adaptive immune response processes with highly interactive associations involving complement factor H (CFH), Toll-like receptor pathways, the innate-immunity, cytokine production, apoptosis and neural cell decline; and (iv) as a potential biomarker for viral infection, TSE and AD and other neurological diseases in both animals and humans. In this report, we review the recent data supporting the idea that miRNA-146a may represent a novel and unique sncRNA-based biomarker for inflammatory neurodegeneration in multiple species. This paper further reviews the current state of knowledge regarding the nature and mechanism of miRNA-146a in viral and prion infection of the human brain and CNS with reference to AD wherever possible.


Asunto(s)
Encéfalo/patología , Enfermedades Virales del Sistema Nervioso Central/inmunología , Regulación de la Expresión Génica/inmunología , MicroARNs/metabolismo , Enfermedades por Prión/inmunología , Apoptosis/genética , Apoptosis/inmunología , Biomarcadores/análisis , Biomarcadores/metabolismo , Encéfalo/inmunología , Encéfalo/virología , Enfermedades Virales del Sistema Nervioso Central/diagnóstico , Enfermedades Virales del Sistema Nervioso Central/genética , Enfermedades Virales del Sistema Nervioso Central/virología , Factor H de Complemento/metabolismo , Citocinas/metabolismo , Humanos , MicroARNs/análisis , MicroARNs/genética , FN-kappa B/metabolismo , Enfermedades por Prión/diagnóstico , Enfermedades por Prión/genética , Enfermedades por Prión/patología , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptores Toll-Like/metabolismo
16.
Arch Biochem Biophys ; 679: 108193, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31738890

RESUMEN

This work was purposed to speculate the possible association of rs2910164hsa-miR-146a C>G gene single nucleotide polymorphism in the pathogenesis of schizophrenia and subsequently their relevance to neuro-inflammatory, vascular and oxidative stress pathways as acute ischemic stroke (AIS) risk factors in chronic schizophrenic patients. 450 subjects, 150 healthy controls (group I), 150 chronic schizophrenic patients without any evidences of stroke (group II) and 150 chronic schizophrenic patients with AIS (group III) were included. Genotypes (CC, CG&GG) for hsa-mir-146a gene polymorphism were identified using polymerase chain reaction-restriction fragment length polymorphism PCR-RFLP technique. Tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), interleukin 1ß (IL-1 ß), plasminogen activator-inhibitor 1 (PAI-1), thrombomodulin (TM) and 8-Hydroxy-2-deoxyguanosine (8-OHdG) serum levels were immunoassayed. Complete lipid profile was estimated. The CG and GG hsa-miR-146a genotypes were associated with increased risk of both schizophrenia and AIS in schizophrenic patients with thrombomodulin levels decrement in group II& III. On the other side, the risk genotypes were associated significantly with positive and negative syndrome scale PANSS scores, TNF-α, IL-6, IL-1 ß, PAI-1, and 8-OHdG increment levels in both groups II & III. By contrast, the CG and GG hsa-miR-146a genotypes did not affect the neuro-inflammatory and oxidative stress markers in healthy controls. These findings illustrate a new mechanism strengthening the occurrence of oxidative stress and DNA damage as a result of the neuro-inflammatory and endothelial dysfunction status originated from the hsa-miR-146a C>G gene single nucleotide polymorphism, thus, confirming their role in the pathogenesis of schizophrenia and its AIS risk.


Asunto(s)
Isquemia Encefálica/complicaciones , Células Endoteliales/patología , MicroARNs/genética , Estrés Oxidativo/genética , Polimorfismo de Nucleótido Simple , Esquizofrenia/genética , Accidente Cerebrovascular/genética , Enfermedad Aguda , Enfermedad Crónica , Daño del ADN/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Persona de Mediana Edad , Esquizofrenia/metabolismo , Esquizofrenia/patología , Transducción de Señal/genética , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Factor de Necrosis Tumoral alfa/metabolismo
17.
Mol Biol Rep ; 47(10): 7459-7466, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32929657

RESUMEN

Genetic variations of microRNA encoding genes influence various sorts of diseases by modifying the expression or activity of microRNAs. MicroRNA 146a is an epigenetic regulator of immune response through controlling the type I interferon (IFN) and nuclear factor kappa B (NF-κB) pathways. Genetic variations of microRNA 146a impact the susceptibility to systemic lupus erythematosus (SLE) and its clinical presentations. This study aimed to investigate the polymorphisms of microRNA-146a gene (rs2431697 and rs57095329) in patients with SLE and its association with disease activity. Sixty-five patients with SLE and 40 apparently healthy controls were enrolled in this study. Patients were subjected to history taking, clinical examination, and disease activity evaluation by SLEDAI score. The microRNA-146a variants were determined by allele discrimination real-time PCR method in all participants. We found a statistically significant association between rs2431697 T allele and SLE (P-value < 0.05), but there was no significant association between rs57095329 and SLE. The T/T genotype of microRNA-146a rs2431697 was associated with lupus nephritis, higher disease activity, and autoantibodies production. The microRNA-146a rs2431697 T allele could be a potential risk factor that contributes to SLE susceptibility, development of lupus nephritis, and disease activity.


Asunto(s)
Alelos , Predisposición Genética a la Enfermedad , Nefritis Lúpica/genética , MicroARNs/genética , Polimorfismo Genético , Adulto , Femenino , Humanos , Masculino , Factores de Riesgo
18.
Int J Mol Sci ; 21(14)2020 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-32708414

RESUMEN

Exosomes (EXs) and extracellular microvesicles (EMVs) represent a diverse assortment of plasma membrane-derived nanovesicles, 30-1000 nm in diameter, released by all cell lineages of the central nervous system (CNS). They are examples of a very active and dynamic form of extracellular communication and the conveyance of biological information transfer essential to maintain homeostatic neurological functions and contain complex molecular cargoes representative of the cytoplasm of their cells of origin. These molecular cargoes include various mixtures of proteins, lipids, proteolipids, cytokines, chemokines, carbohydrates, microRNAs (miRNA) and messenger RNAs (mRNA) and other components, including end-stage neurotoxic and pathogenic metabolic products, such as amyloid beta (Aß) peptides. Brain microglia, for example, respond to both acute CNS injuries and degenerative diseases with complex reactions via the induction of a pro-inflammatory phenotype, and secrete EXs and EMVs enriched in selective pathogenic microRNAs (miRNAs) such as miRNA-34a, miRNA-125b, miRNA-146a, miRNA-155, and others that are known to promote neuro-inflammation, induce complement activation, disrupt innate-immune signaling and deregulate the expression of neuron-specific phosphoproteins involved in neurotropism and synaptic signaling. This communication will review our current understanding of the trafficking of miRNA-containing EXs and EMVs from astrocytes and "activated pro-inflammatory" microglia to target neurons in neurodegenerative diseases with an emphasis on Alzheimer's disease wherever possible.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Micropartículas Derivadas de Células/metabolismo , Exosomas/metabolismo , Inflamación/metabolismo , MicroARNs/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Enfermedad de Alzheimer/genética , Astrocitos/metabolismo , Transporte Biológico/genética , Exosomas/genética , Vesículas Extracelulares/metabolismo , Humanos , Inflamación/patología , MicroARNs/genética , Neuroglía/metabolismo
19.
Am J Physiol Cell Physiol ; 316(3): C299-C311, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30462539

RESUMEN

Hepatocellular carcinoma (HCC) is known for its high mortality rate worldwide. Based on intensive studies, microRNA (miRNA) expression functions in tumor suppression. Therefore, we aimed to evaluate the contribution of miR-146a-5p to radiosensitivity in HCC through the activation of the DNA damage repair pathway by binding to replication protein A3 (RPA3). First, the limma package of R was performed to differentially analyze HCC expression chip, and regulative miRNA of RPA3 was predicted. Expression of miR-146a-5p, RPA3, and DNA damage repair pathway-related factors in tissues and cells was determined. The effects of radiotherapy on the expression of miR-146a-5p and RPA3 as well as on cell radiosensitivity, proliferation, cell cycle, and apoptosis were also assessed. The results showed that there exists a close correlation between miR-146a and the radiotherapy effect on HCC progression through regulation of RPA3 and the DNA repair pathway. The positive rate of ATM, pCHK2, and Rad51 in HCC tissues was higher when compared with that of the paracancerous tissues. SMMC-7721 and HepG2 cell proliferation were significantly inhibited following 8 Gy 6Mv dose. MiR-146a-5p restrained the expression of RPA3 and promoted the expression of relative genes associated with the DNA repair pathway. In addition, miR-146a-5p overexpression suppresses cell proliferation and enhances radiosensitivity and cell apoptosis in HCC cells. In conclusion, the present study revealed that miR-146a-5p could lead to the restriction of proliferation and the promotion of radiosensitivity and apoptosis in HCC cells through activation of DNA repair pathway and inhibition of RPA3.


Asunto(s)
Carcinoma Hepatocelular/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Neoplasias Hepáticas/genética , MicroARNs/genética , Tolerancia a Radiación/genética , Adulto , Anciano , Apoptosis/genética , Línea Celular , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Células HEK293 , Células Hep G2 , Humanos , Masculino , Persona de Mediana Edad , Transducción de Señal/genética
20.
J Cell Biochem ; 120(8): 13861-13866, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30989711

RESUMEN

Periodontitis is characterized by increased levels of proinflammatory factors, such as interleukin-17 (IL-17) and IL-35. In this study, the expression of microRNA-146a (miRNA-146a), IL-17, and IL-35 in the plasma of patients with periodontitis and healthy controls were detected by quantitative reverse transcription polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. miRNA-146a mimic was transfected into periodontal ligament stem cells (PDLSCs) isolated from periodontitis-affected teeth and healthy teeth. Cell proliferation and expression of IL-17 and IL-35 were detected by cell counting kit-8 assay and Western blot analysis, respectively. It was observed that miRNA-146a was downregulated but IL-17 and IL-35 were upregulated in the plasma of patients with periodontitis than in healthy controls. miRNA-146a was inversely correlated with IL-17 and IL-35 in patients with periodontitis. miRNA-146a overexpression inhibited proliferation of PDLSCs derived from both periodontitis-affected teeth and healthy teeth. miRNA-146a overexpression led to downregulated IL-17 and IL-35 expression in PDLSCs isolated from periodontitis-affected teeth. We, therefore, conclude that miRNA-146a may improve periodontitis by downregulating IL-17 and IL-35 expression and inhibiting proliferation of human PDLSCs.


Asunto(s)
Regulación hacia Abajo/genética , Interleucina-17/genética , Interleucinas/genética , MicroARNs/metabolismo , Ligamento Periodontal/citología , Células Madre/citología , Proliferación Celular , Humanos , Interleucina-17/sangre , Interleucina-17/metabolismo , Interleucinas/sangre , Interleucinas/metabolismo , MicroARNs/sangre , MicroARNs/genética , Periodontitis/sangre , Periodontitis/genética , Diente/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA