Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Small ; 19(35): e2207888, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37127878

RESUMO

Spinal cord injury (SCI), following explosive oxidative stress, causes an abrupt and irreversible pathological deterioration of the central nervous system. Thus, preventing secondary injuries caused by reactive oxygen species (ROS), as well as monitoring and assessing the recovery from SCI are critical for the emergency treatment of SCI. Herein, an emergency treatment strategy is developed for SCI based on the selenium (Se) matrix antioxidant system to effectively inhibit oxidative stress-induced damage and simultaneously real-time evaluate the severity of SCI using a reversible dual-photoacoustic signal (680 and 750 nm). Within the emergency treatment and photoacoustic severity assessment (ETPSA) strategy, the designed Se loaded boron dipyrromethene dye with a double hydroxyl group (Se@BDP-DOH) is simultaneously used as a sensitive reporter group and an excellent antioxidant for effectively eliminating explosive oxidative stress. Se@BDP-DOH is found to promote the recovery of both spinal cord tissue and locomotor function in mice with SCI. Furthermore, ETPSA strategy synergistically enhanced ROS consumption via the caveolin 1 (Cav 1)-related pathways, as confirmed upon treatment with Cav 1 siRNA. Therefore, the ETPSA strategy is a potential tool for improving emergency treatment and photoacoustic assessment of SCI.


Assuntos
Selênio , Traumatismos da Medula Espinal , Ratos , Camundongos , Animais , Antioxidantes/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/diagnóstico por imagem , Traumatismos da Medula Espinal/tratamento farmacológico , Estresse Oxidativo , Tratamento de Emergência
2.
J Cell Mol Med ; 26(6): 1776-1784, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-33219594

RESUMO

Hypoxia/reoxygenation (H/R)-induced myocardial cell injury is the main cause of acute myocardial infarction (AMI). Many proofs show that circular RNA plays an important role in the development of AMI. The purpose of this study was to investigate the role of circSAMD4A in H/R-induced myocardial injury. The levels of circular SAMD4A (circSAMD4A) were detected in the heart tissues of AMI mice and H/R-induced H9C2 cells, and the circSAMD4A was suppressed in AMI mice and H/R-induced H9C2 cells to investigate its' function in AMI. The levels of circSAMD4A and miR-138-5p were detected by real-time quantitative PCR, and MTT assay was used to detect cell viability. TUNEL analysis and Annexin V-FITC were used to determine apoptosis. The expression of Bcl-2 and Bax proteins was detected by Western blot. IL-1ß, TNF-α and IL-6 were detected by ELISA kits. The study found that the levels of circSAMD4A were up-regulated after H/R induction and inhibition of circSAMD4A expression would reduce the H/R-induced apoptosis and inflammation. MiR-138-5p was down-regulated in H/R-induced H9C2 cells. circSAMD4A was a targeted regulator of miR-138-5p. CircSAMD4A inhibited the expression of miR-138-5p to promote H/R-induced myocardial cell injury in vitro and vivo. In conclusion, CircSAMD4A can sponge miR-138-5p to promote H/R-induced apoptosis and inflammatory response.


Assuntos
MicroRNAs , Infarto do Miocárdio , Traumatismo por Reperfusão Miocárdica , RNA Circular/genética , Animais , Apoptose/genética , Hipóxia/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo
3.
J Cell Mol Med ; 25(1): 120-131, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33205602

RESUMO

Osteoarthritis (OA) is an ageing-related disease characterized by articular cartilage degradation and joint inflammation. circRNA has been known to involve in the regulation of multiple inflammatory diseases including OA. However, the mechanism underlying how circRNA regulates OA remains to be elucidated. Here, we report circANKRD36 prevents OA chondrocyte apoptosis and inflammation by targeting miR-599, which specifically degrades Casz1. We performed circRNA sequencing in normal and OA tissues and found the expression of circANKRD36 is decreased in OA tissues. circANKRD36 is also reduced in IL-1ß-treated human chondrocytes. FACS analysis and Western blot showed that the knockdown of circANKRD36 promotes the apoptosis and inflammation of chondrocytes in IL-1ß stress. We then found miR-599 to be the target of circANKRD36 and correlate well with circANKRD36 both in vitro and in vivo. By database analysis and luciferase assay, Casz1 was found to be the direct target of miR-599. Casz1 helps to prevent apoptosis and inflammation of chondrocytes in response to IL-1ß. In conclusion, our results proved circANKRD36 sponge miR-599 to up-regulate the expression of Casz1 and thus prevent apoptosis and inflammation in OA.


Assuntos
Apoptose/genética , Condrócitos/patologia , Proteínas de Ligação a DNA/genética , Inflamação/genética , MicroRNAs/metabolismo , Osteoartrite/genética , RNA Circular/metabolismo , Fatores de Transcrição/genética , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Condrócitos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Humanos , Interleucina-1beta/metabolismo , MicroRNAs/genética , Pessoa de Meia-Idade , RNA Circular/genética , Fatores de Transcrição/metabolismo
4.
J Cell Physiol ; 235(1): 563-572, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31318050

RESUMO

Sepsis is a severe clinical disease, which is resulted from the excessive host inflammation response to the infection. Growing evidence indicates that Staphylococcus aureus pneumonia is a significant cause of sepsis, which can lead to intestinal injury, inflammation, and apoptosis. Studies have shown that miR-182-5p can serve as a tumor oncogene or a tumor suppressive microRNA in various cancers, however, its biological role in sepsis is still uninvestigated. Here, we reported that miR-182-5p was obviously increased in S. aureus pneumonia mice models. Loss of miR-182-5p inhibited intestinal damage and intestinal apoptosis as indicated by the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. In addition, we observed the lack of miR-182-5p altered the local inflammatory response to pneumonia in the intestine. Elevated tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) levels were observed in intestinal tissue of pneumonia groups compared with the shams. Furthermore, miR-182-5p knockout (KO) pneumonia group demonstrated decreased levels of intestinal TNF-α and IL-6. Primary murine intestinal epithelial cells were isolated and cultured in our investigation. We exhibited downregulation of miR-182-5p repressed intestinal epithelial cells apoptosis and rescued the cell viability. Meanwhile, miR-182-5p caused elevated cell apoptosis and reduced cell proliferation. Moreover, the surfactant protein D (SP-D) binds with the bacterial pathogens and remove the pathogens and apoptotic bodies, which exhibits important roles in modulating immune responses. It was displayed in our study that SP-D was greatly decreased in pneumonia mice models. SP-D was predicted as a downstream target of miR-182-5p. These data concluded that miR-182-5p promoted intestinal injury in S. aureus pneumonia-induced sepsis via targeting SP-D.


Assuntos
Mucosa Intestinal/patologia , MicroRNAs/genética , Pneumonia Estafilocócica/patologia , Proteína D Associada a Surfactante Pulmonar/metabolismo , Animais , Apoptose/genética , Sobrevivência Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/patologia , Técnicas de Inativação de Genes , Inflamação/patologia , Interleucina-6/metabolismo , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Oncogenes/genética , Pneumonia Estafilocócica/genética , Sepse/patologia , Transdução de Sinais , Staphylococcus aureus/patogenicidade , Fator de Necrose Tumoral alfa/metabolismo
5.
Mediators Inflamm ; 2020: 8232734, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32410866

RESUMO

Sepsis and intestinal injury triggered by sepsis are common in intensive care units, which can contribute to a high mortality. lncRNAs can modulate gene expression, and they are closely involved in multiple diseases, including sepsis. In our present study, we investigated the biological function of MEG3 in sepsis, especially during the intestinal injury. Currently, we observed that in LPS-induced sepsis mouse models, the intestinal injury was triggered. Meanwhile, we reported that MEG3 was greatly decreased in vivo, with an increase of miR-129-5p and inhibition of SP-D. Then, MEG3 was overexpressed, and we found that its overexpression repressed the intestinal injury via downregulating miR-129-5p in sepsis mice. Moreover, TNF-α and IL-6 expression was elevated in intestinal tissues compared to the control groups. MEG3 restrained the activation of TNF-α and IL-6, in sepsis models. Subsequently, to induce the inflammatory injury of sepsis, human colorectal Caco2 cells were treated with 10 ng/ml LPS. 10 ng/ml LPS significantly inhibited Caco2 cell proliferation and increased the apoptosis. Additionally, MEG3 was decreased whereas miR-129-5p was obviously increased in Caco2 cells incubated with LPS. Interestingly, we showed that MEG3 repressed cell apoptosis partly and enhanced Caco2 cell proliferation. miR-129-5p overexpression could reverse the effect of MEG3 in vitro. Previously, we proved SP-D was reduced in sepsis and it depressed the intestinal injury in vivo. Finally, the correlation among MEG3, miR-129-5p, and SP-D was predicted and confirmed in our investigation. These findings indicated that MEG3 might be a potential target for intestinal damage caused by sepsis via regulating miR-129-5p and SP-D.


Assuntos
MicroRNAs/metabolismo , Proteína D Associada a Surfactante Pulmonar/metabolismo , RNA Longo não Codificante/metabolismo , Sepse/metabolismo , Animais , Apoptose , Células CACO-2 , Proliferação de Células , Células Epiteliais/metabolismo , Citometria de Fluxo , Humanos , Inflamação , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
6.
Mediators Inflamm ; 2020: 8275026, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32508527

RESUMO

Sepsis is a severe disease, which results from the excessive inflammatory response to the infection. Dysfunction of intestinal barrier is a crucial problem in various pathological conditions. Meanwhile, microRNAs exhibit significant roles in the modulation of many diseases, including sepsis. Multiple investigations indicate that miR-199a-5p participates in different human diseases. Nevertheless, little is known on the roles of miR-199a-5p in sepsis. Herein, we evaluated the mechanism of miR-199a-5p on the intestinal barrier dysfunction in sepsis. Intestinal mucosa permeability indicators including D-lactic acid, DAO, and FD-40 levels were determined, and they were greatly increased in sepsis. Then, we proved that miR-199a-5p was induced in sepsis mice tissues and isolated intestinal epithelial cells. Moreover, miR-199a-5p increased D-lactic acid, DAO, and FD-40 while inhibition of miR-199a-5p exhibited a reversed process. Additionally, we observed that miR-199a-5p affected the oxidative damage and inflammation in the intestine tissues from sepsis mice. The content of MDA was elevated whereas SOD was remarkably repressed in the miR-199a-5p mimic group. IL-6, IL-1ß, and TNF-α were induced by miR-199a-5p overexpression while IL-10 was reduced by miR-199a-5p. Subsequently, surfactant protein D (SP-D) was predicted as the target of miR-199a-5p. The activation of NF-κB has been identified in sepsis. Herein, we demonstrated that inhibitor of miR-199a-5p contributed to IEC injury via targeting SP-D and inactivating the NF-κB pathway. These revealed miR-199a-5p exacerbated the intestinal barrier dysfunction via inhibiting SP-D and activating the NF-κB pathway in sepsis.


Assuntos
MicroRNAs/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Proteína D Associada a Surfactante Pulmonar/metabolismo , Sepse/metabolismo , Animais , Apoptose , Regulação da Expressão Gênica , Células HEK293 , Humanos , Inflamação , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mucosa/metabolismo , Estresse Oxidativo , Permeabilidade , Transfecção
7.
J Cell Physiol ; 234(8): 14154-14160, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30633352

RESUMO

Long noncoding RNA UCA1 has exerted a significant effect in cardiovascular disease. The biological role of UCA1 in atherosclerosis is unclear. Our study was to identify the potential mechanisms in the progression of atherosclerosis. Here, we observed that ox-LDL increased UCA1 expression greatly in THP-1 cells. Knockdown of UCA1 greatly inhibited CD36 expression, a crucial biomarker in atherosclerosis. Meanwhile, 20 µg/ml ox-LDL induced foam cell formation, which can be reversed by downregulation of UCA1. In addition, TC and TG levels induced by ox-LDL was rescued by UCA1 small interfering RNA. Accumulating studies have indicated that oxidative stress contributes to atherosclerosis progression. Here, we also found that reactive oxygen species, MDA, and THP-1 cell apoptosis were restrained by decreased of UCA1 with an increase of the superoxide dismutase activity. Moreover, miR-206 was predicted as a target of UCA1 and knockdown of UCA1 was able to repress miR-206 expression. Furthermore, overexpression of miR-206 inhibited oxidative stress process and it was reversed by UCA1 upregulation in vitro. In conclusion, we indicated that UCA1 sponged miR-206 to exacerbate atherosclerosis events induced by ox-LDL in THP-1 cells.


Assuntos
Aterosclerose/genética , Lipoproteínas LDL/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Apoptose/genética , Aterosclerose/patologia , Antígenos CD36/genética , Linhagem Celular , Células Espumosas/metabolismo , Células Espumosas/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Lipoproteínas LDL/farmacologia , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Estresse Oxidativo/genética , RNA Longo não Codificante/antagonistas & inibidores , RNA Interferente Pequeno/genética
8.
J Cell Physiol ; 234(10): 18792-18800, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30927265

RESUMO

Atherosclerosis is still the major cause of morbidity and mortality all over the world. Recently, it has been reported increased levels of tissue iron increase the risk of atherosclerosis. However, the detailed mechanism of iron-induced atherosclerosis progression is barely known. Here, we used apoE-deficient mice models to investigate the effects of low iron diet (<0 mg iron carbonyl/kg), high iron diet (25,000 mg iron carbonyl/kg) on atherosclerosis in vivo. As exhibited, we observed that CD68 was significant enriched by high iron diet in apoE-deficient mice. In addition, transforming growth factor ß, tumor necrosis factor α, interleukin 6 (IL-6), IL-23, IL-10, and IL-1ß levels were also greatly induced by high iron diet. Then, we found that the iron load promoted the inflammation response in macrophages. Moreover, macrophage polarization is a process by which macrophage can express various functional programs in activating macrophages. Here, we observed that iron-load macrophages were polarized toward a proinflammatory macrophage phenotype. The polarization of M1 macrophage was promoted by ferric ammonium citrate (FAC) in bone marrow derived macrophages (BMDMs). Furthermore, ECAR and cellular OCR in BMDM with or without FAC was examined. As shown, BMDM indicated with 50 µM FAC showed a significant increase in basic state and maximal ECAR in contrast to the control group. However, there was no significant difference in OCR. This indicated that the glycolysis was involved in the polarization of M1 macrophage triggered by iron-load. In conclusion, we indicated that the iron load exacerbates the progression of atherosclerosis via inducing inflammation and enhancing glycolysis in macrophages.


Assuntos
Aterosclerose/patologia , Glicólise , Inflamação/patologia , Ferro/efeitos adversos , Macrófagos/metabolismo , Índice de Gravidade de Doença , Animais , Aterosclerose/complicações , Polaridade Celular , Feminino , Compostos Férricos/efeitos adversos , Inflamação/complicações , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Fenótipo
9.
J Cell Physiol ; 234(10): 18773-18780, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30950059

RESUMO

Myocardial ischemia-reperfusion (IR) injury is a common cardiovascular problem, which remains a major cause of death in the world. Emerging evidence has suggested that long noncoding RNAs are crucial players in myocardial injury. However, the functional involvement of nuclear enriched abundant transcript 1 (NEAT1) in myocardial IR injury remains poorly investigated. Our study focused on the mechanism of NEAT1 in myocardial IR injury. Here, we reported a crucial role for NEAT1 in exacerbating cardiac IR injury. NEAT1 was greatly increased in myocardial IR injury mice models. As exhibited knockdown of NEAT1 resulted in attenuated myocardial IR injury in vivo. In addition, we found that NEAT1 was dramatically induced by hypoxia/reoxygenation in H9c2 cells. Lactate dehydrogenase (LDH), malondialdehyde, reactive oxygen species levels, and endoplasmic reticulum stress-regulated cardiomyocyte apoptosis were inhibited by the downregulation of NEAT1. Here, it was shown that knockdown of NEAT1 was able to repress tumor necrosis factor-α, interleukin-1ß, and IL-6 expression. The silence of NEAT1 protected against IR injury via decreasing troponin levels, cardiocytes apoptosis, creatine kinase, and lactate LDH release in vivo. Meanwhile, the mitogen-activated protein kinase (MAPK) signaling was involved in NEAT1-mediated myocardial IR injury. In summary, our data indicated that NEAT1 contributed to myocardial IR injury via activating the MAPK pathway.


Assuntos
Sistema de Sinalização das MAP Quinases , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/genética , RNA Longo não Codificante/metabolismo , Animais , Apoptose/genética , Linhagem Celular , Citocinas/genética , Citocinas/metabolismo , Regulação para Baixo/genética , Técnicas de Silenciamento de Genes , Mediadores da Inflamação/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Camundongos Endogâmicos C57BL , RNA Longo não Codificante/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo
10.
J Cell Physiol ; 233(8): 5847-5855, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29219175

RESUMO

An increasing number of studies have reported that lncRNAs are responsible for the development of neuropathic pain. In our current study, chronic constriction injury (CCI) rat models were established and we observed that lncRNA XIST was greatly increased. Knockdown of XIST can relieve pain characteristics including both mechanical and thermal hyperalgesia in CCI rats. Meanwhile, XIST down-regulation could inhibit neuro-inflammation by reducing expression of inflammatory cytokines including tumor necrosis factor (TNF)-α, IL-1ß, and IL-6 and in CCI rats. By performing bioinformatics technology, miR-544 was predicted to have interactions with XIST and dual-luciferase reporter assays validated the correlation between them. A negative correlation between miR-544 and XIST was observed by carrying out XIST loss or gain of function tests. miR-544 markedly alleviated neuropathic pain development in CCI rats via targeting inflammatory cytokines, which was reversed by XIST over-expression. Moreover, STAT3 was manifested to be a target gene of miR-544 by bioinformatics predictions and it was activated in CCI rats. Over-expression of STAT3 was able to induce neuropathic pain and miR-544 inhibited this process in vivo. Furthermore, XIST increased STAT3 expression by sponging miR-544 in neuropathic pain development. To conclude, our present study indicated that XIST can contribute to neuropathic pain progression in rats through down-regulating miR-544 and up-regulating STAT3. Our results suggested that XIST/miR-544/STAT3 axis can serve as a novel therapeutic target in neuropathic pain development.


Assuntos
MicroRNAs/biossíntese , Neuralgia/genética , RNA Longo não Codificante/metabolismo , Fator de Transcrição STAT3/biossíntese , Animais , Linhagem Celular , Regulação para Baixo , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Inflamação/genética , Interleucina-1beta/biossíntese , Interleucina-6/biossíntese , Modelos Animais , Neuralgia/patologia , RNA Longo não Codificante/genética , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/biossíntese
11.
J Cell Physiol ; 233(10): 6996-7003, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29737518

RESUMO

Atherosclerosis is a kind of chronic inflammation disease with lipid accumulation in in blood vessel linings. Increasing evidence has reported that microRNAs can exert crucial roles in atherosclerosis. In previous study, miR-181a has been implicated to be abnormally expressed in atherosclerosis mice, however its detailed function in atherosclerosis remains uninvestigated. Hence, in our current study, we focused on the biological role of miR-181a in atherosclerosis progression. Ox-LDL has been commonly identified as an important atherosclerosis regulator. We observed that ox-LDL induced THP-1 cell apoptosis dose-dependently and time- dependently. Meanwhile, 25 µg/ml ox-LDL can promote foam cell formation and increased miR-181a expression significantly. CD36 has been involved in atherosclerosis progression and it was found that overexpression of miR-181a inhibited its protein levels. Moreover, miR-181a mimics repressed foam cell formation, TC and TG levels induced by ox-LDL dramatically. In addition, miR-181a mimics were able to reverse THP-1 cell apoptosis, increased IL-6, IL-1ß, and TNF-α protein expression triggered by 25 µg/ml ox-LDL. TLR4 has been linked to various inflammation-associated diseases. In our present study, TLR4 was indicated as miR-181a target and the binding correlation between them was validated by dual-luciferase reporter assay. In conclusion, these results improves the understanding of atherosclerosis modulated by miR-181a/TLR4 and can contribute to development of new approaches for atherosclerosis.


Assuntos
Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , MicroRNAs/genética , Receptor 4 Toll-Like/genética , Apoptose/genética , Aterosclerose/genética , Aterosclerose/metabolismo , Humanos , Inflamação/genética , Inflamação/metabolismo , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
12.
J Cell Biochem ; 119(7): 6154-6161, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29663503

RESUMO

Plenty of microRNAs have been identified as critical mediators in atherosclerosis progression, which is still a great threat to human health. Oxidative stress and inflammation have been implicated to contribute a lot to atherosclerosis development. MiR-135a is abnormally expressed in various cancer types, however its function in atherosclerosis is largely unexplored. Ox-LDL is commonly recognized as a crucial atherosclerosis regulator. In our current study, we observed ox-LDL was able to induce RAW264.7 cell apoptosis and meanwhile miR-135a was restrained by ox-LDL both dose-dependently and time- dependently. CD36 has been reported to participate in atherosclerosis process and miR-135a mimics can inhibit its expression while miR-135a inhibitors exhibited a reverse phenomenon. Meanwhile, miR-135a overexpression can suppress foam cell formation, TC, TG levels, and cell apoptosis induced by 20 µg/mL ox-LDL. Subsequently, it was found that miR-135a overexpression can inhibit oxidative stress by decreasing ROS, MDA levels, and increasing SOD levels. Reversely, miR-135a inhibition demonstrated an inhibitory effect in vitro. Apart from these, miR-135a can also modulate inflammation molecules including IL-6, IL-1ß, and TNF-α. TLR4 was predicted as a target of miR-135a and the negative correlation between them was confirmed by dual-luciferase reporter assay in our study. This work improves our understanding of atherosclerosis events mediated by miR-135a/TLR4 and helps to develop new approaches for atherosclerosis.


Assuntos
Aterosclerose/complicações , Inflamação/prevenção & controle , Macrófagos/patologia , MicroRNAs/genética , Estresse Oxidativo/genética , Receptor 4 Toll-Like/antagonistas & inibidores , Animais , Apoptose , Antígenos CD36/metabolismo , Células Cultivadas , Inflamação/etiologia , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
13.
J Cell Biochem ; 119(7): 6231-6237, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29663491

RESUMO

Atherosclerosis, as a chronic cardiovascular disease, still remains a serious threat to human health. Inflammation and oxidative stress are commonly involved in various stages of atherosclerosis development. MicroRNAs are reported to play important roles in macrophages, which can respond to inflammation and oxidative stress. In our current study, we focused on the biological roles of miR-370 in atherosclerosis. According to the previously research, miR-370 was downregulated in AS mice models. Oxidized low-density lipoprotein (Ox-LDL) is regarded as a crucial regulator of atherosclerosis and we observed that miR-370 was decreased by ox-LDL dose-dependently and time-dependently in THP-1 cells. Then, it was found that miR-370 overexpression was able to inhibit inflammation molecules including IL-6 and IL-1ß. Meanwhile, ROS levels, and malondialdehyde (MDA) were also restrained by miR-370 mimics in vitro. Toll-like receptor 4 (TLR4) has been implicated in many inflammation diseases. It can serve as a target of miR-370 and TLR4 expression was greatly increased in ox-LDL-incubated THP-1 cells in a time and dose dependent manner. The negative correlation was validated using a dual-luciferase reporter assay in our study. In conclusion, our present study revealed that miR-370 can reduce inflammatory reaction and inhibit the ROS production by targeting TLR4 in THP-1 cells.


Assuntos
Aterosclerose/prevenção & controle , Inflamação/prevenção & controle , Lipoproteínas LDL/farmacologia , MicroRNAs/genética , Monócitos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Receptor 4 Toll-Like/antagonistas & inibidores , Aterosclerose/genética , Aterosclerose/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/genética , Inflamação/patologia , Monócitos/metabolismo , Monócitos/patologia , Células THP-1
14.
Cell Physiol Biochem ; 45(2): 523-536, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29393141

RESUMO

BACKGROUND/AIMS: LINC00037 has previously been reported to be up-regulated in clear cell renal cell carcinoma (ccRCC), however, the underlying mechanism remained unknown. In this study, we designed to investigate the functional role of LINC00037 in ccRCC Methods: LINC00037 knockdown and re-expressing 786-O and A498 cells were established. CCK8 assay and EdU assay were performed to evaluate the proliferation rates of ccRCC cells. Flow cytometry assay was performed to detect the cell apoptosis and cell cycle. Subcutaneous injection xenotransplantation mouse model was used to observe the role of LINC00037 in tumor growth in vivo. Mass spectrometry (MS) was performed to find the interacting partner of LINC00037 and RNA immunoprecipitation (RIP) was carried out to validate their interaction. RESULTS: We found that knockdown of LINC00037 resulted in inhibited cell proliferation with activated apoptosis and cell cycle arrest in vitro. Over-expression of LINC00037 in LINC00037 knockdown cells restored and enhanced cell proliferation. In vivo mouse model indicated reduced tumor progression by LINC00037 depletion and promoted tumor progression by LINC00037 overexpression. LINC00037 could bind to epidermal growth factor receptor (EGFR) and increase the protein level of EGFR. CONCLUSION: LINC00037 could inhibit proliferation of ccRCC in an epidermal growth factor receptor-dependent way.


Assuntos
Carcinoma de Células Renais/patologia , Receptores ErbB/metabolismo , Neoplasias Renais/patologia , RNA Longo não Codificante/metabolismo , Animais , Apoptose , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Humanos , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Interferência de RNA , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/genética , RNA Interferente Pequeno/metabolismo , Transplante Heterólogo , Regulação para Cima
15.
Biochem Biophys Res Commun ; 495(1): 526-532, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29101035

RESUMO

Innate immune molecule surfactant protein D (SP-D), a member of the C-type lectin protein family, plays an indispensable role in host defense and the regulation of inflammation in the lung and other tissues. Osteoarthritis (OA) is a degenerative disease of cartilage, with inflammation that causes pathologic changes and tissue damage. However, it is unknown whether there exist SP-D expression and its potential role in the pathogenesis of OA. In this study, we examined SP-D expression and explored its biological function in a sodium nitroprusside (SNP)-stimulated rat chondrocytes and surgically-induced rat OA model. We found SP-D expression in both human and rat articular chondrocytes, with higher level in normal chondrocytes compared to in OA chondrocytes. Furthermore, In vivo study demonstrated that recombinant human SP-D (rhSP-D) ameliorated cartilage degeneration in surgically-induced rat OA model. In vitro cell culture study showed that rhSP-D markedly inhibited the expression of caspase-3 as an apoptosis biomarker, and decreased phosphorylation of p38 mitogen-activated protein kinase (MAPK), which resulted in maintaining normal nuclear morphology and increasing mitochondrial membrane potential in SNP-stimulated rat chondrocytes. Collectively, these findings indicate that SP-D expresses in articular chondrocytes and suppresses SNP-stimulated chondrocyte apoptosis and ameliorates cartilage degeneration via suppressing p38 MAPK activity.


Assuntos
Apoptose/imunologia , Condrócitos/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Óxido Nítrico , Osteoartrite/imunologia , Proteína D Associada a Surfactante Pulmonar/imunologia , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Condrócitos/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Osteoartrite/induzido quimicamente , Ratos , Ratos Sprague-Dawley
16.
J Surg Res ; 222: 39-47, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29273374

RESUMO

BACKGROUND: Reactive oxygen species are increased in multiple gastrointestinal diseases and contribute to their pathogenesis. glutathione (GSH) is an antioxidant that helps to prevent reactive oxygen species-mediated mucosal damage. This study examines the mechanisms by which GSH attenuates hydrogen peroxide (H2O2)-induced injury in intestinal epithelial cells. METHODS: IEC-6 cells were cultured and treated with H2O2 ± GSH. Inflammation was measured by nuclear factor kappa-B (NF-κB) P65 expression, NF-κB nuclear translocation, iκBα phosphorylation, and interleukin 1 beta secretion. Terminal deoxynucleotidyl transferase-mediated UTP end-labeling staining and cleaved caspase-3 were used to assess apoptosis. The role of P38 mitogen-activated protein kinase (P38 MAPK) signaling was examined using the P38 MAPK agonist U46619 and inhibitor SB203580 in H2O2 and GSH-treated cells. Phosphorylated and total P38 MAPKs and cleaved caspase-3 were measured by Western blot. Data are means ± standard deviation, statistical significance P < 0.05 by student's t-test, or one-way analysis of variance. RESULTS: Pretreatment with GSH attenuates the activation of NF-κB and P38 MAPK signaling pathways by H2O2. GSH also decreased H2O2-mediated increases in interleukin 1 beta secretion, cleaved caspase-3 activation, and apoptosis in IEC-6 cells. SB203580 attenuated the increase in apoptosis and cleaved caspase-3 in H2O2-treated cells. The increase in apoptotic index and cleaved caspase-3 observed in U46619-treated cells was also diminished by GSH. CONCLUSIONS: GSH appears to ameliorate oxidative injury in intestinal epithelial cells by attenuating H2O2-mediated activation of NF-κB and P38 MAPK signaling pathways that regulate intestinal inflammation and apoptosis.


Assuntos
Glutationa/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Glutationa/uso terapêutico , Peróxido de Hidrogênio , Interleucina-1beta/metabolismo , Enteropatias/prevenção & controle , Mucosa Intestinal/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/metabolismo , Ratos , Sesquiterpenos
17.
Mol Biol Rep ; 41(6): 3853-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24549718

RESUMO

High mobility group box 1 protein (HMGB1) plays an important role in myocardial ischemia and reperfusion (I/R) injury. Preconditioning of exendin-4 (Ex), a glucagon-like peptide-1 receptor agonist, has been reported to attenuate myocardial I/R injury. The current study investigated whether Ex postconditioning also attenuated myocardial I/R injury and the potential mechanisms. Anesthetized male rats were subjected to ischemia for 30 min and treated with Ex (5 µg/kg, i.v.) 5 min before reperfusion, in the absence and/or presence of exendin (9-39) (an antagonist of glucagon-like peptide-1 receptor, 5 µg/kg, i.v.), followed by reperfusion for 4 h. Lactate dehydrogenase (LDH), creatine kinase (CK), tumor necrosis factor-α, interleukin-6, and infarct size were measured. HMGB1 expression was assessed by immunoblotting. Postconditioning with Ex significantly decreased infarct size and levels of LDH and CK after 4 h reperfusion (all p < 0.05). Ex also significantly inhibited the increase in malondialdehyde level and decreased the level of superoxide dismutase (both p < 0.05). In addition, the increase in HMGB1 expression induced by I/R was significantly attenuated by Ex postconditioning. Administration of exendin (9-39) abolished the protective effect of Ex postconditioning (all p < 0.05). The present study suggests that Ex postconditioning may attenuate myocardial I/R injury, which may in turn be associated with inhibiting inflammation.


Assuntos
Inflamação/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Peptídeos/administração & dosagem , Receptores de Glucagon/agonistas , Peçonhas/administração & dosagem , Animais , Creatina Quinase/biossíntese , Exenatida , Regulação da Expressão Gênica/efeitos dos fármacos , Receptor do Peptídeo Semelhante ao Glucagon 1 , Proteína HMGB1/biossíntese , Proteína HMGB1/metabolismo , Humanos , Inflamação/genética , Inflamação/patologia , Interleucina-6/biossíntese , L-Lactato Desidrogenase/biossíntese , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Fragmentos de Peptídeos/administração & dosagem , Peptídeos/metabolismo , Ratos , Receptores de Glucagon/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Peçonhas/metabolismo
18.
Sci China Life Sci ; 67(5): 913-939, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38332216

RESUMO

Fulminant myocarditis is an acute diffuse inflammatory disease of myocardium. It is characterized by acute onset, rapid progress and high risk of death. Its pathogenesis involves excessive immune activation of the innate immune system and formation of inflammatory storm. According to China's practical experience, the adoption of the "life support-based comprehensive treatment regimen" (with mechanical circulation support and immunomodulation therapy as the core) can significantly improve the survival rate and long-term prognosis. Special emphasis is placed on very early identification,very early diagnosis,very early prediction and very early treatment.


Assuntos
Miocardite , Miocardite/diagnóstico , Miocardite/terapia , Humanos , China , Adulto , Cardiologia/métodos , Cardiologia/normas , Prognóstico , Sociedades Médicas
19.
Redox Biol ; 56: 102432, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35981417

RESUMO

OBJECTIVE: Inflammation and oxidative stress contribute to the progression of sepsis-induced acute lung injury (ALI). SAM domain, SH3 domain and nuclear localization signals 1 (SAMSN1) is a signaling adaptor protein, and mainly regulates inflammatory response of various immune cells. The present study generates macrophage-specific SAMSN1-knockout (Samsn1MKO) and SAMSN1-transgenic (Samsn1MTG) mice to investigate its role and mechanism in sepsis-induced ALI. METHODS: Samsn1MKO and Samsn1MTG mice were exposed to lipopolysaccharide (LPS) instillation or cecal ligation and puncture (CLP) surgery to induce sepsis-induced ALI. Bone marrow transplantation, cellular depletion and non-invasive adoptive transfer of bone marrow-derived macrophages (BMDMs) were performed to validate the role of macrophage SAMSN1 in sepsis-induced ALI in vivo. Meanwhile, BMDMs were isolated from Samsn1MKO or Samsn1MTG mice to further clarify the role of SAMSN1 in vitro. RESULTS: Macrophage SAMSN1 expression was increased in response to LPS stimulation, and negatively correlated with LPS-induced ALI in mice. Macrophage SAMSN1 deficiency exacerbated, while macrophage SAMSN1 overexpression ameliorated LPS-induced inflammation, oxidative stress and ALI in mice and in BMDMs. Mechanistically, we found that macrophage SAMSN1 overexpression prevented LPS-induced ALI though activating AMP-activated protein kinase α2 (AMPKα2) in vivo and in vitro. Further studies revealed that SAMSN1 directly bound to growth factor receptor bound protein 2-associated protein 1 (GAB1) to prevent its protein degradation, and subsequently enhanced protein kinase A (PKA)/AMPKα2 activation in a protein tyrosine phosphatase, non-receptor type 11 (PTPN11, also known as SHP2)-dependent manner. Moreover, we observed that macrophage SAMSN1 overexpression diminished CLP-induced ALI in mice. CONCLUSION: Our study documents the protective role of macrophage SAMSN1 against sepsis-induced inflammation, oxidative stress and ALI through activating AMPKα2 in a GAB1/SHP2/PKA pathway, and defines it as a promising biomarker and therapeutic target to treat sepsis-induced ALI.


Assuntos
Lesão Pulmonar Aguda , Proteínas Adaptadoras de Transporte Vesicular , Sinais de Localização Nuclear , Sepse , Proteínas Quinases Ativadas por AMP/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteína Adaptadora GRB2/metabolismo , Inflamação/metabolismo , Lipopolissacarídeos/efeitos adversos , Pulmão/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Sinais de Localização Nuclear/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Sepse/complicações , Sepse/metabolismo
20.
Front Med (Lausanne) ; 8: 665464, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34055839

RESUMO

Sepsis is one of the main causes of death in critically ill patients. Despite the continuous development of medical technology in recent years, its morbidity and mortality are still high. This is mainly related to the delay in starting treatment and non-adherence of clinical guidelines. Artificial intelligence (AI) is an evolving field in medicine, which has been used to develop a variety of innovative Clinical Decision Support Systems. It has shown great potential in predicting the clinical condition of patients and assisting in clinical decision-making. AI-derived algorithms can be applied to multiple stages of sepsis, such as early prediction, prognosis assessment, mortality prediction, and optimal management. This review describes the latest literature on AI for clinical decision support in sepsis, and outlines the application of AI in the prediction, diagnosis, subphenotyping, prognosis assessment, and clinical management of sepsis. In addition, we discussed the challenges of implementing and accepting this non-traditional methodology for clinical purposes.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa