Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cancer Nurs ; 2024 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-38335462

RESUMO

BACKGROUND: Identifying the definition of "being a good parent" facilitates the understanding of parents' personal beliefs and deeds regarding their ill child. OBJECTIVE: The aim of this study was to explore the concept of "being a good parent to my ill child" during pediatric cancer treatment from the perspective of Chinese children, parents, and providers. METHODS: A descriptive qualitative study was conducted with 6 children, 18 parents, 5 doctors, 19 nurses, and 3 social workers by semistructured interviews at 3 Chinese hospitals. RESULTS: Except for "letting the Lord lead," 7 themes from the original conceptual model were validated, for example, "being there for my child" (n = 51, 100.0%); "doing right by my child" (n = 38, 74.5%), "being an advocate for my child" (n = 27, 52.9%), "conveying love to my child" (n = 26, 51.0%), "making my child healthy" (n = 18, 35.3%), "being a good life example" (n = 13, 25.5%), and "not allowing suffering" (n = 13, 25.5%). A new theme, "rebuilding myself" (n = 39, 76.5%), emerged in the Chinese context. "Being a good parent to my ill child" is perceived differently among stakeholders. Healthcare professionals' facilitation to fulfill the concept included "recognizing the individualized good-parent definition," "providing best available care" and "establishing a supportive environment." CONCLUSION: "Being a good parent to my ill child" is meaningfully expressed by Chinese parents and recognized by children and providers during pediatric cancer treatment. IMPLICATIONS FOR PRACTICE: It is important to support parents in conveying their internal good parent definition and sharing it with stakeholders. Attention should be paid to related cultural influencers, a supportive family-friendly environment, and shared decision making involving the child's voice.

2.
Int J Nurs Sci ; 9(4): 430-437, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36285075

RESUMO

Objective: Unmet supportive care needs(SCNs) impact pediatric cancer patients and their parents. This study aimed to explore the unmet SCNs from the perspective of Chinese children with cancer and their parents through lived experiences. Methods: The data of this study was collected using face-to-face semi-structured interviews. The participants were recruited from the oncology units of three children's hospitals in China's cities (Shanghai, Guangzhou, and Hefei) from October 2020 to December 2021. Data were analyzed using Colaizzi's seven-step phenomenological analysis method. Results: Eight pediatric cancer patients and twenty-four parents were enrolled in the study. Four main themes and eight subthemes (both children's and parent's perspectives) were generated: 1) meeting the ongoing needs along the cancer trajectory (can you tell me what comes next; our needs are growing); 2) communicating with a family focus (they only talk to my parents; let each family member have a voice); 3) providing care beyond the treatment (I am bigger than my body [the children's needs for emotional consolidation and information about their prognosis]; there are things beyond treatment); 4) getting support from the community (I am not a monster [the children were unhappy about being treated differently]; we want to connect with the resources near us). Conclusion: This study revealed multiple unmet SCNs from the perspective of Chinese children with cancer and their parents. The findings call for comprehensive and in-depth supportive care beyond treatment, integration of the family member voice in pediatric cancer care, and a coordinated pediatric cancer support mechanism in the Chinese healthcare system.

3.
Cancer Nurs ; 45(1): E68-E82, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34870939

RESUMO

BACKGROUND: Evidence suggests that digital health interventions are feasible and acceptable in pediatric cancer survivors. Efforts to synthesize user experiences, however, are limited. OBJECTIVE: The aim of this study was to systematically identify, appraise, and synthesize qualitative evidence on the user experiences of pediatric cancer survivors participating in digital health interventions. METHODS: We searched 4 databases to identify studies with qualitative data of the digital health intervention user experiences from childhood, adolescent, and young adult pediatric cancer survivors, published from the databases' inception to January 31, 2020. Studies that met inclusion criteria were selected and reviewed for quality. The extracted data were analyzed by thematic synthesis. RESULTS: Sixteen articles met the inclusion criteria. Two analytic themes emerged, each with 3 descriptive themes: (1) the favorable experience of using digital health interventions, including "knowing about oneself and one's cancer," "connecting with others in a trustworthy and safe way," and "getting empowered and rewarded"; and (2) the challenges of using digital health interventions, including "challenges from the content of the program," "technology challenges," and "user challenges." CONCLUSIONS: This systematic review supports the value of digital health interventions as an acceptable and convenient approach for delivering care to pediatric cancer survivors. It is essential to understand the users' positive experiences and challenges to customize interventions and achieve positive health outcomes. IMPLICATIONS FOR PRACTICE: Nurses can contribute to the development and implementation of digital health interventions for pediatric cancer survivors through advocacy, navigation, feedback, and multidisciplinary collaboration. We recommended system-level support in modifying and developing relevant workflows or practice guidelines for implementation.


Assuntos
Sobreviventes de Câncer , Neoplasias , Adolescente , Criança , Humanos , Neoplasias/terapia , Pesquisa Qualitativa , Sobreviventes , Adulto Jovem
4.
J Pediatr Oncol Nurs ; 38(4): 262-270, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33686896

RESUMO

Background: Fatigue is a prevalent and distressing symptom in children and adolescents with cancer. Objectives: This study aimed to (1) investigate the current fatigue status reported by Chinese children and adolescents with cancer during active cancer treatment and (2) examine whether sociodemographic information, disease and treatment information, co-occurring symptoms, function and related clinical data are significantly associated with fatigue according to the biopsychosocial model. Methods: Participants were children aged 8-17 years, who had undergone treatment for cancer at four hospitals in China. Children completed the Chinese version of the Pediatric Patient-Reported Outcomes Measurement Information System short forms. Results: In total, 187 children (33.16% female, mean age 10.28 years) participated. The mean T-score for child-reported fatigue was 48.52 (34-72). Multiple linear regression analysis showed that fatigue in pediatric active cancer treatment could be significantly predicted by greater child-reported pain interference (ß = 0.391, p < .001), greater depressive symptoms (ß = 0.443, p < .001), and reduced mobility (ß = -0.226, p = .004) (adjusted R2 = 0.613, F = 16.476, p < .001). Conclusions: Children and adolescents with cancer experience multiple, intersecting troubling symptoms during their treatment. There is a need to attend to the biopsychosocial aspects of care for children and adolescents during active cancer treatment. To reduce pediatric oncology patients' fatigue level, clinicians could develop culturally sensitive interventions to alleviate children's pain interference, treat depressive symptoms, and maximize their physical mobility.


Assuntos
Fadiga , Neoplasias , Adolescente , Criança , China , Fadiga/epidemiologia , Fadiga/etiologia , Feminino , Humanos , Masculino , Neoplasias/complicações , Neoplasias/terapia , Qualidade de Vida , Autorrelato
5.
Int J Nurs Sci ; 8(1): 22-29, 2021 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-33575441

RESUMO

OBJECTIVE: Pediatric cancer patients endure multiple symptoms during treatment and also in survivorship. Digital health technologies provide an innovative way to support their symptom management. This review aimed to examine the effect of digital health technologies on managing symptoms among across pediatric cancer continuum. METHODS: A systematic literature search of six English and three Chinese electronic databases was combined with hand searching, to identify eligible research studies from database establishment to November 30, 2019. Two reviewers carried out data selection, data extraction, and quality appraisal independently. A narrative approach was taken to summarize data. RESULTS: Four randomized control trials, two quasi-experiments, and five one group pre-posttest designed studies, were included in the review with a total of 425 participants. The methodological quality of the studies was generally fair. Seven symptoms (anxiety, depression, pain, anger, fatigue, fear, distress) and seven digital health technologies (visual reality, website, humanoid robot, app, wearable devices, short messages and videoconference) were reported in the included studies. CONCLUSIONS: Current evidence supports the effect of digital health technologies is generally mixed and inconclusive. There is a trend of positive effects found in the interventions that feature digital health technologies' interactive function. This review highlights the need for further investigation with rigorous research designs and the consideration of influencing factors from the symptoms, participants, and context levels to inform a better digital health implementation.

6.
Free Radic Biol Med ; 160: 303-318, 2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-32846217

RESUMO

Ferroptosis is a reactive oxygen species (ROS)- and iron-dependent form of regulated cell death (RCD), playing critical roles in organ injury and targeting therapy of cancers. Previous studies have demonstrated that ferroptosis participates in the development of cardiomyopathy including cardiac hypertrophy, diabetic cardiomyopathy and doxorubicin-induced cardiotoxicity. However, the role of ferroptosis in sepsis-induced cardiac injury remains unclear. This study aimed to explore the role and underlying mechanism of ferroptosis on lipopolysaccharide (LPS)-induced cardiac injury. Mice were injected with LPS (10 mg/kg) for 12 h to generate experimental sepsis. Ferrostatin-1 (Fer-1) and Dexrazoxane (DXZ) were used to suppress ferroptosis of mice with sepsis-induced cardiac injury. LPS increased the levels of ferroptotic markers involving prostaglandin endoperoxide synthase 2 (PTGS2), malonaldehyde (MDA) and lipid ROS, apart from resulting in obvious mitochondria damage, which were alleviated by Fer-1 and DXZ. In vitro experiments showed that Fer-1 inhibited LPS-induced lipid peroxidation and injury of H9c2 myofibroblasts while erastin and sorafenib aggravated LPS-induced ferroptosis. Additionally, Fer-1 and DXZ improved survival rate and cardiac function of mice with sepsis. Mechanistically, LPS increased the expression of nuclear receptor coactivator 4 (NCOA4) and the level of intracellular Fe2+ but decreased the level of ferritin. NCOA4 could directly interact with ferritin and degrade it in a ferritinophagy-dependent manner, which subsequently released a great amount of iron. Cytoplasmic Fe2+ further activated the expression of siderofexin (SFXN1) on mitochondrial membrane, which in turn transported cytoplasmic Fe2+ into mitochondria, giving rise to the production of mitochondrial ROS and ferroptosis. Based on these findings, we concluded that ferritinophagy-mediated ferroptosis is one of the critical mechanisms contributing to sepsis-induced cardiac injury. Targeting ferroptosis in cardiomyocytes may be a therapeutic strategy for preventing sepsis in the future.


Assuntos
Ferroptose , Sepse , Animais , Autofagia , Ferritinas , Ferro/metabolismo , Camundongos
7.
Cell Death Dis ; 11(3): 160, 2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-32123163

RESUMO

High-mobility group AT-hook1 (HMGA1, formerly HMG-I/Y), an architectural transcription factor, participates in a number of biological processes. However, its effect on cardiac remodeling (refer to cardiac inflammation, apoptosis and dysfunction) in diabetic cardiomyopathy remains largely indistinct. In this study, we found that HMGA1 was upregulated in diabetic mouse hearts and high-glucose-stimulated cardiomyocytes. Overexpression of HMGA1 accelerated high-glucose-induced cardiomyocyte inflammation and apoptosis, while HMGA1 knockdown relieved inflammation and apoptosis in cardiomyocytes in response to high glucose. Overexpression of HMGA1 in mice heart by adeno-associated virus 9 (AAV9) delivery system deteriorated the inflammatory response, increased apoptosis and accelerated cardiac dysfunction in streptozotocin-induced diabetic mouse model. Knockdown of HMGA1 by AAV9-shHMGA1 in vivo ameliorated cardiac remodeling in diabetic mice. Mechanistically, we found that HMGA1 inhibited the formation rather than the degradation of autophagy by regulating P27/CDK2/mTOR signaling. CDK2 knockdown or P27 overexpression blurred HMGA1 overexpression-induced deteriorating effects in vitro. P27 overexpression in mice heart counteracted HMGA1 overexpression-induced increased cardiac remodeling in diabetic mice. The luciferase reporter experiment confirmed that the regulatory effect of HMGA1 on P27 was mediated by miR-222. In addition, a miR-222 antagomir counteracted HMGA1 overexpression-induced deteriorating effects in vitro. Taken together, our data indicate that HMGA1 aggravates diabetic cardiomyopathy by directly regulating miR-222 promoter activity, which inhibits P27/mTOR-induced autophagy.


Assuntos
Autofagia/fisiologia , Diabetes Mellitus Experimental/metabolismo , Cardiomiopatias Diabéticas/metabolismo , Proteína HMGA1a/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Diabetes Mellitus Experimental/complicações , Cardiomiopatias Diabéticas/complicações , Proteína HMGA1a/genética , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Estreptozocina
8.
Int J Biol Sci ; 16(1): 12-26, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31892842

RESUMO

Adverse cardiac remodeling after myocardial infarction (MI) is associated with extremely high mortality rates worldwide. Although optimized medical therapy, Preservation of lusitropic and inotropic function and protection against adverse remodeling in ventricular structure remain relatively frequent. This study demonstrated that Andrographolide (Andr) significantly ameliorated adverse cardiac remodeling induced by myocardial infarction and improves contractile function in mice with LAD ligation compared with the control group. Briefly, Andr markedly attenuated cardiac fibrosis and relieved inflammation after myocardial infarction. Specifically, Andr significantly blocked oxidative stress and the nuclear translocation of p-P65 following myocardial infarction. At the mechanistic level, antioxidant effect of Andr was achieved through strengthening antioxidative stress capacity and attributed to the activation of Nrf2/HO-1 Signaling. Consistently, H9C2 administrated with Andr showed a decreased oxidative stress caused by hypoxia precondition, but treatment with specific Nrf2 inhibitor (ML385) or the silence of Nrf2 blunted the activation of Nrf2/HO-1 Signaling and removed the protective effects of Andr in vitro. Thus, we suggest that Andr alleviates adverse cardiac remodeling following myocardial infarction through enhancing Nrf2 signaling pathway.


Assuntos
Diterpenos/uso terapêutico , Infarto do Miocárdio/tratamento farmacológico , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Western Blotting , Ecocardiografia , Hemodinâmica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos
9.
Mediators Inflamm ; 2019: 6168340, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31686985

RESUMO

Andrographolide (Andr) is a major component isolated from the plant Andrographis paniculata. Inflammation, apoptosis, and impaired angiogenesis are implicated in the pathogenesis of high glucose (HG)-induced injury of vascular endotheliocytes. Our study is aimed at evaluating the effect of Andr on HG-induced HUVEC injury and the underlying mechanism. HUVECs were exposed to HG levels (33 mM) and treated with Andr (0, 12.5, 25, and 50 µM). Western blot analysis, real-time PCR, immunofluorescence staining, the scratch test, and the tube formation assay were performed to assess the effects of Andr. We discovered that Andr inhibited the inflammatory response (IL-1ß, IL-6, and TNFα), decreased the apoptosis ratio and cell migration, and promoted tube formation in response to HG stimulation. Andr ameliorated the levels of phosphorylated PI3K (p-PI3K), phosphorylated AKT (p-AKT), and phosphorylated eNOS (p-eNOS). The expression of vascular endothelial growth factor (VEGF) protein, a vital factor in angiogenesis, was improved by Andr treatment under HG stimulation. LY294002 is a blocker of PI3K, MK-2206 2HCI (MK-2206) is a highly selective AKT inhibitor, and L-NAME is a suppressor of eNOS, all of which significantly reduce Andr-mediated protective effects in vitro. Hence, Andr may be involved in regulating HG-induced injury by activating PI3K/AKT-eNOS signalling in HUVECs.


Assuntos
Diterpenos/farmacologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Inflamação/prevenção & controle , Apoptose/efeitos dos fármacos , Western Blotting , Movimento Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Marcação In Situ das Extremidades Cortadas , Inflamação/induzido quimicamente , Inflamação/metabolismo , L-Lactato Desidrogenase/metabolismo , Malondialdeído/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
10.
Oxid Med Cell Longev ; 2019: 7536803, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31781348

RESUMO

Zileuton has been demonstrated to be an anti-inflammatory agent due to its well-known ability to inhibit 5-lipoxygenase (5-LOX). However, the effects of zileuton on cardiac remodeling are unclear. In this study, the effects of zileuton on pressure overload-induced cardiac remodeling were investigated and the possible mechanisms were examined. Aortic banding was performed on mice to induce a cardiac remodeling model, and the mice were then treated with zileuton 1 week after surgery. We also stimulated neonatal rat cardiomyocytes with phenylephrine (PE) and then treated them with zileuton. Our data indicated that zileuton protected mice from pressure overload-induced cardiac hypertrophy, fibrosis, and oxidative stress. Zileuton also attenuated PE-induced cardiomyocyte hypertrophy in a time- and dose-dependent manner. Mechanistically, we found that zileuton activated PPARα, but not PPARγ or PPARθ, thus inducing Keap and NRF2 activation. This was confirmed with the PPARα inhibitor GW7647 and NRF2 siRNA, which abolished the protective effects of zileuton on cardiomyocytes. Moreover, PPARα knockdown abolished the anticardiac remodeling effects of zileuton in vivo. Taken together, our data indicate that zileuton protects against pressure overload-induced cardiac remodeling by activating PPARα/NRF2 signaling.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Hidroxiureia/análogos & derivados , Inibidores de Lipoxigenase/farmacologia , PPAR alfa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hidroxiureia/farmacologia , Masculino , Camundongos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Ratos
11.
J Pharmacol Exp Ther ; 371(2): 348-359, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31467086

RESUMO

Aucubin (AUB), which is extracted from Eucommia ulmoides Oliver seeds, has been found to possess anti-inflammatory and antiapoptotic properties. Recent studies have indicated that inflammation, oxidative stress, and apoptosis are involved in the pathophysiology of lipopolysaccharide (LPS)-induced cardiac dysfunction. Our study aimed to investigate the effect of AUB on LPS-induced acute cardiac injury. Male C57BL/6 mice were injected with LPS (one 6 mg/kg injection) to induce cardiac dysfunction without or with AUB pretreatment (20 or 80 mg/kg per day) for 1 week. We found that AUB ameliorated cardiac dysfunction, inflammation, oxidative stress, and apoptosis induced by LPS stimulation. Mechanistically, AUB inhibited LPS-induced oxidative stress by decreasing reactive oxygen species and thioredoxin interaction protein (TXNIP) levels. Moreover, AUB suppressed LPS-induced inflammation and apoptosis by reducing nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3)/apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)/caspase-1 inflammasome formation. Overexpression of NLRP3 in cardiomyocytes attenuated the protective effects of AUB. Interestingly, NLRP3 deficiency ameliorated cardiac function and reduced the inflammatory response and oxidative stress after LPS insult in mice, whereas AUB could not further prevent LPS-induced cardiac dysfunction in NLRP3-deficient mice. In summary, AUB exerts a protective effect against LPS-induced inflammation, oxidative stress, and apoptosis in vivo and in vitro by regulating the TXNIP pathway and inactivating the NLRP3/ASC/caspase-1 inflammasome. Hence, AUB may be a promising agent against LPS-induced cardiac dysfunction. SIGNIFICANCE STATEMENT: Aucubin exerts a protective effect against lipopolysaccharide-induced cardiac dysfunction by regulating nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 inflammasome.


Assuntos
Cardiotônicos/uso terapêutico , Frutas , Cardiopatias/metabolismo , Cardiopatias/prevenção & controle , Glucosídeos Iridoides/uso terapêutico , Lipopolissacarídeos/toxicidade , Animais , Cardiotônicos/farmacologia , Células Cultivadas , Cardiopatias/induzido quimicamente , Glucosídeos Iridoides/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/deficiência , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Distribuição Aleatória , Ratos
12.
J Cell Mol Med ; 23(9): 6466-6478, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31293067

RESUMO

Cardiac remodelling refers to a series of changes in the size, shape, wall thickness and tissue structure of the ventricle because of myocardial injury or increased pressure load. Studies have shown that cardiac remodelling plays a significant role in the development of heart failure. Zingerone, a monomer component extracted from ginger, has been proven to possess various properties including antioxidant, anti-inflammatory, anticancer and antidiabetic properties. As oxidative stress and inflammation contribute to acute and chronic myocardial injury, we explored the role of zingerone in cardiac remodelling. Mice were subjected to aortic banding (AB) or sham surgery and then received intragastric administration of zingerone or saline for 25 days. In vitro, neonatal rat cardiomyocytes (NRCMs) were treated with zingerone (50 and 250 µmol/L) when challenged with phenylephrine (PE). We observed that zingerone effectively suppressed cardiac hypertrophy, fibrosis, oxidative stress and inflammation. Mechanistically, Zingerone enhanced the nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/antioxidant response element (ARE) activation via increasing the phosphorylation of endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) production. Additionally, we used Nrf2-knockout (KO) and eNOS-KO mice and found that Nrf2 or eNOS deficiency counteracts these cardioprotective effects of zingerone in vivo. Together, we concluded that zingerone may be a potent treatment for cardiac remodelling that suppresses oxidative stress via the eNOS/Nrf2 pathway.


Assuntos
Aorta/efeitos dos fármacos , Guaiacol/análogos & derivados , Fator 2 Relacionado a NF-E2/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Transdução de Sinais/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Aorta/metabolismo , Cardiomegalia/tratamento farmacológico , Cardiomegalia/metabolismo , Células Cultivadas , Fibrose/tratamento farmacológico , Fibrose/metabolismo , Guaiacol/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fenilefrina/farmacologia , Ratos
13.
J Cell Mol Med ; 23(8): 5715-5727, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31225721

RESUMO

Increase of myocardial oxidative stress is closely related to the occurrence and development of cardiac hypertrophy. Cordycepin, also known as 3'-deoxyadenosine, is a natural bioactive substance extracted from Cordyceps militaris (which is widely cultivated for commercial use in functional foods and medicine). Since cordycepin suppresses oxidative stress both in vitro and in vivo, we hypothesized that cordycepin would inhibit cardiac hypertrophy by blocking oxidative stress-dependent related signalling. In our study, a mouse model of cardiac hypertrophy was induced by aortic banding (AB) surgery. Mice were intraperitoneally injected with cordycepin (20 mg/kg/d) or the same volume of vehicle 3 days after-surgery for 4 weeks. Our data demonstrated that cordycepin prevented cardiac hypertrophy induced by AB, as assessed by haemodynamic parameters analysis and echocardiographic, histological and molecular analyses. Oxidative stress was estimated by detecting superoxide generation, superoxide dismutase (SOD) activity and malondialdehyde levels, and by detecting the protein levels of gp91phox and SOD. Mechanistically, we found that cordycepin activated activated protein kinase α (AMPKα) signalling and attenuated oxidative stress both in vivo in cordycepin-treated mice and in vitro in cordycepin treated cardiomyocytes. Taken together, the results suggest that cordycepin protects against post-AB cardiac hypertrophy through activation of the AMPKα pathway, which subsequently attenuates oxidative stress.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Cardiomegalia/tratamento farmacológico , Desoxiadenosinas/uso terapêutico , Transdução de Sinais , Angiotensina II/farmacologia , Animais , Cardiomegalia/diagnóstico por imagem , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Cardiotônicos/farmacologia , Cardiotônicos/uso terapêutico , Desoxiadenosinas/farmacologia , Fibrose , Hemodinâmica/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Pressão , Transdução de Sinais/efeitos dos fármacos
14.
Redox Biol ; 24: 101215, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31121492

RESUMO

Mountainous evidence suggests that inflammation, cardiomyocyte apoptosis and pyroptosis are involved in the development of sepsis and sepsis-induced cardiomyopathy (SIC). Stimulator of interferon genes (STING) is an indispensable molecule that could regulate inflammation and immune response in multiple diseases. However, the role of STING in cardiovascular disease, especially SIC remains unclear. This study was designed to investigate the potential molecular mechanisms of STING in lipopolysaccharide (LPS)-induced cardiac injury using STING global knockout mice. In wild type mice and cardiomyocytes, LPS stimulation triggered the perinuclear translocation of STING, which further bound to Type-I interferons (IFN) regulatory factor 3 (IRF3) and phosphorylated IRF3. Phosphorylated (P-) IRF3 subsequently translocated into nucleus and increased the expression of NOD-like receptor protein 3 (NLRP3). Knockout of STING in mice significantly improved survival rate and cardiac function, apart from suppressing myocardial and serum inflammatory cytokines, apoptosis, as well as cardiomyocyte pyroptosis. In vitro experiments revealed that NLRP3 overexpression by adenovirus could offset protective effects of STING knockdown in LPS-induced cardiomyocytes. Additionally, LPS stimulation also promoted the production of intracellular reactive oxygen (ROS), which further induced the NLRP3 translocation to the cytoplasm from the nucleus. Dissociative TXNIP could directly interact with cytoplasmic NLRP3 and form inflammasome, eventually triggering cardiomyocyte injury. Collectively, our findings disclose that STING deficiency could alleviate LPS-induced SIC in mice. Hence, targeting STING in cardiomyocytes may be a promising therapeutic strategy for preventing SIC.


Assuntos
Apoptose , Cardiopatias/etiologia , Cardiopatias/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Lipopolissacarídeos/efeitos adversos , Proteínas de Membrana/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Citocinas/sangue , Citocinas/metabolismo , Modelos Animais de Doenças , Ecocardiografia , Cardiopatias/diagnóstico , Cardiopatias/fisiopatologia , Fator Regulador 3 de Interferon/genética , Espaço Intracelular , Masculino , Proteínas de Membrana/genética , Camundongos , Miócitos Cardíacos/metabolismo , Piroptose , Espécies Reativas de Oxigênio
15.
J Mol Cell Cardiol ; 128: 160-178, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30711544

RESUMO

High mobility group protein AT-hook 2 (HMGA2), an architectural transcription factor, has previously been reported to play an essential role in regulating the expression of many genes through architectural remodeling processes. However, the effects of HMGA2 on cardiovascular disease, especial cardiac remodeling, is unclear. This study was aimed at investigating the functional role of HMGA2 in pressure overload-induced cardiac remodeling. Mice that were subjected to aortic banding (AB) for 8 weeks developed myocardial hypertrophy and cardiac dysfunction, which were associated with altered expression of HMGA2. Cardiac-specific expression of the human HMGA2 gene in mice with an adeno-related virus 9 delivery system ameliorated cardiac remodeling and improve cardiac function in response to pressure overload by activating PPARγ/NRF2 signaling. Knockdown of HMGA2 by AAV9-shHMGA2 accelerated cardiac remodeling after 1 weeks of AB surgery. Additionally, knockdown of heart PPARγ largely abolished HMGA2 overexpression-mediated cardioprotection. HMGA2-mediated cardiomyocyte protection was largely abrogated by knocking down NRF2 and inhibiting PPARγ in cardiomyocytes. PPARγ activation was mediated by C/EBPß, which directly interacted with HMGA2. Knocking down C/EBPß offset the effects of HMGA2 on PPARγ activation and cardioprotection. These findings show that the overexpression of HMGA2 ameliorates the remodeling response to pressure overload, and they also imply that the upregulation of HMGA2 may become a treatment strategy in cardiac pathologies.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/genética , Doenças Cardiovasculares/genética , Proteína HMGA2/genética , PPAR gama/genética , Aorta/metabolismo , Aorta/patologia , Remodelamento Atrial/genética , Cardiomegalia/genética , Cardiomegalia/patologia , Doenças Cardiovasculares/patologia , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fator 2 Relacionado a NF-E2/genética , Pressão/efeitos adversos
16.
Br J Pharmacol ; 175(9): 1548-1566, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29447430

RESUMO

BACKGROUND AND PURPOSE: Aucubin, the predominant component of Eucommia ulmoides Oliv., has been shown to have profound effects on oxidative stress. As oxidative stress has previously been demonstrated to contribute to acute and chronic myocardial injury, we tested the effects of aucubin on cardiac remodelling and heart failure. EXPERIMENTAL APPROACH: Initially, H9c2 cardiomyocytes and neonatal rat cardiomyocytes pretreated with aucubin (1, 3, 10, 25 and 50 µM) were challenged with phenylephrine. Secondly, the transverse aorta was constricted in C57/B6 and neuronal NOS (nNOS)-knockout mice, then aucubin (1 or 5 mg·kg-1 body weight day-1 ) was injected i.p. for 25 days. Hypertrophy was evaluated by assessing morphological changes, echocardiographic parameters, histological analyses and hypertrophic markers. Oxidative stress was evaluated by examining ROS generation, oxidase activity and NO generation. NOS expression was determined by Western blotting. KEY RESULTS: Aucubin effectively suppressed cardiac remodelling; in mice, aucubin substantially inhibited pressure overload-induced cardiac hypertrophy, fibrosis and inflammation, whereas knocking out nNOS abolished these cardioprotective effects of aucubin. Blocking or knocking down the ß3 -adrenoceptor abolished the protective effects of aucubin in vitro. Furthermore, aucubin enhanced the protective effects of a ß3 -adrenoceptor agonist in vitro by increasing cellular cAMP levels, whereas treatment with an adenylate cyclase (AC) inhibitor abolished the cardioprotective effects of aucubin. CONCLUSIONS AND IMPLICATIONS: Aucubin suppresses oxidative stress during cardiac remodelling by increasing the expression of nNOS in a process that requires activation of the ß3 -adrenoceptor/AC/cAMP pathway. These findings suggest that aucubin could have potential as a treatment for cardiac remodelling and heart failure.


Assuntos
Insuficiência Cardíaca/prevenção & controle , Glucosídeos Iridoides/farmacologia , Óxido Nítrico Sintase Tipo I/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Remodelação Ventricular/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos beta 3/farmacologia , Antagonistas de Receptores Adrenérgicos beta 3/farmacologia , Animais , Cardiotônicos/farmacologia , Cardiotônicos/uso terapêutico , Células Cultivadas , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Hipertrofia/tratamento farmacológico , Hipertrofia/patologia , Glucosídeos Iridoides/uso terapêutico , Masculino , Camundongos , Camundongos Knockout , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Óxido Nítrico Sintase Tipo I/biossíntese , Óxido Nítrico Sintase Tipo I/genética , Estresse Oxidativo/efeitos dos fármacos , Fenilefrina/farmacologia , Substâncias Protetoras/farmacologia , Ratos , Receptores Adrenérgicos beta 3/genética
17.
Planta Med ; 84(2): 91-99, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28841738

RESUMO

Fibrosis is a key feature of various cardiovascular diseases and compromises cardiac systolic and diastolic performance. The lack of effective anti-fibrosis drugs is a major contributor to the increasing prevalence of heart failure. The present study was performed to investigate whether the iridoid aucubin alleviates cardiac fibroblast activation and its underlying mechanisms. Neonatal rat cardiac fibroblasts were incubated with aucubin (1, 10, 20, 50 µM) followed by transforming growth factor ß1 (TGFß1, 10 ng/mL) stimulation for 24 h. Fibrosis proliferation was measured by cell counting kit-8 assay. The differentiation of fibroblasts into myofibroblasts was determined by measuring the expression of α-smooth muscle actin. Then, the expressions levels of cardiac fibrosis-related proteins in myofibroblasts were analyzed by western blot and real-time PCR to confirm the anti-fibrosis effect of aucubin. As a result, aucubin suppressed TGFß1-induced proliferation in fibroblasts and inhibited the TGFß1-induced activation of fibroblasts to myofibroblasts. In addition, aucubin further attenuated fibrosis-related protein expression in myofibroblasts. Furthermore, this protective effect was related to increased adenosine 5'-monophosphate-activated protein kinase (AMPK) phosphorylation and decreased mammalian target of rapamycin (mTOR) phosphorylation, which was confirmed by an mTOR inhibitor (rapamycin), an AMPK agonist (AICAR) and an AMPKα inhibitor compound C. Collectively, our findings suggest that aucubin protects against TGFß1-induced fibroblast proliferation, activation and function by regulating the AMPKα/mTOR signal axis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Fibroblastos/efeitos dos fármacos , Glucosídeos Iridoides/farmacologia , Miocárdio/citologia , Serina-Treonina Quinases TOR/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fibroblastos/metabolismo , Fibrose/prevenção & controle , Coração/efeitos dos fármacos , Miocárdio/metabolismo , Ratos , Ratos Sprague-Dawley , Serina-Treonina Quinases TOR/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...