Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Korean Circ J ; 54(5): 253-255, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38767338
2.
Nat Commun ; 15(1): 46, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38167804

RESUMEN

Addressing age-related immunological defects through therapeutic interventions is essential for healthy aging, as the immune system plays a crucial role in controlling infections, malignancies, and in supporting tissue homeostasis and repair. In our study, we show that stimulating toll-like receptor 5 (TLR5) via mucosal delivery of a flagellin-containing fusion protein effectively extends the lifespan and enhances the healthspan of mice of both sexes. This enhancement in healthspan is evidenced by diminished hair loss and ocular lens opacity, increased bone mineral density, improved stem cell activity, delayed thymic involution, heightened cognitive capacity, and the prevention of pulmonary lung fibrosis. Additionally, this fusion protein boosts intestinal mucosal integrity by augmenting the surface expression of TLR5 in a certain subset of dendritic cells and increasing interleukin-22 (IL-22) secretion. In this work, we present observations that underscore the benefits of TLR5-dependent stimulation in the mucosal compartment, suggesting a viable strategy for enhancing longevity and healthspan.


Asunto(s)
Longevidad , Receptor Toll-Like 5 , Animales , Ratones , Flagelina/metabolismo , Mucosa Intestinal/metabolismo , Longevidad/genética , Pulmón/metabolismo
3.
Mol Ther Nucleic Acids ; 34: 102071, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38046397

RESUMEN

Heart failure is a leading cause of death and is often accompanied by activation of quiescent cardiac myofibroblasts, which results in cardiac fibrosis. In this study, we aimed to identify novel circular RNAs that regulate cardiac fibrosis. We applied transverse aortic constriction (TAC) for 1, 4, and 8 weeks in mice. RNA sequencing datasets were obtained from cardiac fibroblasts isolated by use of a Langendorff apparatus and then further processed by use of selection criteria such as differential expression and conservation in species. CircSMAD4 was upregulated by TAC in mice or by transforming growth factor (TGF)-ß1 in primarily cultured human cardiac fibroblasts. Delivery of si-circSMAD4 attenuated myofibroblast activation and cardiac fibrosis in mice treated with isoproterenol (ISP). si-circSmad4 significantly reduced cardiac fibrosis and remodeling at 8 weeks. Mechanistically, circSMAD4 acted as a sponge against the microRNA miR-671-5p in a sequence-specific manner. miR-671-5p was downregulated during myofibroblast activation and its mimic form attenuated cardiac fibrosis. miR-671-5p mimic destabilized fibroblast growth factor receptor 2 (FGFR2) mRNA in a sequence-specific manner and interfered with the fibrotic action of FGFR2. The circSMAD4-miR-671-5p-FGFR2 pathway is involved in the differentiation of cardiac myofibroblasts and thereby the development of cardiac fibrosis.

4.
Int J Mol Sci ; 24(17)2023 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-37686120

RESUMEN

Macrophages are the major primary immune cells that mediate the inflammatory response. In this process, long non-coding RNAs (lncRNAs) play an important, yet largely unknown role. Therefore, utilizing several publicly available RNA sequencing datasets, we predicted and selected lncRNAs that are differentially expressed in M1 or M2 macrophages and involved in the inflammatory response. We identified SUGCT-AS1, which is a human macrophage-specific lncRNA whose expression is increased upon M1 macrophage stimulation. Conditioned media of SUGCT-AS1-depleted M1 macrophages induced an inflammatory phenotype of vascular smooth muscle cells, which included increased expression of inflammatory genes (IL1B and IL6), decreased contractile marker proteins (ACTA2 and SM22α), and increased cell migration. Depletion of SUGCT-AS1 promoted the expression and secretion of proinflammatory cytokines, such as TNF, IL1B, and IL6, in M1 macrophages, and transcriptomic analysis showed that SUGCT-AS1 has functions related to inflammatory responses and cytokines. Furthermore, we found that SUGCT-AS1 directly binds to hnRNPU and regulates its nuclear-cytoplasmic translocation. This translocation of hnRNPU altered the proportion of the MALT1 isoforms by regulating the alternative splicing of MALT1, a mediator of NF-κB signaling. Overall, our findings suggest that lncRNAs can be used for future studies on macrophage regulation. Moreover, they establish the SUGCT-AS1/hnRNPU/MALT1 axis, which is a novel inflammatory regulatory mechanism in macrophages.


Asunto(s)
ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , Interleucina-6/genética , Empalme Alternativo , Proteínas Contráctiles , Citocinas/genética , Macrófagos
5.
Int J Cardiol ; 388: 131164, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37429444

RESUMEN

BACKGROUND: Thin-cap fibroatheroma is a rupture-prone vulnerable plaque that leads to acute coronary syndrome (ACS). However, its underlying mechanisms are not fully understood. Several studies have investigated the clinical association between angiopoietin-like protein 4 (ANGPTL4) and coronary artery disease. Therefore, this study aimed to investigate the correlation of plasma ANGPTL4 in culprit lesion of ACS patients using intravascular ultrasound (IVUS) and virtual-histology IVUS (VH-IVUS). METHODS: Fifty patients newly diagnosed with ACS between March to September 2021 were selected. Blood samples for baseline laboratory tests, including ANGPTL4, were collected before percutaneous coronary intervention (PCI), and all pre- and post-PCI IVUS examinations were performed of the culprit lesions. RESULTS: Linear regression analysis between plasma ANGPTL4 and grayscale IVUS/VH-IVUS parameters revealed that plasma ANGPTL4 was strongly correlated with the necrotic core (NC) of the minimal lumen site (r = -0.666, p = 0.003) and largest NC site (r = -0.687, p < 0.001), and patients with lower plasma ANGPTL4 levels showed a significantly higher proportion of TFCA. CONCLUSION: The present study further demonstrated the protective role of ANGPTL4 in the spectrum of atherosclerotic development in patients with ACS by culprit lesion morphology analysis using IVUS and VH-IVUS.


Asunto(s)
Síndrome Coronario Agudo , Enfermedad de la Arteria Coronaria , Intervención Coronaria Percutánea , Placa Aterosclerótica , Humanos , Síndrome Coronario Agudo/diagnóstico por imagen , Síndrome Coronario Agudo/patología , Angiopoyetinas , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/patología , Vasos Coronarios/diagnóstico por imagen , Necrosis/patología , Placa Aterosclerótica/diagnóstico por imagen , Placa Aterosclerótica/patología , Ultrasonografía Intervencional
6.
Korean J Physiol Pharmacol ; 27(4): 407-416, 2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-37386838

RESUMEN

The regeneration of myocardium following acute circulatory events remains a challenge, despite numerous efforts. Mesenchymal stem cells (MSCs) present a promising cell therapy option, but their differentiation into cardiomyocytes is a time-consuming process. Although it has been demonstrated that PSME4 degrades acetyl-YAP1, the role of PSME4 in the cardiac commitment of MSCs has not been fully elucidated. Here we reported the novel role of PSME4 in MSCs cardiac commitment. It was found that overnight treatment with apicidin in primary-cultured mouse MSCs led to rapid cardiac commitment, while MSCs from PSME4 knock-out mice did not undergo this process. Cardiac commitment was also observed using lentivirus-mediated PSME4 knockdown in immortalized human MSCs. Immunofluorescence and Western blot experiments revealed that YAP1 persisted in the nucleus of PSME4 knockdown cells even after apicidin treatment. To investigate the importance of YAP1 removal, MSCs were treated with shYAP1 and apicidin simultaneously. This combined treatment resulted in rapid YAP1 elimination and accelerated cardiac commitment. However, overexpression of acetylation-resistant YAP1 in apicidin-treated MSCs impeded cardiac commitment. In addition to apicidin, the universal effect of histone deacetylase (HDAC) inhibition on cardiac commitment was confirmed using tubastatin A and HDAC6 siRNA. Collectively, this study demonstrates that PSME4 is crucial for promoting the cardiac commitment of MSCs. HDAC inhibition acetylates YAP1 and facilitates its translocation to the nucleus, where it is removed by PSME4, promoting cardiac commitment. The failure of YAP1 to translocate or be eliminated from the nucleus results in the MSCs' inability to undergo cardiac commitment.

7.
ACS Nano ; 17(13): 12290-12304, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37339066

RESUMEN

Myocardial infarction (MI) is a major cause of death worldwide. After the occurrence of MI, the heart frequently undergoes serious pathological remodeling, leading to excessive dilation, electrical disconnection between cardiac cells, and fatal functional damage. Hence, extensive efforts have been made to suppress pathological remodeling and promote the repair of the infarcted heart. In this study, we developed a hydrogel cardiac patch that can provide mechanical support, electrical conduction, and tissue adhesiveness to aid in the recovery of an infarcted heart function. Specifically, we developed a conductive and adhesive hydrogel (CAH) by combining the two-dimensional titanium carbide (Ti3C2Tx) MXene with natural biocompatible polymers [i.e., gelatin and dextran aldehyde (dex-ald)]. The CAH was formed within 250 s of mixing the precursor solution and could be painted. The hydrogel containing 3.0 mg/mL MXene, 10% gelatin, and 5% dex-ald exhibited appropriate material characteristics for cardiac patch applications, including a uniform distribution of MXene, a high electrical conductivity (18.3 mS/cm), cardiac tissue-like elasticity (30.4 kPa), strong tissue adhesion (6.8 kPa), and resistance to various mechanical deformations. The CAH was cytocompatible and induced cardiomyocyte (CM) maturation in vitro, as indicated by the upregulation of connexin 43 expression and a faster beating rate. Furthermore, CAH could be painted onto the heart tissue and remained stably adhered to the beating epicardium. In vivo animal studies revealed that CAH cardiac patch treatment significantly improved cardiac function and alleviated the pathological remodeling of an infarcted heart. Thus, we believe that our MXene-based CAH can potentially serve as a promising platform for the effective repair of various electroactive tissues including the heart, muscle, and nerve tissues.


Asunto(s)
Hidrogeles , Infarto del Miocardio , Animales , Hidrogeles/farmacología , Gelatina/metabolismo , Adhesivos/farmacología , Infarto del Miocardio/patología , Miocitos Cardíacos , Polímeros/farmacología , Conductividad Eléctrica
8.
Exp Mol Med ; 55(2): 426-442, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36782020

RESUMEN

Atherosclerosis, the leading cause of death, is a vascular disease of chronic inflammation. We recently showed that angiopoietin-like 4 (ANGPTL4) promotes cardiac repair by suppressing pathological inflammation. Given the fundamental contribution of inflammation to atherosclerosis, we assessed the role of ANGPTL4 in the development of atherosclerosis and determined whether ANGPTL4 regulates atherosclerotic plaque stability. We injected ANGPTL4 protein twice a week into atherosclerotic Apoe-/- mice and analyzed the atherosclerotic lesion size, inflammation, and plaque stability. In atherosclerotic mice, ANGPTL4 reduced atherosclerotic plaque size and vascular inflammation. In the atherosclerotic lesions and fibrous caps, the number of α-SMA(+), SM22α(+), and SM-MHC(+) cells was higher, while the number of CD68(+) and Mac2(+) cells was lower in the ANGPTL4 group. Most importantly, the fibrous cap was significantly thicker in the ANGPTL4 group than in the control group. Smooth muscle cells (SMCs) isolated from atherosclerotic aortas showed significantly increased expression of CD68 and Krüppel-like factor 4 (KLF4), a modulator of the vascular SMC phenotype, along with downregulation of α-SMA, and these changes were attenuated by ANGPTL4 treatment. Furthermore, ANGPTL4 reduced TNFα-induced NADPH oxidase 1 (NOX1), a major source of reactive oxygen species, resulting in the attenuation of KLF4-mediated SMC phenotypic changes. We showed that acute myocardial infarction (AMI) patients with higher levels of ANGPTL4 had fewer vascular events than AMI patients with lower levels of ANGPTL4 (p < 0.05). Our results reveal that ANGPTL4 treatment inhibits atherogenesis and suggest that targeting vascular stability and inflammation may serve as a novel therapeutic strategy to prevent and treat atherosclerosis. Even more importantly, ANGPTL4 treatment inhibited the phenotypic changes of SMCs into macrophage-like cells by downregulating NOX1 activation of KLF4, leading to the formation of more stable plaques.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Ratones , Animales , Placa Aterosclerótica/patología , Factor 4 Similar a Kruppel , Músculo Liso Vascular , Regulación hacia Abajo , Ratones Noqueados para ApoE , Aterosclerosis/patología , Fenotipo , Miocitos del Músculo Liso/metabolismo , Inflamación/metabolismo , Ratones Endogámicos C57BL , Células Cultivadas
9.
Pharmaceutics ; 14(8)2022 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-36015285

RESUMEN

Intensive research has focused on minimizing the infarct area and stimulating endogenous regeneration after myocardial infarction. Our group previously elucidated that apicidin, a histone deacetylase (HDAC) inhibitor, robustly accelerates the cardiac commitment of naïve mesenchymal stem cells (MSCs) through acute loss of YAP1. Here, we propose the novel regulation of YAP1 in MSCs. We found that acute loss of YAP1 after apicidin treatment resulted in the mixed effects of transcriptional arrest and proteasomal degradation. Subcellular fractionation revealed that YAP1 was primarily localized in the cytoplasm. YAP1 was acutely relocalized into the nucleus and underwent proteasomal degradation. Interestingly, phosphor-S127 YAP1 was shuttled into the nucleus, suggesting that a mechanism other than phosphorylation governed the subcellular localization of YAP1. Apicidin successfully induced acetylation and subsequent dissociation of YAP1 from 14-3-3, an essential molecule for cytoplasmic restriction. HDAC6 regulated both acetylation and subcellular localization of YAP1. An acetylation-dead mutant of YAP1 retarded nuclear redistribution upon apicidin treatment. We failed to acquire convincing evidence for polyubiquitination-dependent degradation of YAP1, suggesting that a polyubiquitination-independent regulator determined YAP1 fate. Nuclear PSME4, a subunit of the 26 S proteasome, recognized and degraded acetyl YAP1 in the nucleus. MSCs from PSME4-null mice were injected into infarcted heart, and aberrant sudden death was observed. Injection of immortalized human MSCs after knocking down PSME4 failed to improve either cardiac function or the fibrotic scar area. Our data suggest that acetylation-dependent proteasome subunit PSME4 clears acetyl-YAP1 in response to apicidin treatment in the nucleus of MSCs.

10.
ACS Nano ; 16(5): 7471-7485, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35438981

RESUMEN

Implantable bioelectrodes enable precise recording or stimulation of electrical signals with living tissues in close contact. However, their performance is frequently compromised owing to inflammatory tissue reactions, which macrophages either induce or resolve by polarizing to an inflammatory (M1) or noninflammatory (M2) phenotype, respectively. Thus, we aimed to fabricate biocompatible and functional implantable conductive polymer bioelectrodes with optimal topography for the modulation of macrophage responses. To this end, we produced heparin-doped polypyrrole (PPy/Hep) electrodes of different surface roughness, with Ra values from 5.5 to 17.6 nm, by varying the charge densities during electrochemical synthesis. In vitro culture revealed that macrophages on rough PPy/Hep electrodes preferentially polarized to noninflammatory phenotypes. In particular, PPy/Hep-900 (Ra = 14 nm) was optimal with respect to electrochemical properties and the suppression of inflammatory M1 polarization. In vivo implantation indicated that PPy/Hep-900 significantly reduced macrophage recruitment, suppressed inflammatory polarization, and mitigated fibrotic tissue formation. In addition, the implanted PPy/Hep-900 electrodes could successfully record electrocardiographic signals for up to 10 days without substantial decreases in sensitivity, while other electrodes substantially lost their signal sensitivity during implantation. Altogether, we demonstrate that modulating the surface features of PPy/Hep can benefit the design and applications of high-performance and high-biocompatibility bioelectrodes.


Asunto(s)
Polímeros , Pirroles , Polímeros/química , Pirroles/química , Conductividad Eléctrica , Macrófagos , Electrodos Implantados
11.
Biomaterials ; 281: 121327, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34952262

RESUMEN

CRISPR/Cas9-mediated gene-editing technology has gained attention as a new therapeutic method for intractable diseases. However, the use of CRISPR/Cas9 for cardiac conditions such as myocardial infarction remains challenging due to technical and biological barriers, particularly difficulties in delivering the system and targeting genes in the heart. In the present study, we demonstrated the in vivo efficacy of the CRISPR/Cas9 magnetoplexes system for therapeutic genome editing in myocardial infarction. First, we developed CRISPR/Cas9 magnetoplexes that magnetically guided CRISPR/Cas9 system to the heart for efficient in vivo therapeutic gene targeting during heart failures. We then demonstrated that the in vivo gene targeting of miR34a via these CRISPR/Cas9 magnetoplexes in a mouse model of myocardial infarction significantly improved cardiac repair and regeneration to facilitate improvements in cardiac function. These results indicated that CRISPR/Cas9 magnetoplexes represent an effective in vivo therapeutic gene-targeting platform in the myocardial infarction of heart, and that this strategy may be applicable for the treatment of a broad range of cardiac failures.


Asunto(s)
Edición Génica , Infarto del Miocardio , Animales , Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Marcación de Gen , Terapia Genética/métodos , Ratones , Infarto del Miocardio/genética , Infarto del Miocardio/terapia
12.
Cell Death Dis ; 12(4): 307, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33753732

RESUMEN

Resistin-like alpha (Retnla) is a member of the resistin family and known to modulate fibrosis and inflammation. Here, we investigated the role of Retnla in the cardiac injury model. Myocardial infarction (MI) was induced in wild type (WT), Retnla knockout (KO), and Retnla transgenic (TG) mice. Cardiac function was assessed by echocardiography and was significantly preserved in the KO mice, while worsened in the TG group. Angiogenesis was substantially increased in the KO mice, and cardiomyocyte apoptosis was markedly suppressed in the KO mice. By Retnla treatment, the expression of p21 and the ratio of Bax to Bcl2 were increased in cardiomyocytes, while decreased in cardiac fibroblasts. Interestingly, the numbers of cardiac macrophages and unsorted bone marrow cells (UBCs) were higher in the KO mice than in the WT mice. Besides, phosphorylated histone H3(+) cells were more frequent in bone marrow of KO mice. Moreover, adiponectin in UBCs was notably higher in the KO mice compared with WT mice. In an adoptive transfer study, UBCs were isolated from KO mice to transplant to the WT infarcted heart. Cardiac function was better in the KO-UBCs transplanted group in the WT-UBCs transplanted group. Taken together, proliferative and adiponectin-rich bone marrow niche was associated with substantial cardiac recovery by suppression of cardiac apoptosis and proliferation of cardiac fibroblast.


Asunto(s)
Adipoquinas/metabolismo , Células de la Médula Ósea/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Infarto del Miocardio/fisiopatología , Animales , Apoptosis , Masculino , Ratones
13.
Mol Immunol ; 128: 98-105, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33099154

RESUMEN

Macrophages are the primary cell type in the immune system and are activated in response to diverse external stimuli. They can exert inflammatory or anti-inflammatory responses depending on the type of stimuli in the system, and as such rely on a complex network of diverse signaling pathways to function. Non-coding RNAs have recently been established as an important class of regulators linked to the activation of macrophages. Although the roles of many microRNAs have been described, the majority of the other types of regulatory non-coding RNAs, including long non-coding RNAs (lncRNAs), remain undefined. In this study, we performed a comprehensive analysis of several large RNA sequencing datasets to identify common lncRNAs differentially expressed after M1 or M2 macrophage induction. We identified the lncRNAs that underwent significant changes in each dataset and established a list of commonly altered lncRNAs. We expect that our data will act as a valuable resource for future studies designed to uncover the roles of these lncRNAs in determining macrophage fates.


Asunto(s)
Redes Reguladoras de Genes/genética , Activación de Macrófagos/genética , Macrófagos/fisiología , ARN Largo no Codificante/genética , Animales , Línea Celular , Perfilación de la Expresión Génica/métodos , Humanos , Ratones , Células RAW 264.7
14.
ASAIO J ; 66(4): 433-440, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31192851

RESUMEN

Extracorporeal membrane oxygenation (ECMO) is a well-known therapy for refractory cardiac and respiratory failure. Stem cell therapy has been investigated as an adjunctive treatment for use during ECMO, but little is known about the viability of stem cells during ECMO support. We evaluated the viability and activity of mesenchymal stem cells (MSCs) in ex vivo circulation (EVC) conditions. The experimental groups were divided into two subgroups: EVC with oxygenator (OXY group) and EVC without oxygenator (Non-OXY group). Mesenchymal stem cells (1.0 × 10) were injected into the EVC system. Cell counting, a lactate dehydrogenase (LDH) cytotoxicity assay, and the mitochondrial functions of viable MSCs were analyzed. The post-EVC oxygen consumption rate (OCR) was significantly lower than the pre-EVC OCR, regardless of whether the oxygenator was used. The LDH levels were significantly higher in the OXY group than in the Non-OXY group. The cellular loss was mainly due to lysis of the cells whereas the loss of cellular activity was attributed to the nonphysiologic condition itself, as well as the oxygenator. We concluded that direct infusion of MSCs during ECMO support did not serve as adjunctive therapy. Further studies are needed to improve the viability in an ECMO setting.


Asunto(s)
Oxigenación por Membrana Extracorpórea , Células Madre Mesenquimatosas/fisiología , Animales , Supervivencia Celular , Oxigenadores , Porcinos
15.
Cell Death Dis ; 10(11): 835, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31685805

RESUMEN

Ischaemic heart disease (IHD) is the leading cause of death worldwide. Although myocardial cell death plays a significant role in myocardial infarction (MI), its underlying mechanism remains to be elucidated. To understand the progression of MI and identify potential therapeutic targets, we performed tandem mass tag (TMT)-based quantitative proteomic analysis using an MI mouse model. Gene ontology (GO) analysis and gene set enrichment analysis (GSEA) revealed that the glutathione metabolic pathway and reactive oxygen species (ROS) pathway were significantly downregulated during MI. In particular, glutathione peroxidase 4 (GPX4), which protects cells from ferroptosis (an iron-dependent programme of regulated necrosis), was downregulated in the early and middle stages of MI. RNA-seq and qRT-PCR analyses suggested that GPX4 downregulation occurred at the transcriptional level. Depletion or inhibition of GPX4 using specific siRNA or the chemical inhibitor RSL3, respectively, resulted in the accumulation of lipid peroxide, leading to cell death by ferroptosis in H9c2 cardiomyoblasts. Although neonatal rat ventricular myocytes (NRVMs) were less sensitive to GPX4 inhibition than H9c2 cells, NRVMs rapidly underwent ferroptosis in response to GPX4 inhibition under cysteine deprivation. Our study suggests that downregulation of GPX4 during MI contributes to ferroptotic cell death in cardiomyocytes upon metabolic stress such as cysteine deprivation.


Asunto(s)
Regulación hacia Abajo , Ferroptosis , Regulación Enzimológica de la Expresión Génica , Infarto del Miocardio/enzimología , Miocitos Cardíacos/enzimología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/biosíntesis , Animales , Línea Celular , Humanos , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Proteómica , Ratas , Ratas Sprague-Dawley
16.
Biomaterials ; 225: 119513, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31569016

RESUMEN

Mesenchymal stem cell (MSC) transplantation is promising for repairing heart tissues post myocardial infarction (MI). In particular, paracrine effects of the transplanted MSCs have been highlighted to play major roles in heart regeneration by secreting multiple growth factors and immune-modulatory cytokines. Nevertheless, its therapeutic efficacy still remains low, which is strongly associated with low viability and activity of the transplanted stem cells, because the transplanted MSCs are exposed to high shear stress during injection and harsh environments (e.g., high oxidative stress and host immune reactions) post injection. In this study, we aimed to develop novel injectable MSC-delivery microgel systems possessing high anti-oxidant activities. Specifically, we encapsulated MSCs in graphene oxide (GO)/alginate composite microgels by electrospraying. To further enhance the anti-oxidizing activities of the gels, we developed reduced MSC-embedded GO/alginate microgels (i.e., r(GO/alginate)), which have the potential to protect MSCs from the abovementioned harsh environments within MI tissues. Our in vitro studies demonstrated that the MSCs encapsulated in the r(GO/alginate) microgels showed increased viability under oxidative stress conditions with H2O2. Furthermore, cardiomyocytes (CMs), co-cultured with the encapsulated MSCs in transwells with H2O2 treatment, showed higher cell viability and cardiac maturation compared to monolayer cultured CMs, likely due to ROS scavenging by the gels and positive paracrine signals from the encapsulated MSCs. In vivo experiments with acute MI models demonstrated improved therapeutic efficacy of MSC delivery in r(GO/alginate) microgels, exhibiting significant decreases in the infarction area and the improvement of cardiac function. We believe that our novel MSC encapsulation system with GO, alginate, and mild reduction, which exhibits high cell protection capacity (e.g., anti-oxidant activity), will serve as an effective platform for the delivery of stem cells and other therapeutic cell types to treat various injuries and diseases, including MI.


Asunto(s)
Alginatos/farmacología , Antioxidantes/farmacología , Células Inmovilizadas/citología , Grafito/farmacología , Células Madre Mesenquimatosas/citología , Microgeles , Infarto del Miocardio/terapia , Regeneración , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Inmovilizadas/efectos de los fármacos , Citocinas/biosíntesis , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Remodelación Ventricular/efectos de los fármacos
17.
Korean Circ J ; 49(12): 1196-1198, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31642218
18.
JCI Insight ; 4(16)2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31434807

RESUMEN

Mesenchymal stem cells (MSCs) can suppress pathological inflammation. However, the mechanisms underlying the association between MSCs and inflammation remain unclear. Under coculture conditions with macrophages, MSCs highly expressed angiopoietin-like 4 (ANGPTL4) to blunt the polarization of macrophages toward the proinflammatory phenotype. ANGPTL4-deficient MSCs failed to inhibit the inflammatory macrophage phenotype. In inflammation-related animal models, the injection of coculture medium or ANGPTL4 protein increased the antiinflammatory macrophages in both peritonitis and myocardial infarction. In particular, cardiac function and pathology were markedly improved by ANGPTL4 treatment. We found that retinoic acid-related orphan receptor α (RORα) was increased by inflammatory mediators, such as IL-1ß, and bound to ANGPTL4 promoter in MSCs. Collectively, RORα-mediated ANGPTL4 induction was shown to contribute to the antiinflammatory activity of MSCs against macrophages under pathological conditions. This study suggests that the capability of ANGPTL4 to induce tissue repair is a promising opportunity for safe stem cell-free regeneration therapy from a translational perspective.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/fisiología , Activación de Macrófagos , Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/terapia , Peritonitis/terapia , Proteína 4 Similar a la Angiopoyetina/genética , Proteína 4 Similar a la Angiopoyetina/metabolismo , Animales , Antiinflamatorios no Esteroideos , Polaridad Celular , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Inflamación/terapia , Mediadores de Inflamación/metabolismo , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infarto del Miocardio/inmunología , Miocarditis/etiología , Miocarditis/prevención & control , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Peritonitis/inmunología , Receptores de Ácido Retinoico/metabolismo
19.
Invest New Drugs ; 37(4): 796, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30937691

RESUMEN

The blots of control and docetaxel for caspase-9, caspase-3, caspase-8, Bcl-XL, and tubulin in the Figure 4f were reused from Figure 4 of our previous paper published in Journal of Urology in 2010 ( https://doi.org/10.1016/j.juro.2010.07.035 ).

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA