Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Mol Cell Biochem ; 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38652214

RESUMEN

Chronic liver damage (CLD) encompasses a spectrum of conditions and poses a significant global health challenge, affecting millions of individuals. Currently, there is a deficiency of clinically validated therapeutics with minimal side effects. Emerging evidence underscores the significant potential of extracellular vesicles derived from bone marrow mesenchymal stem cells (BMSC-EVs) as a promising therapeutic method for CLD. This study aimed to evaluate the influence of BMSC-EVs containing microRNA-136-5p (BMSC-EVs-miR-136-5p) on macrophage polarization during chronic liver injury and elucidate the mechanisms associated with the GNAS/PI3K/ERK/STAT3 axis. Surface markers of BMSCs were detected via Immunofluorescent Staining. Subsequently, EVs were harvested from the BMSC culture medium. In vivo fluorescence imaging was employed to locate the BMSC-EVs. Additionally, fluorescence microscopy was used to visualize the uptake of DIR-labeled BMSC-EVs by RAW264.7 cells. Various methods were employed to assess the impact of BMSC-EVs on the expression levels of inflammatory factors (IL-1ß, IL-6, IL-10, and TNF-α), M1/M2 macrophage markers (iNOS and Arg-1), and members of inflammation-related signaling pathways (GNAS, PI3K, ERK, and STAT3) in RAW264.7 cells co-cultured with BMSC-EVs. Loss-of-function approaches targeting miR-136-5p in RAW264.7 cells were subsequently utilized to validate the role of BMSC-EVs-miR-136-5p. The Luciferase Reporter Assay indicates that GNAS was identified to be a target of miR-136-5p, and miR-136-5p demonstrating increased within BMSC-EVs compared to Raw264.7-EVs. BMSC-EVs-miR-136-5p mitigated CCl4-induced liver inflammation and improved liver function by Suppressing the GNAS/STAT3 Signaling. Notably, miR-136-5p suppressed lipopolysaccharide (LPS)-induced inflammation in RAW264.7 cells. BMSC-EVs-miR-136-5p alleviates CLD by activating M2 polarization through the GNAS-mediated PI3K/ERK/STAT3 axis. Accordingly, the members of this axis may serve as therapeutic targets.

2.
Acta Pharmacol Sin ; 44(12): 2492-2503, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37468692

RESUMEN

Endothelial dysfunction, a central hallmark of cardiovascular pathogenesis in diabetes mellitus, is characterized by impaired endothelial nitric oxide synthase (eNOS) and NO bioavailability. However, the underlying mechanisms remain unclear. Here in this study, we aimed to identify the role of calmodulin (CaM) in diabetic eNOS dysfunction. Human umbilical vein endothelial cells and murine endothelial progenitor cells (EPCs) treated with high glucose (HG) exhibited downregulated CaM mRNA/protein and vascular endothelial growth factor (VEGF) expression with impeded eNOS phosphorylation and cell migration/tube formation. These perturbations were reduplicated in CALM1-knockdown cells but prevented in CALM1-overexpressing cells. EPCs from type 2 diabetes animals behaved similarly to HG-treated normal EPCs, which could be rescued by CALM1-gene transduction. Consistently, diabetic animals displayed impaired eNOS phosphorylation, endothelium-dependent dilation, and CaM expression in the aorta, as well as deficient physical interaction of CaM and eNOS in the gastrocnemius. Local CALM1 gene delivery into a diabetic mouse ischemic hindlimb improved the blunted limb blood perfusion and gastrocnemius angiogenesis, and foot injuries. Diabetic patients showed insufficient foot microvascular autoregulation, eNOS phosphorylation, and NO production with downregulated CaM expression in the arterial endothelium, and abnormal CALM1 transcription in genome-wide sequencing analysis. Therefore, our findings demonstrated that downregulated CaM expression is responsible for endothelium dysfunction and angiogenesis impairment in diabetes, and provided a novel mechanism and target to protect against diabetic endothelial injury.


Asunto(s)
Diabetes Mellitus Tipo 2 , Humanos , Ratones , Animales , Diabetes Mellitus Tipo 2/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Endotelio/metabolismo , Isquemia/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Neovascularización Fisiológica
3.
Circulation ; 144(10): 788-804, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34162222

RESUMEN

BACKGROUND: Calsequestrins (Casqs), comprising the Casq1 and Casq2 isoforms, buffer Ca2+ and regulate its release in the sarcoplasmic reticulum of skeletal and cardiac muscle, respectively. Human inherited diseases associated with mutations in CASQ1 or CASQ2 include malignant hyperthermia/environmental heat stroke (MH/EHS) and catecholaminergic polymorphic ventricular tachycardia. However, patients with an MH/EHS event often experience arrhythmia for which the underlying mechanism remains unknown. METHODS: Working hearts from conventional (Casq1-KO) and cardiac-specific (Casq1-CKO) Casq1 knockout mice were monitored in vivo and ex vivo by ECG and electric mapping, respectively. MH was induced by 2% isoflurane and treated intraperitoneally with dantrolene. Time-lapse imaging was used to monitor intracellular Ca2+ activity in isolated mouse cardiomyocytes or neonatal rat ventricular myocytes with knockdown, overexpression, or truncation of the Casq1 gene. Conformational change in both Casqs was determined by cross-linking Western blot analysis. RESULTS: Like patients with MH/EHS, Casq1-KO and Casq1-CKO mice had faster basal heart rate and ventricular tachycardia on exposure to 2% isoflurane, which could be relieved by dantrolene. Basal sinus tachycardia and ventricular ectopic electric triggering also occurred in Casq1-KO hearts ex vivo. Accordingly, the ventricular cardiomyocytes from Casq1-CKO mice displayed dantrolene-sensitive increased Ca2+ waves and diastole premature Ca2+ transients/oscillations on isoflurane. Neonatal rat ventricular myocytes with Casq1-knockdown had enhanced spontaneous Ca2+ sparks/transients on isoflurane, whereas cells overexpressing Casq1 exhibited decreased Ca2+ sparks/transients that were absent in cells with truncation of 9 amino acids at the C terminus of Casq1. Structural evaluation showed that most of the Casq1 protein was present as a polymer and physically interacted with ryanodine receptor-2 in the ventricular sarcoplasmic reticulum. The Casq1 isoform was also expressed in human myocardium. Mechanistically, exposure to 2% isoflurane or heating at 41 °C induced Casq1 oligomerization in mouse ventricular and skeletal muscle tissues, leading to a reduced Casq1/ryanodine receptor-2 interaction and increased ryanodine receptor-2 activity in the ventricle. CONCLUSIONS: Casq1 is expressed in the heart, where it regulates sarcoplasmic reticulum Ca2+ release and heart rate. Casq1 deficiency independently causes MH/EHS-like ventricular arrhythmia by trigger-induced Casq1 oligomerization and a relief of its inhibitory effect on ryanodine receptor-2-mediated Ca2+ release, thus revealing a new inherited arrhythmia and a novel mechanism for MH/EHS arrhythmogenesis.


Asunto(s)
Calsecuestrina/genética , Hipertermia Maligna/etiología , Miocardio/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Frecuencia Cardíaca/fisiología , Ventrículos Cardíacos/fisiopatología , Hipertermia Maligna/diagnóstico , Ratones , Ratones Noqueados , Canal Liberador de Calcio Receptor de Rianodina , Retículo Sarcoplasmático/fisiología , Taquicardia Ventricular , Tórax
4.
Acta Pharmacol Sin ; 43(8): 1970-1978, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34931018

RESUMEN

Cx43 is the major connexin in ventricular gap junctions, and plays a pivotal role in control of electrical and metabolic communication among adjacent cardiomyocytes. We previously found that Cx43 dephosphorylation at serine 282 (pS282) caused cardiomyocyte apoptosis, which is involved in cardiac ischemia/reperfusion injury. In this study we investigated whether Cx43-S282 hyper-phosphorylation could protect cardiomyocytes against apoptosis. Adenovirus carrying rat full length Cx43 gene (Cx43-wt) or a mutant gene at S282 substituted with aspartic acid (S282D) were transfected into neonatal rat ventricular myocytes (NRVMs) or injected into rat ventricular wall. Rat abdominal aorta constriction model (AAC) was used to assess Cx43-S282 phosphorylation status. We showed that Cx43 phosphorylation at S282 was increased over 2-times compared to Cx43-wt cells at 24 h after transfection, while pS262 and pS368 were unaltered. S282D-transfected cells displayed enhanced gap junctional communication, and increased basal intracellular Ca2+ concentration and spontaneous Ca2+ transients compared to Cx43-wt cells. However, spontaneous apoptosis appeared in NRVMs transfected with S282D for 34 h. Rat ventricular myocardium transfected with S282D in vivo also exhibited apoptotic responses, including increased Bax/Bcl-xL ratio, cytochrome c release as well as caspase-3 and caspase-9 activities, while factor-associated suicide (Fas)/Fas-associated death domain expression and caspase-8 activity remained unaltered. In addition, AAC-induced hypertrophic ventricles had apoptotic injury with Cx43-S282 hyper-phosphorylation compared with Sham ventricles. In conclusion, Cx43 hyper-phosphorylation at S282, as dephosphorylation, also triggers cardiomyocyte apoptosis, but through activation of mitochondrial apoptosis pathway, providing a fine-tuned Cx43-S282 phosphorylation range required for the maintenance of cardiomyocyte function and survival.


Asunto(s)
Apoptosis , Conexina 43 , Miocitos Cardíacos , Animales , Conexina 43/genética , Conexina 43/metabolismo , Mitocondrias , Miocitos Cardíacos/metabolismo , Fosforilación , Ratas , Serina/metabolismo
5.
Biochem Biophys Res Commun ; 513(3): 567-572, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-30981509

RESUMEN

Connexin 43 (Cx43) phosphorylation plays a pivotal role in cardiac electrical and contractile performance. In a previous study we have found that Cx43 phosphorylation at serine 282 (pS282) regulates cardiomyocyte survival. Considering that both sites are altered simultaneously in many studies, we designed this study to identify the status of S279 phosphorylation upon pS282 manipulation. In heterozygous mice with S282 gene substituted with alanine (S282A), we found ventricular arrhythmias with inhibition of Cx43 phosphorylation at both S282 and S279 in the hearts. In cultured neonatal rat ventricular myocytes (NRVMs), transfection of virus carrying S282A mutant also blocked Cx43 phosphorylation at both S279/282 and gap junction coupling, while expression of wild-type Cx43 or S279A did not. Further, NRVMs transfected with S282 phospho-mimicking mutant substituted with aspartate or treated with ATP exhibited promotions of Cx43 phosphorylation at S279/282 and intercellular communication. Therefore, this study demonstrated a regulatory role of Cx43-S282 on S279 phosphorylation in cardiomyocytes, and suggested an involvement of S279 in the Cx43-S282 mediated cardiomyocyte homeostasis.


Asunto(s)
Conexina 43/metabolismo , Miocitos Cardíacos/metabolismo , Serina/metabolismo , Animales , Comunicación Celular , Células Cultivadas , Conexina 43/química , Conexina 43/genética , Doxorrubicina/farmacología , Uniones Comunicantes/metabolismo , Masculino , Ratones Endogámicos C57BL , Mutación , Fosforilación/efectos de los fármacos , Ratas Sprague-Dawley
6.
Cell Physiol Biochem ; 46(3): 1042-1054, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29669320

RESUMEN

BACKGROUND/AIMS: Upon Ca2+ store depletion, stromal interaction molecule 1 (STIM1) oligomerizes, redistributes near plasmalemma to interact with Ca2+ selective channel-forming subunit (Orai1) and initiates store-operated Ca2+ entry (SOCE). Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a regulator of SOCE, but how CaMKII regulates SOCE remains obscure. METHODS: Using Fura2, confocal microscopy, co-immunoprecipitation, specific blocker and overexpression/knockdown approaches, we evaluated STIM1 aggregation and its interaction with Orai1, and SOCE upon Ca2+ store depletion in thapsigargin (TG) treated HEK293 and HeLa cells. RESULTS: Overexpression of CaMKIIδ enhanced TG-induced STIM1 co-localization and interaction with Orai1 as well as SOCE. In contrast, CaMKIIδ knockdown and a specific inhibitor of CaMKII suppressed them. In addition, overexpression or knockdown of CaMKIIδ in TG treated cells exhibited increased or reduced STIM1 clustering and plasmalemma redistribution, respectively. CONCLUSION: CaMKII up-regulates SOCE by increasing STIM1 aggregation and interaction with Orai1. This study provides an additional insight into SOCE regulation and a potential mechanism for CaMKII involvement in some pathological situations through crosstalk with SOCE.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calcio/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Células HEK293 , Células HeLa , Humanos , Inmunoprecipitación , Fosforilación/efectos de los fármacos , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Multimerización de Proteína/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Tapsigargina/farmacología
7.
Biochem Biophys Res Commun ; 500(2): 384-390, 2018 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-29654766

RESUMEN

BACKGROUND/AIMS: It has been suggested that diabetes is associated with immune dysfunction, in which Ca2+ signaling malfunction in lymphocyte may contributes most. However, the pattern of the Ca2+ signal disorder and the mechanism(s) that explains the change are unclear. Here, in this study we aimed to investigate possible changes and mechanism(s) accounting for the internal Ca2+ signals in diabetic T lymphocyte upon stimulation. METHODS AND RESULTS: Using Fura-2-AM, we found a significant decrease in Ca2+ influx induced by thapsigargin (TG) and anti-CD3 antibody (OKT3) in T lymphocytes from blood of both diabetes patients and animals. Furthermore, a downregulated Orai1 protein expression, but not mRNA, was also observed in these cells using western blot and qRT-PCR, respectively. In addition, in high-glucose and agonist treated Jurkat T cells, Ca2+ entry and the release of interleukin-2 (IL-2) were also decreased. Orai1 expression reduced, while stromal interaction molecule 1 (STIM1) and other downstream proteins remained unchanged. CONCLUSION: This study demonstrates that the declined Orai1 expression, at least partly, contributes to the downregulated Ca2+ entry during lymphocyte excitation, providing an important mechanism for T lymphocyte malfunction in diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Regulación hacia Abajo , Linfocitos/metabolismo , Proteína ORAI1/metabolismo , Animales , Calcio/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Diabetes Mellitus Tipo 2/sangre , Regulación hacia Abajo/efectos de los fármacos , Glucosa/toxicidad , Humanos , Linfocitos/efectos de los fármacos , Ratones Endogámicos C57BL , Ratas Wistar
8.
Anal Bioanal Chem ; 409(29): 6885-6892, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28975377

RESUMEN

A sensitive fluorescence immunoassay for the detection of imidaclothiz was established by using magnetic nanoparticles (MNPs) as concentration elements and upconversion nanoparticles (UCNPs) as signal labels. The NaYF4/Yb,Er UCNPs and MNPs were conjugated with imidaclothiz monoclonal antibody and imidaclothiz antigen, respectively. Imidaclothiz could compete with the antigen-conjugated MNPs for binding to the antibody-conjugated UCNPs and resulted in a decreased fluorescence signal when the MNPs were separated by an external magnet. Under the optimal conditions, the concentration of imidaclothiz producing 50% inhibition of the signal (IC50), limit of detection (LOD, IC10), and the linear assay range (IC10-IC90) were 14.59, 0.74, and 0.74-289.30 ng mL-1, respectively. The immunoassay exhibited no obvious cross-reactivity with analogues of imidaclothiz except for imidacloprid, with 89.2% cross-reactivity. The average recoveries measured in paddy water, pear, soil, peach, rice, tomato, wheat, and pakchoi were 75.7-105.2%, and the relative standard deviations (RSDs) were less than 11.2%. In addition, the results of the immunoassay correlated well with that of high-performance liquid chromatography (HPLC) for authentic samples. Graphical abstract Development of an upconversion fluorescence immunoassay for the detection of imidaclothiz by using antibody-modified upconversion nanoparticles (UCNPs) as the detection probe and antigen-modified magnetic nanoparticles (MNPs) as the capture probe.


Asunto(s)
Técnica del Anticuerpo Fluorescente , Inmunoensayo/métodos , Nanopartículas de Magnetita/química , Tiazoles/análisis , Cromatografía Líquida de Alta Presión , Fluorescencia , Límite de Detección , Magnetismo , Tiazoles/química
9.
Heliyon ; 10(9): e29977, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38756592

RESUMEN

Chronic hepatitis (CH) encompasses a prevalent array of liver conditions that significantly contribute to global morbidity and mortality. Yiguanjian (YGJ) is a classical traditional Chinese medicine with a long history of medicinal as a treatment for CH. Although it has been reported that YGJ can reduce liver inflammation, the intricate mechanism requires further elucidation. We used network pharmacology approaches in this work, such as gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and network-based analysis of protein-protein interactions (PPIs), to clarify the pharmacological constituents, potential therapeutic targets, and YGJ signaling pathways associated with CH. Employing the random walk restart (RWR) algorithm, we identified GNAS, GNB1, CYP2E1, SFTPC, F2, MAPK3, PLG, SRC, HDAC1, and STAT3 as pivotal targets within the PPI network of YGJ-CH. YGJ attenuated liver inflammation and inhibited GNAS/STAT3 signaling in vivo. In vitro, we overexpressed the GNAS gene further to verify the critical role of GNAS in YGJ treatment. Our findings highlight GNAS/STAT3 as a promising therapeutic target for CH, providing a basis and direction for future investigations.

10.
Artículo en Inglés | MEDLINE | ID: mdl-38898365

RESUMEN

Sinoatrial node (SAN) is the pacemaker of the heart in charge of initiating spontaneous electronical activity and controlling heart rate. Myocytes from SAN can generate spontaneous rhythmic action potentials, which propagate through the myocardium, thereby triggering cardiac myocyte contraction. Acutely, the method for isolating sinoatrial node myocytes (SAMs) is critical in studying the protein expression and function of myocytes in SAN. Currently, the SAMs were isolated by transferring SAN tissue directly into the digestion solution, but it is difficult to judge the degree of digestion, and the system was unstable. Here, we present a modified protocol for the isolation of SAMs in mice, based on the collagenase II and protease perfusion of the heart using a Langendorff apparatus and subsequent dissociation of SAMs. The appearance and droplet flow rate of the heart could be significantly changed during enzymatic digestion via perfusion, which allowed us to easily judge the degree of digestion and avoid incomplete or excessive digestion. The SAMs with stable yield and viability achieved from our optimized approach would facilitate the follow-up experiments.

12.
Heliyon ; 9(5): e15879, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37215881

RESUMEN

Background: Connexin 43 (Cx43), the predominant gap junction protein in hearts, is modified by specific (de)phosphorylation events under physiological and pathological states to affect myocardium function and structure. Previously we found that deficiency in Cx43 S282 phosphorylation could impair intercellular communication and contribute to cardiomyocyte apoptosis by activating p38 mitogen-activated protein kinase (p38 MAPK)/factor-associated suicide (Fas)/Fas-associating protein with a novel death domain (FADD) pathway, which is involved in myocardium injury in ischemia/reperfusion (I/R) heart. In addition, mutant at Cx43 S282 substituted with alanine heterozygous mice (S282A+/-) exhibited different degrees of ventricular arrhythmias and only some underwent myocardium apoptosis. In this study, we aimed to investigate the role of Cx43 pS282 in different cardiac pathological phenotypes. Methods: We examined cardiac function, structure, and relevant protein expression in S282A+/- mice (aged 2, 10 and 30 weeks) by electrocardiograph, echocardiography, histological staining, and co-immunoprecipitation followed by Western blot. Intraperitoneal isoprenaline injection and I/R surgery were applied in S282A+/- mice as external stimulus. 2,3,5-triphenyltetrazolium chloride staining was used for myocardium infarction evaluation. Results: Adult S282A+/- mice (aged 10 and 30 weeks) still exhibited spontaneous arrhythmia. Unlike neonatal stage (aged around 2 weeks), no apoptosis-related manifestations and the activation of p38 MAPK-Fas-FADD apoptotic pathway were observed in adult S282A+/- hearts. S282A+/- neonatal mice with cardiomyocytes apoptosis exhibited more than 60% dephosphorylation at Cx43 S282 than WT mice, while less than 40% S282 dephosphorylation were found in adult S282A+/- mice. In addition, although S282A+/- mice displayed normal cardiac function, they were highly susceptible to isoproterenol-induced ECG alternans and prone to cardiac injury and deaths upon I/R attack. Conclusions: These results reinforce that Cx43 S282 dephosphorylation acts as a susceptibility factor in regulating cardiomyocyte survival and cardiac electrical homeostasis in basal conditions and contributes to myocardium injury in the setting of I/R. Cx43 S282 phosphorylation was competent to induce spontaneous arrhythmias, cardiomyocyte apoptosis and deaths based on the degree of S282 dephosphorylation.

13.
Front Pharmacol ; 13: 863797, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35721175

RESUMEN

Bone marrow mesenchymal stem cells (BMSCs) can effectively alleviate liver fibrosis, but the efficacy of cell therapy alone is insufficient. In recent years, a combination of traditional Chinese medicine (TCM) and cell therapy has been increasingly used to treat diseases in clinical trials. Ferulic acid (FA) is highly effective in treating liver fibrosis, and a combination of cells and drugs is being tested in clinical trials. Therefore, we combined BMSCs and Ferulic acid to treat CCl4-induced fibrosis and determine whether this combination was more effective than single treatment. We used BMSCs and FA to treat CCl4-induced fibrosis in rat models, observed their therapeutic effects, and investigated the specific mechanism of this combination therapy in liver fibrosis. We created a BMSC/hepatic stellate cell (HSC) coculture system and used FA to treat activated HSCs to verify the specific mechanism. Then, we used cytochalasin D and angiotensin II to investigate whether BMSCs and FA inactivate HSCs through cytoskeletal rearrangement. MiR-19b-3p was enriched in BMSCs and targeted TGF-ß receptor II (TGF-ßR2). We separately transfected miR-19b-3p into HSCs and BMSCs and detected hepatic stellate cell activation. We found that the expression of the profibrotic markers α-SMA and COL1-A1 was significantly decreased in the combination group of rats. α-SMA and COL1-A1 levels were also significantly decreased in the HSCs with the combination treatment. Cytoskeletal rearrangement of HSCs was inhibited in the combination group, and RhoA/ROCK pathway gene expression was decreased. Following angiotensin II treatment, COL1-A1 and α-SMA expression increased, while with cytochalasin D treatment, profibrotic gene expression decreased in HSCs. The expression of COL1-A1, α-SMA and RhoA/ROCK pathway genes was decreased in the activated HSCs treated with a miR-19b-3p mimic, indicating that miR-19b-3p inactivated HSCs by suppressing RhoA/ROCK signalling. In contrast, profibrotic gene expression was significantly decreased in the BMSCs treated with the miR-19b-3p mimic and FA or a miR-19b-3p inhibitor and FA compared with the BMSCs treated with the miR-19b-3p mimic alone. In conclusion, the combination therapy had better effects than FA or BMSCs alone. BMSC and FA treatment attenuated HSC activation and liver fibrosis by inhibiting cytoskeletal rearrangement and delivering miR-19b-3p to activated HSCs, inactivating RhoA/ROCK signalling. FA-based combination therapy showed better inhibitory effects on HSC activation.

14.
Cell Death Differ ; 26(7): 1332-1345, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30770876

RESUMEN

Gap junction protein connexin 43 (Cx43) plays an important role in regulating cardiomyocyte survival in addition to regulating electrical coordination. Cx43 dephosphorylation, found in severe cardiac pathologies, is thought to contribute to myocardial injury. However, the mechanisms underlying Cx43 mediation of cell survival and myocardial lesions remain unknown. Here, we found that transfecting an adenovirus carrying a mutant gene of Cx43-serine 282 substituted with alanine (S282A) into neonatal rat ventricular myocytes (NRVMs) induced cell apoptosis and Ca2+ transient desynchronization, whereas using gap junction inhibitor or knocking down Cx43 expression with Cx43-miRNA caused uncoupled Ca2+ signaling without cell death. Similarly, while Cx43-S282A+/+ failed in generation, Cx43-S282A+/- mice exhibited cardiomyocyte apoptosis and ventricular arrhythmias dependent on S282 dephosphorylation. Further, Cx43 dephosphorylation at S282 activated p38 mitogen-activated protein kinase (p38 MAPK), factor-associated suicide and the caspase-8 apoptotic pathway by physically interacting with p38 MAPK. These findings uncovered a specific Cx43 phosphorylation residue involved in regulating cardiomyocyte homeostasis. S282 phosphorylation deficiency acts as a trigger inducing cardiomyocyte apoptosis and cardiac arrhythmias, providing a potential mechanism for Cx43-mediated myocardial injury in severe cardiac diseases.


Asunto(s)
Apoptosis , Conexina 43/metabolismo , Serina/metabolismo , Animales , Calcio/análisis , Calcio/metabolismo , Células Cultivadas , Colorantes Fluorescentes/química , Isoquinolinas/química , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosforilación
15.
Food Chem ; 257: 289-294, 2018 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-29622212

RESUMEN

An upconversion fluorescence DNA probe which consists of aptamer-conjugated magnet nanoparticles (apt-MNPs) and complementary DNA-conjugated upconversion nanoparticles (cDNA-UCNPs) was developed to detect acetamiprid. Acetamiprid can specifically conjugate with the apt-MNPs to dissociate the cDNA-UCNPs from the apt-MNPs and resulted in reduced fluorescence intensity through an external magnet. The change of fluorescence intensity (△I) is positively related to the concentration of acetamiprid, which can be applied for the quantification of acetamiprid. Under optimal conditions, a linear detection range and detection limit are 0.89-114.18 µg/L and 0.65 µg/L, respectively. The probe was successfully used to detect acetamiprid in spiked paddy water, soil, pear, apple, wheat and cucumber. Average recoveries are 78.2%-103.5% with intra-day relative standard deviations (RSDs) of 2.6%-10.9% and inter-day RSDs of 4.3%-10.2%. The amounts of acetamiprid in the authentic paddy water and pear samples detected by the DNA probe are significantly correlated with that detected by high-performance liquid chromatography (HPLC).


Asunto(s)
Sondas de ADN/química , Colorantes Fluorescentes/química , Contaminación de Alimentos/análisis , Nanopartículas de Magnetita/química , Neonicotinoides/análisis , Aptámeros de Nucleótidos/química , Cucumis sativus/química , Insecticidas/análisis , Límite de Detección , Malus/química , Pyrus/química , Contaminantes del Suelo/análisis , Espectroscopía Infrarroja por Transformada de Fourier , Triticum/química , Contaminantes Químicos del Agua/análisis
16.
J Ethnopharmacol ; 210: 434-442, 2018 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-28917976

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Low molecular weight fucoidan (LMWF), extracted from Laminaria japonica Areschoug, is a traditional Chinese medicine, commonly used to alleviate edema, particularly for feet with numbness and pain. AIM OF THE STUDY: Diabetic mellitus (DM) patients are at high risk of developing peripheral arterial disease (PAD). Individuals with DM and PAD co-morbidity have a much higher risk of critical limb ischemia. LMWF showed several beneficial effects, such as anti-inflammation, anti-thrombosis, and enhancing revascularization. Therefore, we hypothesized that LMWF might be beneficial to diabetes-induced PAD, and investigated the therapeutic potential of LMWF on diabetic PAD rats. MATERIALS AND METHODS: Type 2 diabetic Goto-Kakizaki (GK) rats were made PAD by injection of sodium laurate into femoral artery. LMWF (20, 40 or 80mg/kg/day) or cilostazol (100mg/kg/day) were given to diabetic PAD rats for 4 weeks, respectively. The effects of LMWF on foot ulceration and claudication, plantar blood flow, collateral vessel formation, endothelium morphology, gastrocnemius injury, platelet aggregation, vessel vasodilation, and the expressions of inflammation factors, VEGF, eNOS, and nitric oxide were measured. RESULTS: We found that LMWF markedly ameliorated foot ulceration and claudication, and improved the plantar perfusion by reversing hyperreactive platelet aggregation, ameliorating endothelium-dependent vasodilation and revascularization on diabetic PAD rats. In addition, upregulation of several inflammatory factors, such as ICAM-1 and IL-1ß in the gastrocnemius muscles of ischemic hindlimb were suppressed by LMWF administration. And eNOS phosphorylation at Ser1177 and NO production were significantly enhanced in LMWF-treated diabetic PAD rats. CONCLUSIONS: Taken together, our findings demonstrated that LMWF exhibits therapeutic effect on hindlimb ischemia in type 2 diabetic rats likely through ameliorating endothelium eNOS dysfunction and enhancing revascularization, thus, providing a potential supplementary non-invasive treatment for diabetes-induced PAD.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Isquemia/tratamiento farmacológico , Polisacáridos/farmacología , Animales , Cilostazol , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Relación Dosis-Respuesta a Droga , Miembro Posterior/irrigación sanguínea , Laminaria/química , Masculino , Medicina Tradicional China , Peso Molecular , Óxido Nítrico Sintasa de Tipo III/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Enfermedad Arterial Periférica/tratamiento farmacológico , Enfermedad Arterial Periférica/etiología , Polisacáridos/administración & dosificación , Polisacáridos/aislamiento & purificación , Ratas , Ratas Wistar , Tetrazoles/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
17.
J Diabetes ; 10(11): 820-834, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29633569

RESUMEN

BACKGROUND: Diabetes mellitus (DM) complications are associated with ischemic injury. Angiogenesis is a therapeutic strategy for diabetic foot. The aim of this study was to investigate the possible angiogenic effect of low molecular weight fucoidan (LMWF) in diabetic peripheral arterial disease (PAD). METHODS: Diabetic db/db mice and age-matched C57BL/6 mice underwent femoral artery ligation followed by LMWF (30, 60, 80 mg/kg per day, p.o.) or cilostazol (30 mg/kg/day, p.o.) treatment for 6 weeks. Endothelium-dependent vasodilation and blood flow of the hindlimb were measured. Histological and western blot analyses of CD34, vascular endothelial growth factor (VEGF), eNOS, and inflammatory factors in the gastrocnemius were performed. The effects of LMWF were confirmed in human umbilical vein endothelial cells (HUVEC). RESULTS: Diabetic mice with ligation exhibited hindlimb ulceration, hydrosarca, and necrosis, increased expression of inflammatory factors, and decreased levels of VEGF and eNOS phosphorylation. Treatment with LMWF markedly ameliorated foot lesions, suppressed expression of inflammatory factors, and improved plantar perfusion by promoting endothelium-dependent vasodilation and revascularization in diabetic PAD mice. In high-glucose treated HUVEC, LMWF (40 µg/mL) reversed blunted endothelial cell proliferation, migration, and tube formation, and promoted eNOS phosphorylation and VEGF expression, whereas HUVEC pretreatment with 100 µmol/L NG -nitro-l-arginine methyl ester, an eNOS antagonist, markedly inhibited the effects of LMWF. CONCLUSION: This study demonstrates that LMWF alleviates hindlimb ischemic damage, at least in part by promoting eNOS phosphorylation, nitric oxide production, and VEGF expression, resulting in enhanced angiogenesis in the ischemic region.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Angiopatías Diabéticas/prevención & control , Isquemia/prevención & control , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Polisacáridos/farmacología , Animales , Células Cultivadas , Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/fisiopatología , Angiopatías Diabéticas/enzimología , Angiopatías Diabéticas/patología , Angiopatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Isquemia/enzimología , Isquemia/patología , Isquemia/fisiopatología , Ratones Endogámicos C57BL , Peso Molecular , Óxido Nítrico/metabolismo , Fosforilación , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 28(4): 520-3, 2006 Aug.
Artículo en Zh | MEDLINE | ID: mdl-16995305

RESUMEN

OBJECTIVE: To investigate the role of endothelin-1 and its receptors on hypertrophy or proliferation of cultured cardial cells. METHODS: Cardiomyocytes and cardiac fibroblasts were isolated by trypsin digestion method, DNA and protein synthesis were measured by 3H-dexyribonucleotidethymine (3H-TdR) and 3H-Leucine (3H-Leu) incorporation, while protein content was measured by Bradford method. Atrial natriuretic peptide (ANP) mRNA expression of cardiomyocyte was measured by reverse transcripted-polymerase chain reaction. Selective endothelin (ET) receptor subtype antagonists BQ123 and BQ788 were used to block ET(A) receptors (ET(A)R) and ET(B)R respectively and to observe the effects of the two receptors during cardiac hypertrophy. RESULTS: ET-1 significantly increased the 3H-TdR and 3H-Leu incorporation rate of cardiomyocytes and cardiac fibroblasts in a dose-dependent manner and increased protein content. Furthermore, ET-1 promoted the ANP mRNA expression of cardiomyocyte. ET(A)R antagonist remarkably blocked these effects, while ET(B)R antagonist had no obvious effect. CONCLUSIONS: ET-1 can induce the hypertrophy for cardiomyocytes and the proliferation for cardiac fibroblasts. These effects are mediated by ET(A)R.


Asunto(s)
Endotelina-1/fisiología , Miocitos Cardíacos/citología , ARN Mensajero/biosíntesis , Receptor de Endotelina A/fisiología , Animales , Animales Recién Nacidos , Factor Natriurético Atrial/biosíntesis , Factor Natriurético Atrial/genética , Proliferación Celular , Células Cultivadas , Fibroblastos/citología , Fibroblastos/patología , Hipertrofia , Miocitos Cardíacos/patología , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley
19.
J Tradit Chin Med ; 36(4): 471-8, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-28459511

RESUMEN

OBJECTIVE: To investigate the inhibitory effect of Yiguanjian decoction (YD) on DNA damage in Concanavalin A (Con A)-induced liver injury mice model and to explain the possible mechanism. METHODS: METHODS: Totally 120 male BALB/c mice were randomly divided into 6 groups, 20 mice each: normal group, model group, Bifendate group, YD low dose group, YD middle dose group and YD high dose group. Except normal group, liver injury model induced by Con A was established. While modeling, each mouse in YD group was given YD (0.4 mL/20 g per day) by intragastric administration (0.13 g YD for YD low dose group; 0.26 g for YD middle dose group; 0.52 g for YD high dose group). Bifendate group was given Bifendate (0.2 g·kg-1·d-1) by gavage. Normal group and model group were fed with same volume of physiological saline daily. After 8 weeks, the serum alanine transaminase (ALT) and aspartate transaminase (AST) were tested. The hematoxylin-eosin staining was used to evaluate the grade of liver inflammation and liver fibrosis stage. Hepatocellular DNA damage was detected by single cell gel electrophoresis technology. The protein expression of tumor necrosis factor-α (TNF-α), Bax and MutT Homolog 1 (MTH1) was detected by western blotting and enzyme linked immunosorbent assay. Bax mRNA and MTH1 mRNA were detected by Real-time Polymerase Chain Reaction (PCR). RESULTS: YD can improve the degree of liver inflammation and fibrosis in the liver of chronic hepatitis mice, the dose effect relationship is remarkable (P < 0.05). YD can reduce liver cell DNA damage. The difference between YD middle dose group and model group was statistically significant (P < 0.05). YD middle dose group had decreased the protein expression of TNF-α in the mice liver of immunological liver injury (P < 0.05). YD can increase the protein expression of Bax (P < 0.05). Compared with normal group, the protein expression of MTH1 was decreased (P < 0.05), but there was no statistical significance between YD group and model group (P > 0.05). YD can increase the mRNA expression of Bax and MTH1 (both P < 0.05). CONCLUSION: YD can effectively inhibit the DNA damage in immunological liver injury mice, the mechanism may be that it can decrease the TNF-α and increase the Bax and MTH1 expression.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Concanavalina A/efectos adversos , Daño del ADN/efectos de los fármacos , Medicamentos Herbarios Chinos/administración & dosificación , Sustancias Protectoras/administración & dosificación , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Modelos Animales de Enfermedad , Humanos , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos BALB C
20.
Curr Pharm Des ; 20(11): 1604-13, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23888946

RESUMEN

Nicotinamide phosphoribosyl transferase (Nampt) is the rate-limiting enzyme for the salvage biosynthesis of nicotinamide adenine dinucleotide (NAD). Although elevated level of Nampt expression has been observed in various cancers, the involvement of Nampt promoter regulation was not well understood. We have identified a cluster of MEF2 recognition sites upstream of the functional hypoxia response elements (HREs) within the human Nampt promoter, and demonstrated that the two MEF2 sites at -1272 and -1200 were functional to upregulate the promoter activity by luciferase reporter assays. The Nampt promoter was able to be activated cooperatively following hypoxic stimulation by CoCl2 treatment with associated MEF2C overexpression. During the investigation on MEF2C regulation of endogenous Nampt expression in HeLa cells, the most significant enhancement of Nampt expression observed was by overexpression of MEF2C in combination with sodium butyrate exposure. By chromatin immunoprecipitation with a MEF2C anti-body, we found that MEF2C indeed interacted with endogenous Nampt promoter. The requirement of HDAC inhibition for the MEF2C enhancement of Nampt transcription was verified by RNAi of HDAC. Our results were in support of reports indicating that MEF2 family transcription factors interacted with HDACs and regulated downstream gene expression at the epigenetic levels. Our study provided important evidence to demonstrate the sophisticated mechanism of endogenous Nampt promoter regulation, and therefore, will help to better understand the Nampt overexpression in cancer progression, especially in the context of MEF2C upregulation which frequently occurred in cancer development and drug resistance.


Asunto(s)
Ácido Butírico/farmacología , Citocinas/genética , Inhibidores de Histona Desacetilasas/farmacología , Nicotinamida Fosforribosiltransferasa/genética , Hipoxia de la Célula , Inmunoprecipitación de Cromatina , Epigénesis Genética , Regulación de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Factores de Transcripción MEF2/genética , NAD/biosíntesis , Regiones Promotoras Genéticas , Interferencia de ARN , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA