44 research outputs found

    Humulus japonicus attenuates LPS-and scopolamine-induced cognitive impairment in mice

    Get PDF
    Background : Neuroinflammation plays an important role in cognitive decline and memory impairment in neurodegenerative disorders. Previously, we demonstrated that Humulus japonicus (HJ) has anti-inflammatory effects in rodent models of Alzheimer’s disease and Parkinson’s disease. The present study aimed to examine the protective potential of HJ extracts against lipopolysaccharide (LPS)-induced cognitive impairment and scopolamine-induced amnesia in mouse models. Cognitive improvement of mice was investigated by novel object recognition test. For analyzing effects on neuroinflammation, immunohistochemistry and quantitative real-time polymerase chain reaction (qRT-PCR) assays were performed. Results : We found that the oral administration of HJ significantly improved cognitive dysfunction induced by LPS in a novel object recognition test. The LPS-induced activation of microglia was notably decreased by HJ treatment in the cortex and hippocampus. HJ administration with LPS also significantly increased the mRNA expression of interleukin (IL)-10 and decreased the mRNA expression of IL-12 in the parietal cortex of mice. The increased expression of LPS-induced complement C1q B chain (C1bq) and triggering receptor expressed on myeloid cells 2 (Trem2) genes was significantly suppressed by HJ treatment. In addition, HJ administration significantly improved novel object recognition in a scopolamine-induced amnesia mouse model. Conclusions : These findings revealed that HJ has a beneficial effect on cognitive impairment and neuroinflammation induced by systemic inflammation and on amnesia induced by scopolamine in mice.This study was supported by the KRIBB Research Initiative Program of the Republic of Korea (KGS1042221) and the Development of Platform Technology for Innovative Medical Measurements funded by Korea Research Institute of Standards and Science (KRISS-GP2022-2)

    Impact of Gender on the Association of Epicardial Fat Thickness, Obesity, and Circadian Blood Pressure Pattern in Hypertensive Patients

    Get PDF
    This study aimed to investigate the effects of gender on the association between epicardial fat thickness (EFT) and circadian blood pressure (BP) changes in patients with recently diagnosed essential hypertension (EH). A total of 441 patients with EH (male/female: 236/205, mean age: 50.7 ± 13.8) and 83 control patients underwent 24-hour ambulatory BP monitoring and echocardiography. Obese EH patients had higher circadian BP profile with BP variability, wall thickness, and left ventricular mass than nonobese EH patients and controls (all p’s <0.05) without gender differences. EFT was higher in female than in male patients (7.0 ± 2.5 versus 5.9 ± 2.2 mm, p<0.001) and higher in the obese female EH group (7.5 ± 2.6 mm) than in the control (6.4 ± 2.8 mm) or nonobese EH group (6.7 ± 2.8 mm) among women, whereas EFT did not vary among males (5.9 ± 1.9 versus 6.0 ± 2.7 versus 5.9 ± 2.4 mm, p=0.937). Multivariate logistic regression analysis demonstrated that the 24-hour mean BP variability was associated with SBP (p=0.018) and EFT (p=0.016) in female patients, but not in male patients. The relationships among circadian BP variability, obesity, and EFT were affected by gender in different manners. EFT may be a more valuable parameter in the evaluation of BP severity and obesity in women than in men

    Label-free fluorescent real-time monitoring of adenylyl cyclase

    Full text link
    In cellular signaling, adenylyl cyclase plays a key role in the hydrolysis of ATP to cyclic AMP and pyrophosphate. Using a synthetic fluorescent chemosensor (PyDPA) which binds strongly to the pyrophosphate group, we have developed a label-free fluorescent real-time detection system for adenylyl cyclase. This assay would be the first adenylyl cyclase assay based on chemosensing the production of pyrophosphate. (C) 2009 Elsevier Ltd. All rights reservedclose2

    Comparative Analysis of Human and Porcine derived Pancreatic dECM using hiPSCs-derived IPCs and LC-MS/MS

    Full text link
    To recapitulate the microenvironment of the target tissue in the 3D printed tissue construct, the selection of bioink is critical. In previous study, we suggested decellularized extracellular matrix (dECM) bioink as an appropriate material for mimicking the native microenvironment. However, since the source of dECM is generally derived from porcine, it is questionable whether porcine tissue-derived dECM bioink can completely mimic the function of human tissue. In this study, we investigated the differences between human and porcine-derived dECM through a variety of methods to validate that porcine derived dECM provide a suitable microenvironmental cue as for cellular activities, particularly for pancreatic tissue. To evaluate the composition of ECM, we quantified major ECM components before and after decellularization using various biochemical assays. In human and porcine dECM bioinks, the quantity of major components such as collagen and GAGs were observed in similar level. In addition, we conducted comparative analysis of representative components of human and porcine tissue-derived pancreatic dECM using liquid chromatography-mass spectrometry and immunofluorescence staining. The same types of collagen occupied the largest portion of both ECM in common, and other components also appeared in a similar ratio. In addition, we examined the differences of cell-matrix interactions by culturing human-induced pluripotent stem cells (hiPSCs)-derived insulin-producing cells (IPCs) in human and porcine pancreatic dECM (pdECM) bioinks. To assess the effects of the pdECM on cellular function, insulin secretion and gene expression level of IPCs encapsulated in both pdECM bioinks were conducted. These data confirmed that porcine-derived material can also provide beneficial effect under optimized microenvironment condition similar to human tissue. The developed pdECM bioink will be able to broaden the application of in vitro disease models of diabetes and pancreatic cancer and transplantable constructs for in vivo study.2

    Comparison of Human and Porcine Pancreatic Decellularzied Extracellular Matrix Bioink using Proteomic Approaches

    Full text link
    To promote various functions of 3D printed tissues, the printable bioink should be selected carefully because it can optimally recapitulate the microenvironment of the target tissue. Decellularized extracellular matrix (dECM) is known as very suitable material to recreate the physiological 3D microenvironment that can maintain the cellular organization and provide appropriate biochemical cues for functional tissue remodeling. Although human tissue can be extracted or get a little amount from tissue bank, the accessibility is pretty much lower than porcine one. Here, we identified the compositional differences between the human and the pig tissuederived dECM bioink. To evaluate whether the pig-derived dECM bioink can provide the suitable microenvironmental cue for 3D printed functional tissues, we showed comparison of the dECM components using LC-MS/MS and gene expression level from the human insulin producing cells encapsulated in human and pig tissue-derived dECM.1

    Bioprinting of insulin-producing multicellular aggregates with the pancreatic tissue-derived bioink

    Full text link
    Bioprinting technology enables directly incorporating multiple cells into complex 3D geometries by rapid and precise patterning to implement physiologically relevant architecture. In the pancreatic tissue, native islets are composed of diverse endocrine cell types and connected by vascular networks to maintain glucose homeostasis. In this regard, we fabricated a 3D human pancreatic tissue model using human embryonic stem cells (hESC)-derived insulin producing cells (IPC), human mesenchymal stem cells (hMSC), and human umbilical vein endothelial cells (HUVEC) with 3D aggregate printing approach to emulate native pancreatic islet-like structure and function. Development of bioink as a functional building block, which can support robust cell differentiation and proliferation, is a critical step towards creation of engineered tissue constructs. In previous study, we proposed that pancreatic tissue derived-decellularized extracellular matrix (pdECM) bioink is compatible with advanced pancreatic tissue engineering as encapsulated islets in pdECM bioink revealed functional stability in glucose responsiveness over typical bioink. Here, we organized the list of major components in the pdECM bioink through gene ontology and proteomic analysis to assess whether tissue-derived bioink can provide sufficient pancreatic cell niche. The representative protein of pdECM bioink was collagen type VI, and other important ECM proteins were also abundant compared to that of the collagen bioink. Moreover, differentiation of hESC into IPC using four-stage protocol was performed to generate functional human pancreatic cells and differentiated cells were characterized by gene expression profile and flow cytometry analysis. As a result, beta cell-specific markers including NKX6.1, PDX1, and insulin exhibited high expression level. After generation of IPC, spatial organization of IPC aggregates via 3D bioprinting were validated for recapitulating native pancreatic tissue geometry. Rapid induction of cellular networks between IPC aggregates comprising IPC, HUVEC, and hMSC was observed after 2 days of printing. Future efforts on functional tests will be able to improve the established 3D human pancreatic tissue model, expanding the application of in vitro and in vivo studies for diabetes research.1

    3D Bioprinting of Insulin-Producing Cell Aggregates-Derived from Human Pluripotent Stem Cells with Pancreatic Tissue-Derived Bioink

    Full text link
    Native pancreatic islets are clustered with diverse endocrine cells as functional units and surrounded by microvasculature networks communicating with neighboring cells for releasing hormones in response to glucose stimulation. 3D bioprinting technology has emerged as an promising strategy for the fabrication of engineered pancreatic tissue constructs because it enables implementation of complex tissue microarchitecture by precise positioning of multiple cells within functional bioink. In this study, we fabricated a pancreatic tissue construct through 3D aggregate bioprinting method using tissue-derived bioink with stem cell-derived insulin producing cells (IPC) to replicate the structural and functional features of native human pancreatic tissue. Selection of suitable bioink is a first step towards building functional 3D pancreatic tissue constructs. In previous study, we suggested pancreatic tissue derived-decellularized extracellular matrix (pdECM) as a attractive printable material to recapitulate native pancreatic cell niche. Here, we investigated representative constituents of pdECM bioink through proteomic analysis to evaluate that pdECM bioink can provide sufficient microenviromental cues. Additionally, functional gene classification regarding matrix-mediated characteristics were observed via gene ontology (GO) analysis. Collagen type VI was most abundant protein in pdECM bioink and other crucial ECM proteins for cell-matrix interactions were also enriched compared to the collagen bioink. Furthermore, we differentiated human embryonic stem cells (hESC) into IPC via four-stage protocol to generate functional human pancreatic cells. The differentiated cells we obtained at each stage were characterized by gene expression profile and flow cytometry analysis. Key markers of beta cells including PDX1, NKX6.1, and insulin were highly expressed after stage 3. After generation of IPC, printing conditions regarding the size of IPC aggregates were optimized for mimicking native pancreatic tissue geometry. In addition, we demonstrated interaction capacity between IPC aggregates with co-culture condition using human umbilical vein endothelial cells (HUVEC) and human mesenchymal stem cells (hMSC). Immunofluorescent staining results of printed constructs revealed that rapid induction of IPC aggregates networks can occur in tri-culture condition. The developed 3D human pancreatic tissue constructs will be able to broaden the application of in vitro disease models of diabetes and transplantable constructs for in vivo study.2

    Comparative Analysis of Human and Porcine derived Pancreatic Decellularized Extracellular Matrix Bioinks to Recapitulate Microenvironment of Human Pancreatic Tissue in vitro

    Full text link
    To recapitulate the microenvironment of the target tissue in the 3D printed tissue construct, the selection of bioink is critical. In previous study, we suggested decellularized extracellular matrix (dECM) bioink as an appropriate material for mimicking the native microenvironment. However, since the source of dECM is generally derived from porcine, it is questionable whether porcine tissue-derived dECM bioink can completely mimic the function of human tissue. In this study, we investigated the differences between human and porcine-derived dECM through a variety of methods to validate that porcine derived dECM provide a suitable microenvironmental cue as for cellular activities, particularly for pancreatic tissue. To evaluate the composition of ECM, we quantified major ECM components before and after decellularization using various biochemical assays. In human and porcine dECM bioinks, the quantity of major components such as collagen and GAGs were observed in similar level. In addition, we conducted comparative analysis of representative components of human and porcine tissue-derived pancreatic dECM using liquid chromatography-mass spectrometry and immunofluorescence staining. The same types of collagen occupied the largest portion of both ECM in common, and other components also appeared in a similar ratio. In addition, we examined the differences of cell-matrix interactions by culturing human-induced pluripotent stem cells (hiPSCs)-derived insulin-producing cells (IPCs) in human and porcine pancreatic dECM (pdECM) bioinks. To assess the effects of the pdECM on cellular function, insulin secretion and gene expression level of IPCs encapsulated in both pdECM bioinks were conducted. These data confirmed that porcine-derived material can also provide beneficial effect under optimized microenvironment condition similar to human tissue. The developed pdECM bioink will be able to broaden the application of in vitro disease models of diabetes and pancreatic cancer and transplantable constructs for in vivo study.1
    corecore