Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Int J Stem Cells ; 17(2): 158-181, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38777830

ABSTRACT

This study offers a comprehensive overview of brain organoids for researchers. It combines expert opinions with technical summaries on organoid definitions, characteristics, culture methods, and quality control. This approach aims to enhance the utilization of brain organoids in research. Brain organoids, as three-dimensional human cell models mimicking the nervous system, hold immense promise for studying the human brain. They offer advantages over traditional methods, replicating anatomical structures, physiological features, and complex neuronal networks. Additionally, brain organoids can model nervous system development and interactions between cell types and the microenvironment. By providing a foundation for utilizing the most human-relevant tissue models, this work empowers researchers to overcome limitations of two-dimensional cultures and conduct advanced disease modeling research.

2.
Elife ; 122023 Nov 14.
Article in English | MEDLINE | ID: mdl-37963071

ABSTRACT

In vitro culture systems that structurally model human myogenesis and promote PAX7+ myogenic progenitor maturation have not been established. Here we report that human skeletal muscle organoids can be differentiated from induced pluripotent stem cell lines to contain paraxial mesoderm and neuromesodermal progenitors and develop into organized structures reassembling neural plate border and dermomyotome. Culture conditions instigate neural lineage arrest and promote fetal hypaxial myogenesis toward limb axial anatomical identity, with generation of sustainable uncommitted PAX7 myogenic progenitors and fibroadipogenic (PDGFRa+) progenitor populations equivalent to those from the second trimester of human gestation. Single-cell comparison to human fetal and adult myogenic progenitor /satellite cells reveals distinct molecular signatures for non-dividing myogenic progenitors in activated (CD44High/CD98+/MYOD1+) and dormant (PAX7High/FBN1High/SPRY1High) states. Our approach provides a robust 3D in vitro developmental system for investigating muscle tissue morphogenesis and homeostasis.


Humans contains around 650 skeletal muscles which allow the body to move around and maintain its posture. Skeletal muscles are made up of individual cells that bundle together into highly organized structures. If this group of muscles fail to develop correctly in the embryo and/or fetus, this can lead to muscular disorders that can make it painful and difficult to move. One way to better understand how skeletal muscles are formed, and how this process can go wrong, is to grow them in the laboratory. This can be achieved using induced pluripotent stem cells (iPSCs), human adult cells that have been 'reprogrammed' to behave like cells in the embryo that can develop in to almost any cell in the body. The iPSCs can then be converted into specific cell types in the laboratory, including the cells that make up skeletal muscle. Here, Mavrommatis et al. created a protocol for developing iPSCs into three-dimensional organoids which resemble how cells of the skeletal muscle look and arrange themselves in the fetus. To form the skeletal muscle organoid, Mavrommatis et al. treated iPSCs that were growing in a three-dimensional environment with various factors that are found early on in development. This caused the iPSCs to organize themselves in to embryonic and fetal structures that will eventually give rise to the parts of the body that contain skeletal muscle, such as the limbs. Within the organoid were cells that produced Pax7, a protein commonly found in myogenic progenitors that specifically mature into skeletal muscle cells in the fetus. Pax 7 is also present in 'satellite cells' that help to regrow damaged skeletal muscle in adults. Indeed, Mavrommatis et al. found that the myogenic progenitors produced by the organoid were able to regenerate muscle when transplanted in to adult mice. These findings suggest that this organoid protocol can generate cells that will give rise to skeletal muscle. In the future, these lab-grown progenitors could potentially be created from cells isolated from patients and used to repair muscle injuries. The organoid model could also provide new insights in to how skeletal muscles develop in the fetus, and how genetic mutations linked with muscular disorders disrupt this process.


Subject(s)
Muscle, Skeletal , Satellite Cells, Skeletal Muscle , Humans , Muscle, Skeletal/metabolism , Cell Differentiation , Fetus/metabolism , Satellite Cells, Skeletal Muscle/physiology , Muscle Development/physiology , PAX7 Transcription Factor/metabolism
3.
Nanomaterials (Basel) ; 13(16)2023 Aug 12.
Article in English | MEDLINE | ID: mdl-37630909

ABSTRACT

This paper presents a study that aims to enhance the performance of quantum dot light-emitting didoes (QLEDs) by employing a solution-processed molybdenum oxide (MoOx) nanoparticle (NP) as a hole injection layer (HIL). The study investigates the impact of varying the concentrations of the MoOx NP layer on device characteristics and delves into the underlying mechanisms that contribute to the observed enhancements. Experimental techniques such as an X-ray diffraction and field-emission transmission electron microscopy were employed to confirm the formation of MoOx NPs during the synthesis process. Ultraviolet photoelectron spectroscopy was employed to analyze the electron structure of the QLEDs. Remarkable enhancements in device performance were achieved for the QLED by employing an 8 mg/mL concentration of MoOx nanoparticles. This configuration attains a maximum luminance of 69,240.7 cd/cm2, a maximum current efficiency of 56.0 cd/A, and a maximum external quantum efficiency (EQE) of 13.2%. The obtained results signify notable progress in comparison to those for QLED without HIL, and studies that utilize the widely used poly(3,4-ethylenedioxythiophene):poly(styrene sulfonate) (PEDOT:PSS) HIL. They exhibit a remarkable enhancements of 59.5% and 26.4% in maximum current efficiency, respectively, as well as significant improvements of 42.7% and 20.0% in maximum EQE, respectively. This study opens up new possibilities for the selection of HIL and the fabrication of solution-processed QLEDs, contributing to the potential commercialization of these devices in the future.

4.
Front Biosci (Landmark Ed) ; 28(3): 48, 2023 03 13.
Article in English | MEDLINE | ID: mdl-37005763

ABSTRACT

BACKGROUND: Disinfection byproducts (DBPs) cause endocrine disruption via estrogenic or anti-estrogenic effects on estrogen receptors. However, most studies have focused on human systems, with little experimental data being presented on aquatic biota. This study aimed to compare the effects of nine DBPs on zebrafish and human estrogen receptor alpha (zERα and hERα). METHODS: In vitro enzyme response-based tests, including cytotoxicity and reporter gene assays, were performed. Additionally, statistical analysis and molecular docking studies were employed to compare ERα responses. RESULTS: Iodoacetic acid (IAA), chloroacetonitrile (CAN), and bromoacetonitrile (BAN) showed robust estrogenic activity on hERα(maximal induction ratios of 108.7%, 50.3%, and 54.7%, respectively), while IAA strongly inhibited the estrogenic activity induced by 17ß-estradiol (E2) in zERα (59.8% induction at the maximum concentration). Chloroacetamide (CAM) and bromoacetamide (BAM) also showed robust anti-estrogen effects in zERα (48.1% and 50.8% induction at the maximum concentration, respectively). These dissimilar endocrine disruption patterns were thoroughly assessed using Pearson correlation and distance-based analyses. Clear differences between the estrogenic responses of the two ERαs were observed, whereas no pattern of anti-estrogenic activities could be established. Some DBPs strongly induced estrogenic endocrine disruption as agonists of hERα, while others inhibited estrogenic activity as antagonists of zERα. Principal coordinate analysis (PCoA) showed similar correlation coefficients for estrogenic and anti-estrogenic responses. Reproducible results were obtained from computational analysis and the reporter gene assay. CONCLUSIONS: Overall, the effects of DBPs on both human and zebrafish highlight the importance of controlling their differences in responsiveness for estrogenic activities including the water quality monitoring and endocrine disruption, as DBPs have species-specific ligand-receptor interactions.


Subject(s)
Estrogen Receptor alpha , Zebrafish , Animals , Humans , Estrogen Receptor alpha/genetics , Disinfection , Molecular Docking Simulation , Estrogens/pharmacology , Receptors, Estrogen/genetics
5.
Nat Commun ; 14(1): 1488, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36932093

ABSTRACT

Reconstruction of skin equivalents with physiologically relevant cellular and matrix architecture is indispensable for basic research and industrial applications. As skin-nerve crosstalk is increasingly recognized as a major element of skin physiological pathology, the development of reliable in vitro models to evaluate the selective communication between epidermal keratinocytes and sensory neurons is being demanded. In this study, we present a three-dimensional innervated epidermal keratinocyte layer as a sensory neuron-epidermal keratinocyte co-culture model on a microfluidic chip using the slope-based air-liquid interfacing culture and spatial compartmentalization. Our co-culture model recapitulates a more organized basal-suprabasal stratification, enhanced barrier function, and physiologically relevant anatomical innervation and demonstrated the feasibility of in situ imaging and functional analysis in a cell-type-specific manner, thereby improving the structural and functional limitations of previous coculture models. This system has the potential as an improved surrogate model and platform for biomedical and pharmaceutical research.


Subject(s)
Epidermis , Microfluidics , Coculture Techniques , Epidermis/innervation , Keratinocytes , Skin , Sensory Receptor Cells , Cells, Cultured
6.
Small ; 19(25): e2300236, 2023 06.
Article in English | MEDLINE | ID: mdl-36932895

ABSTRACT

The disruption of thyroid hormones because of chemical exposure is a significant societal problem. Chemical evaluations of environmental and human health risks are conventionally based on animal experiments. However, owing to recent breakthroughs in biotechnology, the potential toxicity of chemicals can now be evaluated using 3D cell cultures. In this study, the interactive effects of thyroid-friendly soft (TS) microspheres on thyroid cell aggregates are elucidated and their potential as a reliable toxicity assessment tool is evaluated. Using state-of-the-art characterization methods coupled with cell-based analysis and quadrupole time-of-flight mass spectrometry, it is shown that TS-microsphere-integrated thyroid cell aggregates exhibit improved thyroid function. Specifically, the responses of zebrafish embryos, which are used for thyroid toxicity analysis, and the TS-microsphere-integrated cell aggregates to methimazole (MMI), a known thyroid inhibitor, are compared. The results show that the thyroid hormone disruption response of the TS-microsphere-integrated thyroid cell aggregates to MMI is more sensitive compared with those of the zebrafish embryos and conventionally formed cell aggregates. This proof-of-concept approach can be used to control cellular function in the desired direction and hence evaluate thyroid function. Thus, the proposed TS-microsphere-integrated cell aggregates may yield new fundamental insights for advancing in vitro cell-based research.


Subject(s)
Thyroid Gland , Zebrafish , Animals , Humans , Antithyroid Agents/pharmacology , Thyroid Hormones/pharmacology , Methimazole/toxicity
7.
Exp Mol Med ; 55(3): 653-664, 2023 03.
Article in English | MEDLINE | ID: mdl-36941461

ABSTRACT

We do not yet understand exactly how corticosteroids attenuate hyperinflammatory responses and alleviate high-risk coronavirus disease 2019 (COVID-19). We aimed to reveal the molecular mechanisms of hyperinflammation in COVID-19 and the anti-inflammatory effects of corticosteroids in patients with high-risk COVID-19. We performed single-cell RNA sequencing of peripheral blood mononuclear cells (PBMCs) from three independent COVID-19 cohorts: cohort 1 was used for comparative analysis of high-risk and low-risk COVID-19 (47 PBMC samples from 28 patients), cohort 2 for longitudinal analysis during COVID-19 (57 PBMC samples from 15 patients), and cohort 3 for investigating the effects of corticosteroid treatment in patients with high-risk COVID-19 (55 PBMC samples from 13 patients). PBMC samples from healthy donors (12 PBMC samples from 12 donors) were also included. Cohort 1 revealed a significant increase in the proportion of monocytes expressing the long noncoding RNAs NEAT1 and MALAT1 in high-risk patients. Cohort 2 showed that genes encoding inflammatory chemokines and their receptors were upregulated during aggravation, whereas genes related to angiogenesis were upregulated during improvement. Cohort 3 demonstrated downregulation of interferon-stimulated genes (ISGs), including STAT1, in monocytes after corticosteroid treatment. In particular, unphosphorylated STAT-dependent ISGs enriched in monocytes from lupus patients were selectively downregulated by corticosteroid treatment in patients with high-risk COVID-19. Corticosteroid treatment suppresses pathologic interferon responses in monocytes by downregulating STAT1 in patients with high-risk COVID-19. Our study provides insights into the mechanisms underlying COVID-19 aggravation and improvement and the effects of corticosteroid treatment.


Subject(s)
COVID-19 , Leukocytes, Mononuclear , Humans , Leukocytes, Mononuclear/metabolism , Interferons , Monocytes/metabolism , Adrenal Cortex Hormones/pharmacology , Adrenal Cortex Hormones/therapeutic use , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism
8.
Materials (Basel) ; 16(2)2023 Jan 08.
Article in English | MEDLINE | ID: mdl-36676338

ABSTRACT

Highly efficient and all-solution processed quantum dot light-emitting diodes (QLEDs) with high performance are demonstrated by employing ZnMgO nanoparticles (NPs) with core/shell structure used as an electron transport layer (ETL). Mg-doping in ZnO NPs exhibits a different electronic structure and degree of electron mobility. A key processing step for synthesizing ZnMgO NPs with core/shell structure is adding Mg in the solution in addition to the remaining Mg and Zn ions after the core formation process. This enhanced Mg content in the shell layer compared with that of the core X-ray photoelectron spectroscopy showed a higher number of oxygen vacancies for the ZnMgO core/shell structure, thereby enhancing the charge balance in the emitting layer and improving device efficiency. The QLED incorporating the as synthesized ZnMgO NP core/shell A exhibited a maximum luminance of 55,137.3 cd/m2, maximum current efficiency of 58.0 cd/A and power efficiency of 23.3 lm/W. The maximum current efficiency and power efficiency of the QLED with ZnMgO NP core/shell A improved by as much as 156.3% and 113.8%, respectively, compared to the QLED with a Zn0.9Mg0.1O NP ETL, thus demonstrating the benefits of ZnMgO NPs with the specified core/shell structure.

9.
Sci Rep ; 12(1): 11449, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35794197

ABSTRACT

The protozoan parasite Toxoplasma gondii (T. gondii) causes one of the most common human zoonotic diseases and infects approximately one-third of the global population. T. gondii infects nearly every cell type and causes severe symptoms in susceptible populations. In previous laboratory animal studies, T. gondii movement and transmission were not analyzed in real time. In a three-dimensional (3D) microfluidic assay, we successfully supported the complex lytic cycle of T. gondii in situ by generating a stable microvasculature. The physiology of the T. gondii-infected microvasculature was monitored in order to investigate the growth, paracellular and transcellular migration, and transmission of T. gondii, as well as the efficacy of T. gondii drugs.


Subject(s)
Toxoplasma , Toxoplasmosis , Animals , Microfluidics , Toxoplasma/physiology , Toxoplasmosis/parasitology , Transendothelial and Transepithelial Migration , Zoonoses
10.
Front Bioeng Biotechnol ; 9: 697657, 2021.
Article in English | MEDLINE | ID: mdl-34671596

ABSTRACT

Lymphangiogenesis is a stage of new lymphatic vessel formation in development and pathology, such as inflammation and tumor metastasis. Physiologically relevant models of lymphatic vessels have been in demand because studies on lymphatic vessels are required for understanding the mechanism of tumor metastasis. In this study, a new three-dimensional lymphangiogenesis model in a tumor microenvironment is proposed, using a newly designed macrofluidic platform. It is verified that controllable biochemical and biomechanical cues, which contribute to lymphangiogenesis, can be applied in this platform. In particular, this model demonstrates that a reconstituted lymphatic vessel has an in vivo-like lymphatic vessel in both physical and biochemical aspects. Since biomechanical stress with a biochemical factor influences robust directional lymphatic sprouting, whether our model closely approximates in vivo, the initial lymphatics in terms of the morphological and genetic signatures is investigated. Furthermore, attempting an incorporation with a tumor spheroid, this study successfully develops a complex tumor microenvironment model for use in lymphangiogenesis and reveals the microenvironment factors that contribute to tumor metastasis. As a first attempt at a coculture model, this reconstituted model is a novel system with a fully three-dimensional structure and can be a powerful tool for pathological drug screening or disease model.

11.
Sci Rep ; 11(1): 18230, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34521931

ABSTRACT

During radiotherapy, microenvironments neighboring the tumor are also exposed to gamma irradiation; this results in unexpected side effects. Blood vessels can serve as microenvironments for tumors and they play an important role in providing nutrients to tumors. This is mostly related to tumor progression, metastasis, and relapse after therapy. Many studies have been performed to obtain a better understanding of tumor vasculature after radiotherapy with in vitro models. However, compared to 3-D models, 2-D in vitro endothelial monolayers cannot physiologically reflect in vivo blood vessels. We previously remodeled the extracellular matrix (ECM) hydrogel that enhanced the tight barrier formation of 3-D blood vessels and the vascular endothelial growth factor (VEGF) gradient induced angiogenesis in a microfluidic device. In this study, the blood vessel model is further introduced to understand how gamma irradiation affects the endothelial monolayer. After the gamma irradiation exposure, we observed a collapsed endothelial barrier and a reduced angiogenic potential. Changes in the cell behaviors of the tip and stalk cells were also detected in the angiogenesis model after irradiation, which is difficult to observe in 2-D monolayer models. Therefore, the 3-D in vitro blood vessel model can be used to understand radiation-induced endothelial injuries.


Subject(s)
Endothelial Cells/radiation effects , Gamma Rays , Neovascularization, Pathologic/metabolism , Tissue Engineering/methods , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Extracellular Matrix/chemistry , Humans , Hydrogels/chemistry , Microfluidics/methods , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism
12.
Biomaterials ; 276: 121004, 2021 09.
Article in English | MEDLINE | ID: mdl-34273688

ABSTRACT

As the importance of organoids increases, the need to develop organoid culture systems suitable for basic biological and clinical applications is being emphasized. However, there is still an unmet need to produce functionally complex and scalable uniform organoids. Here, we demonstrate a scalable organoid production platform with 8 well strips and a total of 8 × 9 microwells per strip using organoids derived from colorectal cancer tissue. The new culture platform is a format in which single cells are self-organized into organoids in culture medium supplemented with 2% Matrigel. It is functionally compatible with existing 96 well plates and Matrigel based conventional organoid culture methods. The consistency, uniformity and reproducibility of organoid produced on the new platform have been significantly improved compared to those of conventional plates. Importantly, Hydro-organoids are functionally identical to conventional Matrigel organoids, but show better consistency in drug screening. Our results show the possibility that Hydro-organoids can be used in high-throughput assays and incorporated into drug screening models to predict clinical outcomes.


Subject(s)
Colorectal Neoplasms , Organoids , Colorectal Neoplasms/drug therapy , Drug Evaluation, Preclinical , Early Detection of Cancer , Humans , Reproducibility of Results
13.
Biomaterials ; 269: 120529, 2021 02.
Article in English | MEDLINE | ID: mdl-33257114

ABSTRACT

Recent advances in pluripotent stem cell technology provide an alternative source of human hepatocytes to overcome the limitations of current toxicity tests. However, this approach requires optimization and standardization before it can be used as a fast and reliable toxicity screening system. Here, we designed and tested microwell culture platforms with various diameters. We found that large quantities of uniformly-sized hepatocyte-like cell (HLC) spheroids (3D-uniHLC-Ss) could be efficiently and reproducibly generated in a short period time from a small number of differentiating human pluripotent stem cells (hPSCs). The hPSC-3D-uniHLC-Ss that were produced in 500-µm diameter microwells consistently exhibited high expressions of hepatic marker genes and had no significant signs of cell death. Importantly, a hepatic master gene hepatocyte nuclear factor 4α (HNF4α) was maintained at high levels, and the epithelial-mesenchymal transition was significantly attenuated in hPSC-3D-uniHLC-Ss. Additionally, when compared with 3D-HLC-Ss that were produced in other 3D platforms, hPSC-3D-uniHLC-Ss showed significantly higher hepatic gene expressions and drug-metabolizing activity of the enzyme, CYP3A4. Imaging-based drug toxicity studies demonstrated that hPSC-3D-uniHLC-Ss exhibited enhanced sensitivity to various hepatotoxicants, compared to HLCs, which were differentiated under 2D conditions. Precise prediction of drug-induced hepatotoxicity is a crucial step in the early phases of drug discovery. Thus, the hPSC-3D-uniHLC-Ss produced using our microwell platform could be used as an imaging-based toxicity screening system to predict drug hepatotoxicity.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Pluripotent Stem Cells , Cell Culture Techniques , Cell Differentiation , Hepatocytes , Humans , Liver
14.
Sci Adv ; 5(11): eaax4520, 2019 11.
Article in English | MEDLINE | ID: mdl-31807701

ABSTRACT

Native pancreatic islets interact with neighboring cells by establishing three-dimensional (3D) structures, and are surrounded by perfusion at an interstitial flow level. However, flow effects are generally ignored in islet culture models, although cell perfusion is known to improve the cell microenvironment and to mimic in vivo physiology better than static culture systems. Here, we have developed functional islet spheroids using a microfluidic chip that mimics interstitial flow conditions with reduced shear cell damage. Dynamic culture, compared to static culture, enhanced islet health and maintenance of islet endothelial cells, reconstituting the main component of islet extracellular matrix within spheroids. Optimized flow condition allowed localization of secreted soluble factors near spheroids, facilitating diffusion-mediated paracrine interactions within islets, and enabled long-term maintenance of islet morphology and function for a month. The proposed model can aid islet preconditioning before transplantation and has potential applications as an in vitro model for diabetic drug testing.


Subject(s)
Islets of Langerhans/metabolism , Lab-On-A-Chip Devices , Models, Biological , Spheroids, Cellular/metabolism , Animals , Cell Culture Techniques , Drug Evaluation , Hypoglycemic Agents/pharmacology , Islets of Langerhans/cytology , Islets of Langerhans Transplantation , Male , Rats , Rats, Sprague-Dawley , Spheroids, Cellular/cytology
15.
J Biol Chem ; 291(27): 14199-14212, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27189941

ABSTRACT

The viral vector-mediated overexpression of the defined transcription factors, Brn4/Pou3f4, Sox2, Klf4, and c-Myc (BSKM), could induce the direct conversion of somatic fibroblasts into induced neural stem cells (iNSCs). However, viral vectors may be randomly integrated into the host genome thereby increasing the risk for undesired genotoxicity, mutagenesis, and tumor formation. Here we describe the generation of integration-free iNSCs from mouse fibroblasts by non-viral episomal vectors containing BSKM. The episomal vector-derived iNSCs (e-iNSCs) closely resemble control NSCs, and iNSCs generated by retrovirus (r-iNSCs) in morphology, gene expression profile, epigenetic status, and self-renewal capacity. The e-iNSCs are functionally mature, as they could differentiate into all the neuronal cell types both in vitro and in vivo Our study provides a novel concept for generating functional iNSCs using a non-viral, non-integrating, plasmid-based system that could facilitate their biomedical applicability.


Subject(s)
Neural Stem Cells/cytology , Animals , Fibroblasts/cytology , Genetic Vectors , Kruppel-Like Factor 4 , Mice , Mice, Inbred C3H , Transfection
16.
Cell Stem Cell ; 10(4): 465-72, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22445517

ABSTRACT

Recent studies have shown that defined sets of transcription factors can directly reprogram differentiated somatic cells to a different differentiated cell type without passing through a pluripotent state, but the restricted proliferative and lineage potential of the resulting cells limits the scope of their potential applications. Here we show that a combination of transcription factors (Brn4/Pou3f4, Sox2, Klf4, c-Myc, plus E47/Tcf3) induces mouse fibroblasts to directly acquire a neural stem cell identity-which we term as induced neural stem cells (iNSCs). Direct reprogramming of fibroblasts into iNSCs is a gradual process in which the donor transcriptional program is silenced over time. iNSCs exhibit cell morphology, gene expression, epigenetic features, differentiation potential, and self-renewing capacity, as well as in vitro and in vivo functionality similar to those of wild-type NSCs. We conclude that differentiated cells can be reprogrammed directly into specific somatic stem cell types by defined sets of specific transcription factors.


Subject(s)
Cell Dedifferentiation , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Transcription Factors/biosynthesis , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Fibroblasts/cytology , Gene Expression Regulation/genetics , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Mice , Neural Stem Cells/cytology , Transcription Factors/genetics
17.
Biochem Biophys Res Commun ; 390(3): 763-8, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19836348

ABSTRACT

The pathogenesis of post-transplant diabetes mellitus (PTDM) is thought to be partly related to the direct toxic effect of cyclosporine (CsA) on pancreatic beta-cells and the resultant decrease in insulin synthesis and secretion. Although rosiglitazone (Rosi) is an insulin sensitizer, recent data has shown that Rosi also directly protects against beta-cell dysfunction and death. This study was undertaken to clarify the effects of Rosi on CsA-induced beta-cell dysfunction and death. The deterioration in glucose tolerance caused by CsA administration was significantly improved by cotreatment with Rosi. The relative volume and absolute mass of beta-cells were significantly reduced by CsA, whereas combined treatment with Rosi had protective effects. Induction of beta-cell death and increased expression of endoplasmic reticulum (ER) stress markers (CHOP and spliced XBP-1) by CsA were rescued by Rosi. Thus, Rosi signaling directly modulates the ER stress response, promoting beta-cell adaptation and survival. Rosi might be an appropriate drug for preventing and treating CsA-induced PTDM.


Subject(s)
Cyclosporine/antagonists & inhibitors , Cytoprotection , Diabetes Mellitus/prevention & control , Hypoglycemic Agents/pharmacology , Immunosuppressive Agents/antagonists & inhibitors , Insulin-Secreting Cells/drug effects , Animals , Cyclosporine/adverse effects , Cyclosporine/toxicity , Diabetes Mellitus/chemically induced , Glucose Tolerance Test , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/toxicity , Insulin/blood , Insulin-Secreting Cells/pathology , Male , Rats , Rats, Sprague-Dawley , Rosiglitazone , Thiazolidinediones , Transplantation/adverse effects
18.
Am J Transplant ; 5(8): 1856-67, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15996232

ABSTRACT

Rosiglitazone (RGTZ) has protective effect against various types of injury. This study was performed to evaluate the effect of RGTZ on pancreatic and renal injury caused by cyclosporine (CsA). CsA (15 mg/kg) and RGTZ (3 mg/kg) were administered alone and together to the rats for 28 days. The effect of RGTZ on CsA-induced pancreatic injury was evaluated by intraperitoneal glucose tolerance test (IPGTT), plasma insulin concentrations and pancreatic beta-cell morphology. The effect of RGTZ on CsA-induced renal injury was evaluated by assessing renal function and pathology; mediators of inflammation and fibrosis such as angiotensin II (AngII), osteopontin (OPN) and transforming growth factor-beta1 (TGF-beta1) and apoptotic cell death. Four weeks of CsA treatment caused diabetes, renal dysfunction, typical pathologic lesions (arteriolopathy, interstitial fibrosis and inflammatory cells infiltration) and apoptotic cell death. RGTZ treatment decreased blood glucose concentration, increased plasma insulin concentration and preserved pancreatic beta islet mass. RGTZ treatment improved renal function and histopathology. Pro-inflammatory and pro-fibrotic molecules such as AngII, OPN and TGF-beta1, and apoptotic cell death also decreased with RGTZ treatment. These data suggest that RGTZ has a protective effect against CsA-induced pancreatic and renal injury.


Subject(s)
Cyclosporine/toxicity , Diabetes Mellitus, Experimental/prevention & control , Hypoglycemic Agents/therapeutic use , Immunosuppressive Agents/toxicity , Islets of Langerhans/drug effects , Kidney/drug effects , Pancreatic Diseases/prevention & control , Thiazolidinediones/therapeutic use , Angiotensin II/genetics , Angiotensin II/metabolism , Animals , Apoptosis , Cyclosporine/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Glucose Tolerance Test , Immunosuppressive Agents/blood , Insulin/blood , Osteopontin , Pancreatic Diseases/chemically induced , Pancreatic Diseases/pathology , Rats , Rats, Sprague-Dawley , Rosiglitazone , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
SELECTION OF CITATIONS
SEARCH DETAIL
...