Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
J Hematol Oncol ; 17(1): 35, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38764068

ABSTRACT

Angiosarcoma is a rare subtype of malignant neoplasm originating from vascular or lymphatic endothelial cells; its low incidence has posed significant challenges for comprehensive investigations into its pathogenic mechanisms and the development of innovative treatment modalities through in vitro and in vivo models. Recent endeavors spearheaded by patient-partnered research initiatives have aimed to elucidate the intricacies of angiosarcomas by leveraging biological omics approaches, with the overarching objective of enhancing prognostic indicators and therapeutic options for this uncommon pathology. To bridge the gap between preclinical research and translational applications, we engineered angiosarcoma-derived organoids from surgically resected primary tumors, hereafter referred to as "sarconoids," as a proof-of-concept model. A novel protocol for the establishment of these sarconoids has been developed and validated. To ensure that the sarconoids faithfully recapitulate the heterogeneity and complexities of the patients' original tumors, including transcriptomic signatures, cell-type specificity, and morphological traits, exhaustive histological and transcriptomic analyses were conducted. Subsequently, we expanded the scope of our study to include an evaluation of a sarconoid-based drug screening platform; for this purpose, a drug library (AOD IX), supplied by the National Cancer Institute's Developmental Therapeutics Program, was screened using 96-well plates. Our findings suggest that sarconoids can be reliably generated from angiosarcoma patient-derived tissues and can serve as accurate models for evaluating therapeutic responses, thereby holding far-reaching implications for translational research and clinical applications aimed at advancing our understanding and treatment of angiosarcoma.


Subject(s)
Hemangiosarcoma , Hemangiosarcoma/pathology , Hemangiosarcoma/drug therapy , Hemangiosarcoma/therapy , Hemangiosarcoma/genetics , Humans , Organoids/pathology , Organoids/drug effects , Female
2.
Biomed Pharmacother ; 174: 116511, 2024 May.
Article in English | MEDLINE | ID: mdl-38574616

ABSTRACT

Human iPSC-derived cardiac organoids (hiPSC-COs) for cardiotoxicity drug testing via the variety of cell lines and unestablished protocols may lead to differences in response results due to a lack of criteria for generation period and size. To ensure reliable drug testing, it is important for researchers to set optimal generation period and size of COs according to the cell line and protocol applied in their studies. Hence, we sought to propose a process to establish minimum criteria for the generation duration and size of hiPSC-COs for cardiotoxic drug testing. We generated hiPSC-COs of different sizes based on our protocol and continuously monitored organoids until they indicated a minimal beating rate change as a control that could lead to more accurate beating rate changes on drug testing. Calcium transients and physiological tests to assess the functionality of hiPSC-COs on selected generation period, which showed regular cardiac beating, and immunostaining assays to compare characteristics were performed. We explained the generation period and size that exhibited and maintained regular beating rate changes on hiPSC-COs, and lead to reliable response results to cardiotoxicity drugs. We anticipate that this study will offer valuable insights into considering the appropriate generation period and size of hiPSC-COs ensuring reliable outcomes in cardiotoxicity drug testing.


Subject(s)
Cardiotoxicity , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Organoids , Humans , Induced Pluripotent Stem Cells/drug effects , Organoids/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Drug Evaluation, Preclinical/methods
3.
Biofabrication ; 13(4)2021 09 21.
Article in English | MEDLINE | ID: mdl-34496353

ABSTRACT

In the tumor microenvironment (TME), the extracellular matrix (ECM) provides a dynamic structure for cell adhesion and cancer cell motility, such as migration and invasion, as well as remodeling. Matrix metalloproteinases (MMPs) promote cancer cell motility, which contributes to inducing drug resistance and thereby acquiring aggressive features. The drug resistance-induced 3Din vitrotumor model can be an effective model for therapeutic strategies for anticancer drugs targeting aggressive cancer cells. Here, we describe highly drug-resistant multicellular tumoroids (MCTs)-ECM tumor grafts under a macroscale dense 3Din vitromodel through a combination of numerous MCTs and a collagen matrix. MCTs-ECM tumor grafts promote the high activity of MMP2 and MMP9 compared to general MCTs and induced cancer cell motility. Then, after the administration of anticancer drugs, the tumor grafts show increased drug resistance, with both the sporadic distribution of necrotic cells and the reduction of apoptotic portions, by activating cancer cell motility. MCTs-ECM tumor graft could be useful as a macroscale tumor graft model for inducing drug resistance by activating cancer cell motility and evaluating the efficacy of anticancer drugs targeting cancer with aggressive features.


Subject(s)
Extracellular Matrix , Neoplasms , Cell Death , Drug Resistance , Humans , Neoplasms/drug therapy , Tumor Microenvironment
5.
Int J Mol Sci ; 22(3)2021 Jan 29.
Article in English | MEDLINE | ID: mdl-33572899

ABSTRACT

Differential chemo-sensitivity of cancer cells, which is attributed to the cellular heterogeneity and phenotypic variation of cancer cells, is considered to be the main reason for tumor recurrence after chemotherapy. Here, we generated small cell lung cancer patient-derived tumor organoids and subjected them to long-term expansion with the addition of WNT3A or R-spondin1. We confirmed that the organoids have similar genetic profiles, molecular characteristics, and morphological architectures to the corresponding patient tumor tissue during and after long-term expansion. Interestingly, the cellular heterogeneity of organoids is reflected in their differential response to cisplatin or etoposide. We propose to utilize the organoids as small cell lung cancer patient avatar models that would be ideal for investigating the mechanisms underlying tumor recurrence after chemotherapy, and would ultimately help to develop personalized medicine.


Subject(s)
Lung Neoplasms/pathology , Organoids/pathology , Small Cell Lung Carcinoma/pathology , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Screening Assays, Antitumor/methods , Etoposide/pharmacology , Humans , Lung Neoplasms/drug therapy , Organ Culture Techniques/methods , Organoids/drug effects , Small Cell Lung Carcinoma/drug therapy , Tumor Cells, Cultured
6.
Biofabrication ; 12(4): 045035, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33000764

ABSTRACT

A biologically relevant in vitro model of hepatic microtissue would be a valuable tool for the preclinical study of pharmacokinetics and metabolism. Although considerable advances have been made in recent years in the establishment of alternative in vitro culture systems that mimic liver tissue, generating an effective liver model remains challenging. Specifically, existing model systems still exhibit limited functions for hepatocellular differentiation potential and cellular complexity. It is essential to improve the in vitro differentiation of liver progenitor cells (LPCs) for disease modeling and preclinical pharmatoxicological research. Here, we describe a rat liver organoid culture system under in vivo-like steady-state flow conditions; this system is capable of controlling the expansion and differentiation of rat liver organoids over 10-15 d. LPCs cultured in medium flow conditions become self-assembled liver organoids that exhibit phenotypic and functional hepato-biliary modeling. In addition, hepatocytes that are differentiated using liver organoids produced albumin and maintained polygonal morphology, which is characteristic of mature hepatocytes.


Subject(s)
Hepatocytes , Organoids , Animals , Cell Differentiation , Liver , Rats , Stem Cells
7.
PLoS One ; 15(7): e0235356, 2020.
Article in English | MEDLINE | ID: mdl-32628693

ABSTRACT

As a new class of cancer therapeutic agents, oncolytic viruses (OVs) have gained much attention not only due to their ability to selectively replicate in and lyse tumor cells, but also for their potential to stimulate antitumor immune responses. As a result, there is an increasing need for in vitro modeling systems capable of recapitulating the 3D physiological tumor microenvironment. Here, we investigated the potential of our recently developed microphysiological system (MPS), featuring a vessel-like channel to reflect the in vivo tumor microenvironment and serving as culture spaces for 3D multicellular tumor spheroids (MCTSs). The MCTSs consist of cancer A549 cells, stromal MRC5 cells, endothelial HUVECs, as well as the extracellular matrix. 3D MCTSs residing in the MPS were infected with oncolytic VSV expressing GFP (oVSV-GFP). Post-infection, GFP signal intensity increased only in A549 cells of the MPS. On the other hand, HUVECs were susceptible to virus infection under 2D culture and IFN-ß secretion was quite delayed in HUVECs. These results thus demonstrate that OV antitumoral characteristics can be readily monitored in the MPS and that its behavior therein somewhat differs compared to its activity in 2D system. In conclusion, we present the first application of the MPS, an in vitro model that was developed to better reflect in vivo conditions. Its various advantages suggest the 3D MCTS-integrated MPS can serve as a first line monitoring system to validate oncolytic virus efficacy.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Vesiculovirus/immunology , A549 Cells , Cell Culture Techniques/methods , Drug Screening Assays, Antitumor/methods , Extracellular Matrix , Human Umbilical Vein Endothelial Cells , Humans , Neoplasms/immunology , Oncolytic Viruses/genetics , Spheroids, Cellular , Vesiculovirus/genetics
8.
Article in English | MEDLINE | ID: mdl-32565421

ABSTRACT

OBJECTIVE: Esterified collagen (EC) can be functionalized with heparin to enhance islet graft stability. Growth factors secreted by human adipose-derived stem cells (hADSCs) can bind efficiently to EC-heparin (EC-Hep), which enhances revascularization and cell protection. We investigated the therapeutic potential of a combined heparin-esterified collagen-hADSC (HCA)-islet sheet to enhance islet engraftment. RESEARCH DESIGN AND METHODS: This study was designed to assess the efficiency of using EC-Hep as a scaffold for subcutaneous islet transplantation in diabetic athymic mice. After the hADSC-cocultured islets were seeded in the EC-Hep scaffold, islet function was measured by glucose-stimulated insulin secretion test and growth factors in the culture supernatants were detected by protein array. Islet transplantation was performed in mice, and graft function and survival were monitored by measuring the blood glucose levels. ß-Cell mass and vascular densities were assessed by immunohistochemistry. RESULTS: The EC-Hep composite allowed sustained release of growth factors. Secretion of growth factors and islet functionality in the HCA-islet sheet were significantly increased compared with the control groups of islets alone or combined with native collagen. In vivo, stable long-term glucose control by the graft was achieved after subcutaneous transplantation of HCA-islet sheet due to enhanced capillary network formation around the sheet. CONCLUSIONS: The findings indicate the potential of the HCA-islet sheet to enhance islet revascularization and engraftment in a hADSC dose-dependent manner, following clinical islet transplantation for the treatment of diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental , Insulin-Secreting Cells , Animals , Collagen , Diabetes Mellitus, Experimental/therapy , Heparin , Mice , Stem Cells
9.
Adv Biosyst ; 4(2): e1900143, 2020 02.
Article in English | MEDLINE | ID: mdl-32293137

ABSTRACT

Replicable oncolytic viruses (OVs) induce tumor cell lysis and release viral progeny. The released progeny virions and cell debris can spread within surrounding tumor cells or blood vessels. These released molecules may also induce bystander damage in additional tumor cells through spreading within surrounding tumor cells or blood vessels. However, this effect has not been clearly demonstrated due to the difficulty of direct observation. Here, the bystander infection of OVs by vessel delivery and selective infection in 3D multicellular tumoroids (MCTs) in an in vitro microphysiological system (MPS) with integrated medium flow is demonstrated. This study uses replicable vesicular stomatitis virus (VSV)-green fluorescence protein (GFP) to identify the location of infection in 3D MCTs. Using this MPS, the oncoselective infection by VSV-GFP and the spreading by delivery of OVs through flow via block-to-block linkage of the primary infected MPS with uninfected 3D MCTs in an integrated MPS is observed. This MPS enables real-time monitoring and various analysis for the bystander infection of OVs. It is expected that the 3D in vitro MPS can be suitable to investigate the oncoselective spreading and bystander infection of OVs.


Subject(s)
Cytological Techniques , Models, Biological , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses , A549 Cells , Cells, Cultured , Cytological Techniques/instrumentation , Cytological Techniques/methods , Equipment Design , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rhabdoviridae Infections/virology , Spheroids, Cellular/virology , Tumor Cells, Cultured/virology , Vesiculovirus/genetics
10.
Tissue Eng Regen Med ; 17(6): 731-745, 2020 12.
Article in English | MEDLINE | ID: mdl-32207030

ABSTRACT

BACKGROUND: Three-dimensional (3D) cell cultures with architectural and biomechanical properties similar to those of natural tissue have been the focus for generating liver tissue. Microarchitectural organization is believed to be crucial to hepatic function, and 3D cell culture technologies have enabled the construction of tissue-like microenvironments, thereby leading to remarkable progress in vitro models of human tissue and organs. Recently, to recapitulate the 3D architecture of tissues, spheroids and organoids have become widely accepted as new practical tools for 3D organ modeling. Moreover, the combination of bioengineering approach offers the promise to more accurately model the tissue microenvironment of human organs. Indeed, the employment of sophisticated bioengineered liver models show long-term viability and functional enhancements in biochemical parameters and disease-orient outcome. RESULTS: Various 3D in vitro liver models have been proposed as a new generation of liver medicine. Likewise, new biomedical engineering approaches and platforms are available to more accurately replicate the in vivo 3D microarchitectures and functions of living organs. This review aims to highlight the recent 3D in vitro liver model systems, including micropatterning, spheroids, and organoids that are either scaffold-based or scaffold-free systems. Finally, we discuss a number of challenges that will need to be addressed moving forward in the field of liver tissue engineering for biomedical applications. CONCLUSION: The ongoing development of biomedical engineering holds great promise for generating a 3D biomimetic liver model that recapitulates the physiological and pathological properties of the liver and has biomedical applications.


Subject(s)
Cell Culture Techniques , Organoids , Biomedical Engineering , Humans , Liver , Tissue Engineering
11.
RSC Adv ; 10(36): 21328, 2020 Jun 02.
Article in English | MEDLINE | ID: mdl-35532503

ABSTRACT

[This corrects the article DOI: 10.1039/D0RA01577F.].

12.
Nat Commun ; 10(1): 3991, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31488816

ABSTRACT

Lung cancer shows substantial genetic and phenotypic heterogeneity across individuals, driving a need for personalised medicine. Here, we report lung cancer organoids and normal bronchial organoids established from patient tissues comprising five histological subtypes of lung cancer and non-neoplastic bronchial mucosa as in vitro models representing individual patient. The lung cancer organoids recapitulate the tissue architecture of the primary lung tumours and maintain the genomic alterations of the original tumours during long-term expansion in vitro. The normal bronchial organoids maintain cellular components of normal bronchial mucosa. Lung cancer organoids respond to drugs based on their genomic alterations: a BRCA2-mutant organoid to olaparib, an EGFR-mutant organoid to erlotinib, and an EGFR-mutant/MET-amplified organoid to crizotinib. Considering the short length of time from organoid establishment to drug testing, our newly developed model may prove useful for predicting patient-specific drug responses through in vitro patient-specific drug trials.


Subject(s)
Drug Screening Assays, Antitumor , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Organoids/pathology , Animals , B7-H1 Antigen/genetics , Early Detection of Cancer , Genomics , Humans , Lung Neoplasms/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Precision Medicine , Xenograft Model Antitumor Assays
13.
Lab Chip ; 19(17): 2854-2865, 2019 09 07.
Article in English | MEDLINE | ID: mdl-31367720

ABSTRACT

Microfluidic devices as translational research tools provide a potential alternative to animal experiments due to their ability to mimic physiological parameters. Several approaches that can be used to predict the efficacy or toxicity of anticancer drugs are available. In general, standard cell culture systems have the advantages of being relatively cost-effective, having high-throughput capability, and providing convenience. However, these models are inadequate to accurately recapitulate the complex organ-level physiological and pharmacological responses. Here, we present a one-stop microfluidic device enabling both 3-dimensional (3D) lung cancer organoid culturing and drug sensitivity tests directly on a microphysiological system (MPS). Our platform reproducibly yields 3D lung cancer organoids in a size-controllable manner and demonstrates for the first time the production of lung cancer organoids from patients with small-cell lung cancer. Lung cancer organoids derived from primary small-cell lung cancer tumors can rapidly proliferate and exhibit disease-specific characteristics in our MPS. Cisplatin and etoposide, the standard regimen for lung cancer, showed increased apoptosis induction in a concentration-dependent manner, but the organoids contained chemo-resistant cells in the core. We envision that this system may provide important information to guide therapeutic approaches at the preclinical level.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Etoposide/pharmacology , Lung Neoplasms/drug therapy , Microfluidic Analytical Techniques , Organ Culture Techniques , Organoids/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Lung Neoplasms/pathology , Microfluidic Analytical Techniques/instrumentation , Organ Culture Techniques/instrumentation , Organoids/pathology , Particle Size , Surface Properties
14.
Biotechnol Bioeng ; 116(11): 3041-3052, 2019 11.
Article in English | MEDLINE | ID: mdl-31294818

ABSTRACT

The purpose of this study was to demonstrate self-organizing in vitro multicellular tumor spheroid (MCTS) formation in a microfluidic system and to observe the behavior of MCTSs under controlled microenvironment. The employed microfluidic system was designed for simple and effective formation of MCTSs by generating nutrient and oxygen gradients. The MCTSs were composed of cancer cells, vascular endothelial cells, and type I collagen matrix to mimic the in vivo tumor microenvironment (TME). Cell culture medium was perfused to the microfluidic device loaded with MCTSs by a passive fluidic pump at a constant flow rate. The dose response to an MMPs inhibitor was investigated to demonstrate the effects of biochemical substances. The result of long-term stability of MCTSs revealed that continuous perfusion of cell culture medium is one of the major factors for the successful MCTS formation. A continuous flow of cell culture medium in the in vitro TME greatly affected both the proliferation of cancer cells in the micro-wells and the sustainability of the endothelial cell-layer integrity in the lumen of microfluidic channels. Addition of MMP inhibitor to the cell culture medium improved the stability of the collagen matrix by preventing the detachment and shrinkage of the collagen matrix surrounding the MCTSs. In summary, the present constant flow assisted microfluidic system is highly advantageous for long-term observation of the MCTS generation, tumorous tissue formation process and drug responses. MCTS formation in a microfluidic system may serve as a potent tool for studying drug screening, tumorigenesis and metastasis.


Subject(s)
Cell Culture Techniques , Lab-On-A-Chip Devices , Lung Neoplasms/metabolism , Microfluidic Analytical Techniques , Spheroids, Cellular/metabolism , Tumor Microenvironment , A549 Cells , Humans , Lung Neoplasms/pathology , Spheroids, Cellular/pathology
15.
PLoS One ; 14(7): e0219834, 2019.
Article in English | MEDLINE | ID: mdl-31344058

ABSTRACT

The 3D multi-cellular tumoroid (MCT) model is an in vivo-like, avascular tumor model that has received much attention as a refined screening platform for drug therapies. Several types of research have been efforted to improve the physiological characteristics of the tumor microenvironment (TME) of the in vivo-like MCTs. Size-controlled MCTs have received much attention for obtaining highly reproducible results in drug screening assays and achieving a homogeneous and meaningful level of biological activities. Here, we describe an effective method for fabricating the size-controlled in vivo-like MCTs using a cell-loss-free (CLF) microwell arrays. The CLF microwell arrays was fabricated by using the simple operation of laser carving of a poly (methyl methacrylate) (PMMA) master mold. We also demonstrated the biophysicochemical effect of tumor microenvironment (TME) resident fibroblasts through the expression of TGFß, αSMA, Type I-, IV collagen, angiogenesis related markers on tumorigenesis, and confirmed the drug response of MCTs with anti-cancer agents. This technology for the fabrication of CLF microwell arrays could be used as an effective method to produce an in vitro tumor model for cancer research and drug discovery.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Culture Techniques/instrumentation , Spheroids, Cellular/cytology , A549 Cells , Cell Culture Techniques/methods , Cell Line , Cell Proliferation/drug effects , Cell Size/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Human Umbilical Vein Endothelial Cells , Humans , Spheroids, Cellular/drug effects , Tumor Microenvironment/drug effects
16.
Sci Rep ; 8(1): 2365, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29403007

ABSTRACT

In vitro three-dimensional (3D) tumour models mimic natural cancer tissue in vivo, bridging the gap between conventional 2D in vitro testing and animal models. Stromal and cancer tissues with extracellular matrix (ECM) can provide a tumour microenvironment (TME) with cell-to-cell and cell-to-ECM interactions. These interactions induce the exchange of biophysical factors, contributing to the progression, metastasis, and drug resistance of cancer. Here, we describe a 3D in vitro lung cancer model cultured in a microfluidic channel that is able to confirm the role and function of various stromal cells in tumourigenesis, thereby representing an in vivo-like TME. We founded that biophysical factors contribute to the role of fibroblast cells in tumour formation, especially, producing a nascent vessel-like tubular structure, resulting in the formation of vascularized tumour tissue. Fibroblast cells altered the gene expression of the cancer cells to enhance metastasis, survival, and angiogenesis. The device could be used for developing and screening anti-cancer drugs through the formation of the same multicellular tumour spheroids under TME interactions. We believe this microfluidic system provides interaction of TME for cancer research by culturing stromal tissue.


Subject(s)
Fibroblasts/physiology , Lung Neoplasms/physiopathology , Microfluidics/methods , Models, Biological , Neovascularization, Physiologic , Organ Culture Techniques/methods , Tumor Microenvironment , Animals , Cells, Cultured , Stromal Cells/physiology
17.
Oncotarget ; 9(98): 37268, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30647858

ABSTRACT

[This corrects the article DOI: 10.18632/oncotarget.21946.].

18.
Oncotarget ; 8(58): 98887-98897, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-29228735

ABSTRACT

Gastric cancer (GC) is a leading cause of death worldwide and in urgent need of targeted drug development. In the current, we investigated the ability of a repositioned drug verteporfin (VP), originally a treatment for macular degeneration, to inhibit GC cell growth. VP inhibited growth of various GC cell lines. Gene expression profiling of GC cell lines treated with VP revealed that migration-related genes and those with oncogenic potential were down-regulated. Of these genes, we found that FAT1, an adhesion molecule promoting cell invasion, was highly suppressed by VP. Silencing of FAT1 suppressed cell migration and invasion as VP did. FAT1 expression was up-regulated in tumors, and patients with high FAT1-expressing tumors had a worse prognosis. We propose that VP- targeting FAT1 to suppress metastatic potential is a promising therapeutic strategy against GC.

19.
PLoS One ; 12(9): e0184595, 2017.
Article in English | MEDLINE | ID: mdl-28886159

ABSTRACT

Homing of peripheral stem cells is regulated by one of the most representative homing factors, stromal cell-derived factor 1 alpha (SDF-1α), which specifically binds to the plasma membrane receptor CXCR4 of mesenchymal stem cells (MSCs) in order to initiate the signaling pathways that lead to directional migration and homing of stem cells. This complex homing process and directional migration of stem cells have been mimicked on a microfluidic device that is capable of generating a chemokine gradient within the collagen matrix and embedding endothelial cell (EC) monolayers to mimic blood vessels. On the microfluidic device, stem cells showed directional migration toward the higher concentration of SDF-1α, whereas treatment with the CXCR4 antagonist AMD3100 caused loss of directionality of stem cells. Furthermore, inhibition of stem cell's main migratory signaling pathways, Rho-ROCK and Rac pathways, caused blockage of actomyosin and lamellipodia formation, decreasing the migration distance but maintaining directionality. Stem cell homing regulated by SDF-1α caused directional migration of stem cells, while the migratory ability was affected by the activation of migration-related signaling pathways.


Subject(s)
Chemokine CXCL12/chemistry , Lab-On-A-Chip Devices , Mesenchymal Stem Cells/cytology , Amides/pharmacology , Aminoquinolines/pharmacology , Benzylamines , Cell Movement/drug effects , Cyclams , Heterocyclic Compounds/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , Mesenchymal Stem Cells/drug effects , Microscopy, Confocal , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/metabolism
20.
Nat Commun ; 7: 11269, 2016 05 09.
Article in English | MEDLINE | ID: mdl-27157977

ABSTRACT

High-aspect ratio micro- and nano-structures have been used for the production of a variety of applications. In this paper, we describe a simple and cost-effective approach to fabricate an arrayed microarchitecture with an ultra-high aspect ratio using soft materials. The shapes and sizes of the honeycomb structure can be easily modulated by changing the dimensions and position of the base mould pattern and the pressure. The honeycomb structure is used to prepare a drug delivery patch and a microwell array to form cell spheroids without cell loss. The honeycomb structures prepared using natural ECM (collagen-Matrigel) materials are successfully fabricated. The hepatocytes and endothelial cells are seeded and co-cultured in the ECM-based micro-honeycomb to prepare a 3D liver model successfully mimicking an ultrastructure of liver and providing enhanced liver function.

SELECTION OF CITATIONS
SEARCH DETAIL
...