Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters

Publication year range
1.
Int Immunopharmacol ; 100: 108054, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34492537

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (HSCT) remains the key for the treatment of malignant hematological diseases, and acute graft-versus-host disease (aGVHD) that might occur after allogenic transplantation can be life threatening and promote disease recurrence. GVHD damages the various parts of the body by upregulating T helper 1 cytokines (Th1) cytokines and stimulating CD4、CD8 + T cells. GVHD can exhibit significant immunoregulatory effects, but could be easily affected by the mesenchymal stem cells (MSC) environment, and hence the MSC immunosuppressive effects on GVHD remain unpredictable. Hence, to better understand the role of MSC in the prevention and treatment of GVHD, umbilical cord derived mesenchymal stem cells (UC-MSC) were pre-treated with Chinese medicine Asarinin and IFN-γ. In the mix lymphocyte reaction, we found that Asarinin pre-treated UC-MSC can exert significantly greater inhibition towards the proliferation of CD4 and CD8 + T cells, down-regulate Th1 type cytokines, up-regulate Th2 type cytokines, and reduce the inflammatory damage to liver, lung and intestine of aGVHD mice model. Moreover, Asarinin can cooperate with IFN-γto promote UC-MSC to secrete indoleamine 2,3-dioxygenase (IDO). Our findings establish that Asarinin pre-treated UC-MSC can significantly promote the immunosuppressive effects of MSC on aGVHD after hematopoietic stem cell transplantation.


Subject(s)
Dioxoles/pharmacology , Drugs, Chinese Herbal/pharmacology , Graft vs Host Disease/therapy , Lignans/pharmacology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/drug effects , Animals , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Mesenchymal Stem Cells/immunology , Mice , Primary Cell Culture/methods , Transplantation, Homologous/adverse effects , Umbilical Cord/cytology
2.
Nutrients ; 13(8)2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34444791

ABSTRACT

Traditional medicines rely mainly on use of plant extracts to mitigate or treat a wide range of disorders, including those that affect skeletal homeostasis. In this study, we investigated for the first time the potential pro-osteogenic effects of hexane, acetone and methanol extracts of the leaves of Cucurbita moschata, a very popular pumpkin cultivar in Western countries. We found that in Cucurbita moschata leaves, there are acetone-extractable substances-in particular, fatty acids such as 13-OH-9Z,11E,15E-octadecatrienoic acid (PU-13OH-FA), which is capable of both stimulating the function of human primary osteoblasts, which are responsible for bone formation, and inhibiting the differentiation of human osteoclasts, which are responsible for bone resorption. This dual effect was monitored by analyzing Runx2 expression, deposition of mineralized matrix, ALP activity, TRAP and actin ring staining respectively. This study suggests that bioactive chemicals from Cucurbita moschata leaves are potentially suitable as therapeutics for managing metabolic bone disorders such as osteoporosis and rheumatoid arthritis, and promoting tissue healing and functional recovery after bone fractures. The data we obtained increase knowledge on the biological activities of Cucurbita moschata, and in particular underline the potential benefits of consuming leaves which are a part of the plant currently little considered in the Western world.


Subject(s)
Cucurbita/chemistry , Osteogenesis/drug effects , Plant Extracts/pharmacology , Plant Leaves/chemistry , Primary Cell Culture/methods , Bone Resorption , Cell Differentiation/drug effects , Cell Survival/drug effects , Dicarboxylic Acids , Humans , Middle Aged , Osteoblasts/drug effects , Osteoclasts/drug effects
3.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33443156

ABSTRACT

Fertility relies upon pulsatile release of gonadotropin-releasing hormone (GnRH) that drives pulsatile luteinizing hormone secretion. Kisspeptin (KP) neurons in the arcuate nucleus are at the center of the GnRH pulse generation and the steroid feedback control of GnRH secretion. However, KP evokes a long-lasting response in GnRH neurons that is hard to reconcile with periodic GnRH activity required to drive GnRH pulses. Using calcium imaging, we show that 1) the tetrodotoxin-insensitive calcium response evoked by KP relies upon the ongoing activity of canonical transient receptor potential channels maintaining voltage-gated calcium channels in an activated state, 2) the duration of the calcium response is determined by the rate of resynthesis of phosphatidylinositol 4,5-bisphosphate (PIP2), and 3) nitric oxide terminates the calcium response by facilitating the resynthesis of PIP2 via the canonical pathway guanylyl cyclase/3',5'-cyclic guanosine monophosphate/protein kinase G. In addition, our data indicate that exposure to nitric oxide after KP facilitates the calcium response to a subsequent KP application. This effect was replicated using electrophysiology on GnRH neurons in acute brain slices. The interplay between KP and nitric oxide signaling provides a mechanism for modulation of the refractory period of GnRH neurons after KP exposure and places nitric oxide as an important component for tonic GnRH neuronal pulses.


Subject(s)
Calcium Signaling/physiology , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Female , Hypothalamus/metabolism , Kisspeptins/metabolism , Luteinizing Hormone/metabolism , Male , Mice , Nitric Oxide/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/physiology , Primary Cell Culture/methods
4.
Clin Transl Sci ; 14(3): 1049-1061, 2021 05.
Article in English | MEDLINE | ID: mdl-33382907

ABSTRACT

Liver microphysiological systems (MPSs) are promising models for predicting hepatic drug effects. Yet, after a decade since their introduction, MPSs are not routinely used in drug development due to lack of criteria for ensuring reproducibility of results. We characterized the feasibility of a liver MPS to yield reproducible outcomes of experiments assaying drug toxicity, metabolism, and intracellular accumulation. The ability of the liver MPS to reproduce hepatotoxic effects was assessed using trovafloxacin, which increased lactate dehydrogenase (LDH) release and reduced cytochrome P450 3A4 (CYP3A4) activity. These observations were made in two test sites and with different batches of Kupffer cells. Upon culturing equivalent hepatocytes in the MPS, spheroids, and sandwich cultures, differences between culture formats were detected in CYP3A4 activity and albumin production. Cells in all culture formats exhibited different sensitivities to hepatotoxicant exposure. Hepatocytes in the MPS were more functionally stable than those of other culture platforms, as CYP3A4 activity and albumin secretion remained prominent for greater than 18 days in culture, whereas functional decline occurred earlier in spheroids (12 days) and sandwich cultures (7 days). The MPS was also demonstrated to be suitable for metabolism studies, where CYP3A4 activity, troglitazone metabolites, diclofenac clearance, and intracellular accumulation of chloroquine were quantified. To ensure reproducibility between studies with the MPS, the combined use of LDH and CYP3A4 assays were implemented as quality control metrics. Overall results indicated that the liver MPS can be used reproducibly in general drug evaluation applications. Study outcomes led to general considerations and recommendations for using liver MPSs. Study Highlights WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC? Microphysiological systems (MPSs) have been designed to recreate organ- or tissue-specific characteristics of extracellular microenvironments that enhance the physiological relevance of cells in culture. Liver MPSs enable long-lasting and stable culture of hepatic cells by culturing them in three-dimensions and exposing them to fluid flow. WHAT QUESTION DID THIS STUDY ADDRESS? What is the functional performance relative to other cell culture platforms and the reproducibility of a liver MPS for assessing drug development and evaluation questions, such as toxicity, metabolism, and pharmacokinetics? WHAT DOES THIS STUDY ADD TO OUR KNOWLEDGE? The liver MPS systematically detected the toxicity of trovafloxacin. When compared with spheroids and sandwich cultures, this system had a more stable function and different sensitivity to troglitazone, tamoxifen, and digoxin. Quantifying phase II metabolism of troglitazone and intracellular accumulation of chloroquine demonstrated the potential use of the liver MPS for studying drug metabolism and pharmacokinetics. Quality control criteria for assessing chip function were key for reliably using the liver MPS. HOW MIGHT THIS CHANGE CLINICAL PHARMACOLOGY OR TRANSLATIONAL SCIENCE? Due to its functional robustness and physiological relevance (3D culture, cells expose to fluid flow and co-culture of different cell types), the liver MPS can, in a reproducible manner: (i) detect inflammatory-induced drug toxicity, as demonstrated with trovafloxacin, (ii) detect the toxicity of other drugs, such as troglitazone, tamoxifen, and digoxin, with different effects than those detected in spheroids and sandwich cultures, (iii) enable studies of hepatic function that rely on prolonged cellular activity, and (iv) detect phase II metabolites and drug accumulation to potentially support the interpretation of clinical data. The integration of MPSs in drug development will be facilitated by careful evaluation of performance and reproducibility as performed in this study.


Subject(s)
Liver/drug effects , Primary Cell Culture/methods , Toxicity Tests/methods , Cells, Cultured , Cytochrome P-450 CYP3A/metabolism , Drug Evaluation, Preclinical , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Lab-On-A-Chip Devices , Liver/cytology , Liver/metabolism , Models, Biological , Primary Cell Culture/instrumentation , Reproducibility of Results , Spheroids, Cellular , Toxicity Tests/instrumentation
5.
J Clin Endocrinol Metab ; 106(3): 843-857, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33212489

ABSTRACT

CONTEXT: Disorders affecting adrenal steroidogenesis promote an imbalance in the normally tightly controlled secretion of mineralocorticoids, glucocorticoids, and androgens. This may lead to differences/disorders of sex development in the fetus, as seen in virilized girls with congenital adrenal hyperplasia (CAH). Despite the important endocrine function of human fetal adrenals, neither normal nor dysregulated adrenal steroidogenesis is understood in detail. OBJECTIVE: Due to significant differences in adrenal steroidogenesis between human and model species (except higher primates), we aimed to establish a human fetal adrenal model that enables examination of both de novo and manipulated adrenal steroidogenesis. DESIGN AND SETTING: Human adrenal tissue from 54 1st trimester fetuses were cultured ex vivo as intact tissue fragments for 7 or 14 days. MAIN OUTCOME MEASURES: Model validation included examination of postculture tissue morphology, viability, apoptosis, and quantification of steroid hormones secreted to the culture media measured by liquid chromatography-tandem mass spectrometry. RESULTS: The culture approach maintained cell viability, preserved cell populations of all fetal adrenal zones, and recapitulated de novo adrenal steroidogenesis based on continued secretion of steroidogenic intermediates, glucocorticoids, and androgens. Adrenocorticotropic hormone and ketoconazole treatment of ex vivo cultured human fetal adrenal tissue resulted in the stimulation of steroidogenesis and inhibition of androgen secretion, respectively, demonstrating a treatment-specific response. CONCLUSIONS: Together, these data indicate that ex vivo culture of human fetal adrenal tissue constitutes a novel approach to investigate local effects of pharmaceutical exposures or emerging therapeutic options targeting imbalanced steroidogenesis in adrenal disorders, including CAH.


Subject(s)
Adrenal Glands/cytology , Drug Evaluation, Preclinical/methods , Fetus/cytology , Primary Cell Culture/methods , Steroids/biosynthesis , Adrenal Glands/drug effects , Adrenal Glands/embryology , Adrenal Glands/metabolism , Adrenal Hyperplasia, Congenital/drug therapy , Adrenal Hyperplasia, Congenital/metabolism , Adrenal Hyperplasia, Congenital/pathology , Adrenocorticotropic Hormone/pharmacology , Androgens/metabolism , Cell Survival , Culture Media/chemistry , Female , Glucocorticoids/pharmacology , Humans , Ketoconazole/pharmacology , Metabolic Networks and Pathways/drug effects , Models, Biological , Pregnancy , Steroids/analysis , Steroids/metabolism
6.
AAPS J ; 22(6): 133, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33063163

ABSTRACT

Suspended human hepatocytes (SHH) have long been used in assessing hepatic drug uptake, while plated human hepatocytes in short-term monolayer culture (PHH) have gained use in recent years. This study aimed to cross-evaluate SHH and PHH in measuring the hepatic uptake mediated by organic anion transporting polypeptide 1Bs (OATP1Bs). We compared the time courses of cell-to-medium (C/M) concentration ratios and initial uptake clearance values of the OATP1B substrates (pitavastatin, rosuvastatin, cerivastatin, pravastatin, dehydropravastatin, and SC-62807) between SHH and PHH. For all compounds except cerivastatin, the C/M ratios in SHH displayed an apparent overshoot (an initial increase followed by a decrease) during the 180-min uptake experiment, but not in PHH. Based on the literature evidence suggesting the possible internalization of OATP1Bs in primary hepatocytes, separate experiments measured the drug uptake after varying lengths of pre-incubation in the drug-free medium. The initial uptake clearances of pitavastatin and rosuvastatin declined in SHH beyond an apparent threshold time of 20-min drug-free pre-incubation, but not in PHH. Kinetic modeling quantitatively captured the decline in the active uptake clearance in SHH, and more than half of the active uptake clearances of pitavastatin and rosuvastatin were prone to loss during the 180-min uptake experiment. These results suggested a partial, time-delayed loss of the functional OATP1Bs in SHH upon prolonged incubation. Our results indicate that PHH is more appropriate for experiments where a prolonged incubation is required, such as estimation of unbound hepatocyte-to-medium concentration ratio (Kp,uu) at the steady-state.


Subject(s)
Hepatocytes/enzymology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Liver-Specific Organic Anion Transporter 1/metabolism , Adult , Cells, Cultured , Child , Culture Media/analysis , Culture Media/metabolism , Drug Evaluation, Preclinical/methods , Hepatobiliary Elimination , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/analysis , Male , Models, Biological , Primary Cell Culture/methods
7.
J Pharmacol Toxicol Methods ; 106: 106916, 2020.
Article in English | MEDLINE | ID: mdl-32890732

ABSTRACT

INTRODUCTION: The evaluation of microvascular permeability is crucial for drug development. Nonetheless, there are few reliable test methods in vitro due to the lack of vascular endothelial models suitable for quantitative analyses. The purpose of this study is to construct a novel microvascular endothelial model with the high endothelial barrier function and sensitivity to physiological stimuli utilizing a collagen vitrigel membrane (CVM) composed of high-density collagen fibrils. METHODS: Human microvascular endothelial cells (HMVECs) were cultured for 6 days in a CVM chamber with or without human dermal fibroblasts (HDFs) cocultured on the reverse surface of the CVM. The endothelial barrier function was evaluated by measurement of transendothelial electrical resistance (TEER) and macromolecular permeation. RESULTS: The TEER value of a monolayer of HMVECs cultured on the CVM was 15-20 Ωï½¥cm2 during the culture period while it reached over 60 Ωï½¥cm2 by coculturing with HDFs. The TEER value was decreased from 5.7 to 3.4 Ωï½¥cm2 by 100 µM histamine in the monolayer model and from 50 to 32 Ωï½¥cm2 by 1 nM histamine in the coculture model, respectively. Interestingly, the permeability coefficient of the compound with a molecular weight of not 376 and 40,000 but 4000 was selectively increased in the histamine-treated coculture model. DISCUSSION: HMVECs cocultured with HDFs via a CVM formed the tight endothelial barrier and showed high responsivity to histamine. The model might be useful for exploring molecules that modulate microvascular permeability and pass through the microvascular endothelial barrier.


Subject(s)
Capillary Permeability/drug effects , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Histamine/pharmacology , Primary Cell Culture/methods , Cells, Cultured , Coculture Techniques , Collagen , Drug Evaluation, Preclinical/methods , Electric Impedance , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Feasibility Studies , Fibroblasts , Humans , Membranes, Artificial
8.
Plast Reconstr Surg ; 146(2): 309-320, 2020 08.
Article in English | MEDLINE | ID: mdl-32740581

ABSTRACT

BACKGROUND: Adipose-derived stem cells are considered as candidate cells for regenerative plastic surgery. Measures to influence cellular properties and thereby direct their regenerative potential remain elusive. Hyperbaric oxygen therapy-the exposure to 100% oxygen at an increased atmospheric pressure-has been propagated as a noninvasive treatment for a multitude of indications and presents a potential option to condition cells for tissue-engineering purposes. The present study evaluates the effect of hyperbaric oxygen therapy on human adipose-derived stem cells. METHODS: Human adipose-derived stem cells from healthy donors were treated with hyperbaric oxygen therapy at 2 and 3 atm. Viability before and after each hyperbaric oxygen therapy, proliferation, expression of surface markers and protein contents of transforming growth factor (TGF)-ß, tumor necrosis factor-α, hepatocyte growth factor, and epithelial growth factor in the supernatants of treated adipose-derived stem cells were measured. Lastly, adipogenic, osteogenic, and chondrogenic differentiation with and without use of differentiation-inducing media (i.e., autodifferentiation) was examined. RESULTS: Hyperbaric oxygen therapy with 3 atm increased viability, proliferation, and CD34 expression and reduced the CD31/CD34/CD45 adipose-derived stem cell subset and endothelial progenitor cell population. TGF-ß levels were significantly decreased after two hyperbaric oxygen therapy sessions in the 2-atm group and decreased after three hyperbaric oxygen therapy sessions in the 3-atm group. Hepatocyte growth factor secretion remained unaltered in all groups. Although the osteogenic and chondrogenic differentiation were not influenced, adipogenic differentiation and autodifferentiation were significantly enhanced, with osteogenic autodifferentiation significantly alleviated by hyperbaric oxygen therapy with 3 atm. CONCLUSION: Hyperbaric oxygen therapy with 3 atm increases viability and proliferation of adipose-derived stem cells, alters marker expression and subpopulations, decreases TGF-ß secretion, and skews adipose-derived stem cells toward adipogenic differentiation. CLINICAL QUESTION/LEVEL OF EVIDENCE: Therapeutic, V.


Subject(s)
Adipogenesis/drug effects , Cell Differentiation/drug effects , Cell Engineering/methods , Mesenchymal Stem Cells/drug effects , Oxygen/administration & dosage , Adipose Tissue/cytology , Adult , Biomarkers/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytokines/metabolism , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mesenchymal Stem Cells/physiology , Middle Aged , Pressure , Primary Cell Culture/methods
9.
Drug Metab Dispos ; 48(9): 804-810, 2020 09.
Article in English | MEDLINE | ID: mdl-32623369

ABSTRACT

The HepatoPac micropatterned coculture (MPCC) hepatocyte system has been shown to be an effective tool to investigate the qualitative human and preclinical species' metabolite profiles of new drug candidates. However, additional improvements to the overall study conditions and execution, layout, and human-donor count could be made. To that end, we have evaluated several ways to increase the amount of data one can generate per MPCC plate and how to more efficiently execute a MPCC study for the purpose of metabolite generation. Herein, we compare a set of compounds using single- and 10-donor pooled human MPCC hepatocytes. Intrinsic clearance and mean metabolic activities assessed by diverse enzyme markers were comparable between the single- and 10-donor pool. We have confirmed that the generated metabolite profiles were indistinguishable between the single- and 10-donor pool and also that rat MPCC can be performed at 400 µl media volume, which greatly simplifies study execution. Additional tips for successful study execution are also described. SIGNIFICANCE STATEMENT: When using the HepatoPac micropatterned coculture (MPCC) system, sometimes simple experimental condition variables or problematic plate designs can hamper productive study execution. We evaluated conditions to increase the amount of data one can generate per MPCC plate and, perhaps more importantly, execute that study more efficiently with less likelihood of error. We describe some of our key learnings, provide an examination of enzyme activity levels and clearance values, and provide some recommendations to simplify the execution of a HepatoPac experiment.


Subject(s)
Hepatobiliary Elimination , Metabolomics/methods , Primary Cell Culture/methods , Animals , Biotransformation , Chromatography, High Pressure Liquid/methods , Coculture Techniques/methods , Datasets as Topic , Drug Evaluation, Preclinical/methods , Female , Fibroblasts , Hepatocytes/metabolism , Humans , Male , Rats , Tandem Mass Spectrometry/methods
10.
Drug Metab Dispos ; 48(8): 690-697, 2020 08.
Article in English | MEDLINE | ID: mdl-32503882

ABSTRACT

Long-term hepatocyte culture systems such as HepatoPac are well suited to evaluate the metabolic turnover of low clearance (CL) drugs because of their sustained metabolic capacity and longer-term viability. Erythromycin (ERY), a moderate, mechanism-based inhibitor of CYP3A, was evaluated as a tool in the HepatoPac model to assess contribution of CYP3A to the clearance of drug candidates. ERY inhibited CYP3A activity by 58% and 80% at 3 and 10 µM, respectively, for up to 72 hours. At 30 µM, ERY inhibited midazolam hydroxylation by >85% for the entire 144-hour duration of the incubation. Alprazolam CLint was inhibited 58% by 3 µM of ERY, 75% by 15 µM of ERY, 89% by 30 µM of ERY, and 94% by 60 µM of ERY. ERY (30 µM) did not markedly affect CLint of substrates for several other major cytochrome P450 isoforms evaluated and did not markedly inhibit uridine diphosphoglucuronosyl transferase (UGT) isoforms 1A1, 1A3, 1A4, 1A6, 1A9, 2B7, or 2B15 as assessed using recombinant UGTs. ERY only mildly increased CYP3A4 gene expression by 2.1-fold (14% of rifampicin induction) at 120 µM, indicating that at effective concentrations for inhibition of CYP3A activity (30-60 µM), arylhydrocarbon receptor, constitutive androstane receptor, and pregnane-X-receptor activation are not likely to markedly increase levels of other drug-metabolizing enzymes or transporters. ERY at concentrations up to 60 µM was not toxic for up to 6 days of incubation. Use of ERY to selectively inhibit CYP3A in high-functioning, long-term hepatocyte models such as HepatoPac can be a valuable strategy to evaluate the contribution of CYP3A metabolism to the overall clearance of slowly metabolized drug candidates. SIGNIFICANCE STATEMENT: This work describes the use of erythromycin as a selective inhibitor of CYP3A to assess the contribution of CYP3A in the metabolism of compounds using long-term hepatocyte cultures.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A/metabolism , Erythromycin/pharmacology , Hepatobiliary Elimination/drug effects , Adult , Alprazolam/pharmacokinetics , Cells, Cultured , Coculture Techniques/methods , Cytochrome P-450 CYP3A Inducers/pharmacology , Drug Evaluation, Preclinical/methods , Female , Glucuronosyltransferase/metabolism , Hepatocytes , Humans , Male , Midazolam/pharmacokinetics , Middle Aged , Primary Cell Culture/methods , Rifampin/pharmacology , Time Factors
11.
Sci Rep ; 10(1): 4867, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32184427

ABSTRACT

High callus production is a feasible way to improve the propagation coefficient of garlic. It remains unknown how genotypes and explants affect garlic callus formation. In the present investigation, we found that there were significant differences in callus formation among garlic varieties. Tip explants were the best calli-producing source, and 91.05% of the explants from four varieties, on average, formed calli after 45 d of primary culturing. Upper leaf parts explants produced lower values. Among the different varieties and explant types, tip explants of variety T141 induced calli in the shortest time and had the greatest callus fresh weight at 45 d. An endogenous hormone contents analysis showed that auxins (indole-3-acetic acid and methyl indole-3-acetic acetate), cytokinins (trans-zeatin and dihydrozeatin), gibberellins4, 9,15,19,24 and 53, abscisic acid, jasmonic acid, jasmonoyl-L-isoleucine, and dihydrojasmonic acid were significantly greater in the tips than those in the upper leaf parts. High endogenous jasmonic acid content might play important roles in callus formation. These results will help us not only establish an efficient garlic callus induction protocol that can be applied to large-scale callus multiplication and regeneration, and to genetically improvement of garlic production, but also understand endogenous hormone roles in tissue/organ differentiation and dedifferentiation.


Subject(s)
Garlic/growth & development , Garlic/genetics , Plant Growth Regulators/metabolism , Abscisic Acid/metabolism , Cytokinins/metabolism , Garlic/metabolism , Genotype , Gibberellins/metabolism , Indoleacetic Acids/metabolism , Organ Specificity , Plant Leaves/genetics , Plant Leaves/growth & development , Plant Leaves/metabolism , Primary Cell Culture/methods
12.
Stem Cells Dev ; 29(11): 728-736, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32122257

ABSTRACT

This study was conducted to compare the in vitro proliferation and osteogenic differentiation potential of mesenchymal stem cells (MSCs) derived from mandibular (M-MSCs) or femur (F-MSCs) tissues of rats. M-MSC and F-MSC cultures were isolated and established from the same rat. Cultures were observed for morphological changes by microscope and growth characteristics by CCK-8 and cloning assays. Cell adhesion ability on a culture plate and titanium sheet was detected by staining with toluidine blue and Hoechst 33258, respectively. The levels of Ca, P, and ALP (serially) during osteogenic differentiation were evaluated. Cultures were analyzed for mineralization potential with alizarin red and ALP staining methods and for differentiation markers with RT-PCR (ALP, Runx2, and OCN). M-MSCs and F-MSCs were successfully isolated from the same rat with uncontaminated culture, which showed significant differences in morphology. The proliferation rate of M-MSCs was higher than F-MSCs in primary culture, but significantly lower after passage. More colonies are formed from F-MSCs than from M-MSCs. M-MSCs showed a significantly higher mineralization and osteogenic differentiation potential, which might be of significance for use in bone/dental tissue engineering. In vitro, cell passage will decrease the proliferation ability of M-MSCs. The higher mineralization and osteogenic differentiation potential of M-MSCs could make them an approachable stem cell source for further application in stem cell-based clinical therapies.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation , Femur/cytology , Mandible/cytology , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Calcium/metabolism , Cell Proliferation , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Organ Specificity , Osteoblasts/metabolism , Osteoblasts/physiology , Osteocalcin/genetics , Osteocalcin/metabolism , Phosphorus/metabolism , Primary Cell Culture/methods , Rats , Rats, Sprague-Dawley , Tissue Engineering/methods
13.
Article in English | MEDLINE | ID: mdl-31678517

ABSTRACT

Full thickness models (FTMs) are 3D-cultured human skin models that mimic many aspects of native human skin (NHS). However, their stratum corneum (SC) lipid composition differs from NHS causing a reduced skin barrier. The most pronounced differences in lipid composition are a reduction in lipid chain length and increased monounsaturated lipids. The liver-X-receptor (LXR) activates the monounsaturated lipid synthesis via stearoyl-CoA desaturase-1 (SCD-1). Therefore, the aim was to improve the SC lipid synthesis of FTMs by LXR deactivation. This was achieved by supplementing culture medium with LXR antagonist GSK2033. LXR agonist T0901317 was added for comparison. Subsequently, epidermal morphogenesis, lipid composition, lipid organization and the barrier functionality of these FTMs were assessed. We demonstrate that LXR deactivation resulted in a lipid composition with increased overall chain lengths and reduced levels of monounsaturation, whereas LXR activation increased the amount of monounsaturated lipids and led to a reduction in the overall chain length. However, these changes did not affect the barrier functionality. In conclusion, LXR deactivation led to the development of FTMs with improved lipid properties, which mimic the lipid composition of NHS more closely. These novel findings may contribute to design interventions to normalize SC lipid composition of atopic dermatitis patients.


Subject(s)
Culture Media/pharmacology , Liver X Receptors/antagonists & inhibitors , Primary Cell Culture/methods , Skin/drug effects , Sulfonamides/pharmacology , Ceramides/metabolism , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Drug Evaluation, Preclinical/methods , Fatty Acids, Nonesterified , Humans , Hydrocarbons, Fluorinated/pharmacology , Lipogenesis/drug effects , Liver X Receptors/agonists , Liver X Receptors/metabolism , Morphogenesis/drug effects , Skin/growth & development , Skin/metabolism , Stearoyl-CoA Desaturase/metabolism
14.
Sci Rep ; 9(1): 18210, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31796850

ABSTRACT

The interaction of light with biological tissues has been considered for various therapeutic applications. Light-induced neurite growth has the potential to be a clinically useful technique for neuron repair. However, most previous studies used either a large illumination area to accelerate overall neurite growth or employed a light spot to guide a growing neurite. It is not clear if optical stimulation can induce the regrowth of a retracted neurite. In the present work, we used blue light (wavelength: 473 nm) to cause neurite retraction, and we proved that using a red-light (wavelength: 650 nm) spot to illuminate the soma near the junction of the retracted neurite could induce neurite regrowth. As a comparison, we found that green light (wavelength 550 nm) had a 62% probability of inducing neurite regrowth, while red light had a 75% probability of inducing neurite regrowth at the same power level. Furthermore, the neurite regrowth length induced by red light was increased by the pre-treatment with inhibitors of myosin functions. We also observed actin propagation from the soma to the tip of the re-growing neurite following red-light stimulation of the soma. The red light-induced extension and regrowth were abrogated in the calcium-free medium. These results suggest that illumination with a red-light spot on the soma may trigger the regrowth of a neurite after the retraction caused by blue-light illumination.


Subject(s)
Light , Nerve Regeneration/radiation effects , Neurites/physiology , Actins/metabolism , Animals , Calcium/metabolism , Cell Line, Tumor , Color , Culture Media/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Hippocampus/cytology , Low-Level Light Therapy/methods , Mice , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , Nerve Regeneration/drug effects , Neurites/radiation effects , Primary Cell Culture/methods , Rats
15.
World J Gastroenterol ; 25(30): 4125-4147, 2019 Aug 14.
Article in English | MEDLINE | ID: mdl-31435168

ABSTRACT

The introduction of biologics such as anti-tumor necrosis factor (TNF) monoclonal antibodies followed by anti-integrins has dramatically changed the therapeutic paradigm of inflammatory bowel diseases (IBD). Furthermore, a newly developed anti-p40 subunit of interleukin (IL)-12 and IL-23 (ustekinumab) has been recently approved in the United States for patients with moderate to severe Crohn's disease who have failed treatment with anti-TNFs. However, these immunosuppressive therapeutics which focus on anti-inflammatory mechanisms or immune cells still fail to achieve long-term remission in a significant percentage of patients. This strongly underlines the need to identify novel treatment targets beyond immune suppression to treat IBD. Recent studies have revealed the critical role of intestinal epithelial cells (IECs) in the pathogenesis of IBD. Physical, biochemical and immunologic driven barrier dysfunctions of epithelial cells contribute to the development of IBD. In addition, the recent establishment of adult stem cell-derived intestinal enteroid/organoid culture technology has allowed an exciting opportunity to study human IECs comprising all normal epithelial cells. This long-term epithelial culture model can be generated from endoscopic biopsies or surgical resections and recapitulates the tissue of origin, representing a promising platform for novel drug discovery in IBD. This review describes the advantages of intestinal enteroids/organoids as a research tool for intestinal diseases, introduces studies with these models in IBD, and gives a description of the current status of therapeutic approaches in IBD. Finally, we provide an overview of the current endeavors to identify a novel drug target for IBD therapy based on studies with human enteroids/organoids and describe the challenges in using enteroids/organoids as an IBD model.


Subject(s)
Drug Discovery/methods , Immunosuppressive Agents/pharmacology , Inflammatory Bowel Diseases/drug therapy , Intestinal Mucosa/drug effects , Organoids/drug effects , Biological Products/pharmacology , Biological Products/therapeutic use , Biopsy , Drug Evaluation, Preclinical/methods , Epithelial Cells , Humans , Immunosuppressive Agents/therapeutic use , Induced Pluripotent Stem Cells , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Organoids/immunology , Primary Cell Culture/methods
16.
Nat Commun ; 10(1): 2621, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31197168

ABSTRACT

The high selectivity of the human blood-brain barrier (BBB) restricts delivery of many pharmaceuticals and therapeutic antibodies to the central nervous system. Here, we describe an in vitro microfluidic organ-on-a-chip BBB model lined by induced pluripotent stem cell-derived human brain microvascular endothelium interfaced with primary human brain astrocytes and pericytes that recapitulates the high level of barrier function of the in vivo human BBB for at least one week in culture. The endothelium expresses high levels of tight junction proteins and functional efflux pumps, and it displays selective transcytosis of peptides and antibodies previously observed in vivo. Increased barrier functionality was accomplished using a developmentally-inspired induction protocol that includes a period of differentiation under hypoxic conditions. This enhanced BBB Chip may therefore represent a new in vitro tool for development and validation of delivery systems that transport drugs and therapeutic antibodies across the human BBB.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Delivery Systems/methods , Endothelial Cells/metabolism , Microfluidics/instrumentation , Antibodies/pharmacology , Astrocytes , Blood-Brain Barrier/cytology , Drug Evaluation, Preclinical/instrumentation , Drug Evaluation, Preclinical/methods , Endothelium, Vascular/cytology , Humans , Lab-On-A-Chip Devices , Microfluidics/methods , Microvessels/cytology , Pericytes , Permeability , Pluripotent Stem Cells , Primary Cell Culture/instrumentation , Primary Cell Culture/methods
17.
Stem Cells Transl Med ; 8(8): 810-821, 2019 08.
Article in English | MEDLINE | ID: mdl-31038850

ABSTRACT

Xenogeneic-free media are required for translating advanced therapeutic medicinal products to the clinics. In addition, process efficiency is crucial for ensuring cost efficiency, especially when considering large-scale production of mesenchymal stem cells (MSCs). Human platelet lysate (HPL) has been increasingly adopted as an alternative for fetal bovine serum (FBS) for MSCs. However, its therapeutic and regenerative potential in vivo is largely unexplored. Herein, we compare the effects of FBS and HPL supplementation for a scalable, microcarrier-based dynamic expansion of human periosteum-derived cells (hPDCs) while assessing their bone forming capacity by subcutaneous implantation in small animal model. We observed that HPL resulted in faster cell proliferation with a total fold increase of 5.2 ± 0.61 in comparison to 2.7 ± 02.22-fold in FBS. Cell viability and trilineage differentiation capability were maintained by HPL, although a suppression of adipogenic differentiation potential was observed. Differences in mRNA expression profiles were also observed between the two on several markers. When implanted, we observed a significant difference between the bone forming capacity of cells expanded in FBS and HPL, with HPL supplementation resulting in almost three times more mineralized tissue within calcium phosphate scaffolds. FBS-expanded cells resulted in a fibrous tissue structure, whereas HPL resulted in mineralized tissue formation, which can be classified as newly formed bone, verified by µCT and histological analysis. We also observed the presence of blood vessels in our explants. In conclusion, we suggest that replacing FBS with HPL in bioreactor-based expansion of hPDCs is an optimal solution that increases expansion efficiency along with promoting bone forming capacity of these cells. Stem Cells Translational Medicine 2019;8:810&821.


Subject(s)
Batch Cell Culture Techniques/methods , Bone Regeneration , Culture Media/pharmacology , Primary Cell Culture/methods , Stem Cells/drug effects , Adipogenesis , Animals , Batch Cell Culture Techniques/instrumentation , Bioreactors , Blood Platelets/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Culture Media/chemistry , Humans , Mice , Mice, Nude , Osteogenesis , Periosteum/cytology , Primary Cell Culture/instrumentation , Stem Cell Transplantation/methods , Stem Cells/physiology
18.
Stem Cells Transl Med ; 8(8): 758-767, 2019 08.
Article in English | MEDLINE | ID: mdl-31020786

ABSTRACT

As common chemotherapeutic agents are associated with an increased risk of acute and chronic cardiovascular complications, a new clinical discipline, cardio-oncology, has recently emerged. At the same time, the development of preclinical human stem cell-derived cardiovascular models holds promise as a more faithful platform to predict the cardiovascular toxicity of common cancer therapies and advance our understanding of the underlying mechanisms contributing to the cardiotoxicity. In this article, we review the recent advances in preclinical cancer-related cardiotoxicity testing, focusing on new technologies, such as human induced pluripotent stem cell-derived cardiomyocytes and tissue engineering. We further discuss some of the limitations of these technologies and present future directions. Stem Cells Translational Medicine 2019;8:758&767.


Subject(s)
Antineoplastic Agents/toxicity , Drug Evaluation, Preclinical/methods , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Precision Medicine/methods , Animals , Cardiotoxicity , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Lab-On-A-Chip Devices , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , Primary Cell Culture/methods
19.
Science ; 364(6438): 399-402, 2019 04 26.
Article in English | MEDLINE | ID: mdl-31023926

ABSTRACT

The maintenance of terminally differentiated cells, especially hepatocytes, in vitro has proven challenging. Here we demonstrated the long-term in vitro maintenance of primary human hepatocytes (PHHs) by modulating cell signaling pathways with a combination of five chemicals (5C). 5C-cultured PHHs showed global gene expression profiles and hepatocyte-specific functions resembling those of freshly isolated counterparts. Furthermore, these cells efficiently recapitulated the entire course of hepatitis B virus (HBV) infection over 4 weeks with the production of infectious viral particles and formation of HBV covalently closed circular DNA. Our study demonstrates that, with a chemical approach, functional maintenance of PHHs supports long-term HBV infection in vitro, providing an efficient platform for investigating HBV cell biology and antiviral drug screening.


Subject(s)
Hepatitis B virus/growth & development , Hepatocytes/physiology , Hepatocytes/virology , Primary Cell Culture/methods , Virus Cultivation/methods , Antiviral Agents/isolation & purification , Antiviral Agents/pharmacology , DNA, Circular/biosynthesis , DNA, Circular/isolation & purification , DNA, Viral/biosynthesis , DNA, Viral/isolation & purification , Drug Evaluation, Preclinical , Hepatitis B virus/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Transcriptome , Virion/drug effects , Virion/growth & development
20.
Biochim Biophys Acta Mol Basis Dis ; 1865(5): 920-928, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30264693

ABSTRACT

Liver diseases negatively impact the quality of life and survival of patients, and often require liver transplantation in cases that progress to organ failure. Understanding the cellular and molecular mechanisms of liver development and pathogenesis has been a challenging task, in part for the lack of adequate cellular models directly relevant to the human diseases. Recent technological advances in the stem cell field have shown the potentiality of induced pluripotent stem cells (iPSC) and liver organoids as the next generation tool to model in vitro liver diseases. Hepatocyte-like cells and cholangiocyte are currently being generated from skin fibroblasts and mononuclear blood cells reprogrammed into iPSC and have been successfully used for disease modeling, drug testing and gene editing, with the hope to be able to find application also in regenerative medicine. Protocols to generate other liver cell types are still under development, but the field is advancing rapidly. On the other end, liver cells can now be isolated from liver specimens (liver explants or liver biopsies) and cultured in specific conditions to form polarized 3D organoids. The purpose of this review is to summarize all these recent technological advances and their potential applications but also to analyze the current issues to be addressed before the technology can reach its full potential.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Liver Diseases/metabolism , Organoids/metabolism , Drug Evaluation, Preclinical/methods , Gene Editing/methods , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/pathology , Liver Diseases/genetics , Liver Diseases/pathology , Liver Diseases/therapy , Organoids/pathology , Primary Cell Culture/methods
SELECTION OF CITATIONS
SEARCH DETAIL