Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Soc Sci Res ; 98: 102580, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34247725

ABSTRACT

We investigate whether white women, black women, and black men earn less than white men because of 1) lower educational attainment and/or 2) lower wage returns to the same levels and academic fields of attainment. Using the 1979-2012 waves of the American National Longitudinal Survey of Youth (NLSY79), we examine how educational attainment and academic fields of study impact pay. Regression decompositions show that differences in attainment and in academic fields explain 13 to 23 percent of the racial pay gaps, but none of the gender pay gaps. Random effects models test for race and gender differences in the wage returns to education. Men of both races receive higher wage returns relative to women, while black women receive lower returns relative to all groups for master's degrees. Our intersectional approach reveals that equalizing educational attainment would reduce racial pay gaps, whereas equalizing wage returns to education would reduce gender pay disparities. Moreover, black women's earnings are multiply disadvantaged, both by their lower attainment relative to white women, and their lower returns to education relative to all groups studied.


Subject(s)
Income , Salaries and Fringe Benefits , Adolescent , Black People , Educational Status , Female , Humans , Male , Sex Factors , United States
2.
Mol Vis ; 27: 288-299, 2021.
Article in English | MEDLINE | ID: mdl-34012231

ABSTRACT

Purpose: To describe the derivation of photoreceptor precursor cells from human embryonic stem cells by coculture with RPE cells. Methods: Human embryonic stem cells were induced to differentiate into neural precursor cells and then cocultured with RPE cells to obtain cells showing retinal photoreceptor features. Immunofluorescent staining, reverse transcription-PCR (RT-PCR), and microarray analysis were performed to identify photoreceptor markers, and a cGMP assay was used for in vitro functional analysis. After subretinal injection in rat animal models, retinal function was determined with electroretinography and optokinetic response detection, and immunofluorescent staining was performed to assess the survival of the injected cells. Results: Cocultured cells were positive for rhodopsin, red and blue opsin, recoverin, and phosphodiesterase 6 beta on immunofluorescent staining and RT-PCR. Serial detection of stem cell-, neural precursor-, and photoreceptor-specific markers was noted in each stage of differentiation with microarray analysis. Increased cGMP hydrolysis in light-exposed conditions compared to that in dark conditions was observed. After the subretinal injection in the rats, preservation of optokinetic responses was noted up to 20 weeks, while electroretinographic response decreased. Survival of the injected cells was confirmed with positive immunofluorescence staining of human markers at 8 weeks. Conclusions: Cells showed photoreceptor-specific features when stem cell-derived neurogenic precursors were cocultured with RPE cells.


Subject(s)
Human Embryonic Stem Cells/cytology , Photoreceptor Cells/cytology , Retinal Pigment Epithelium/cytology , Stem Cells/cytology , Biomarkers/metabolism , Cell Differentiation/physiology , Coculture Techniques , Electroretinography , Eye Proteins/metabolism , Human Embryonic Stem Cells/metabolism , Humans , Nystagmus, Optokinetic/physiology , Photoreceptor Cells/metabolism , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/metabolism , Stem Cells/metabolism
4.
Int J Mol Sci ; 21(10)2020 May 22.
Article in English | MEDLINE | ID: mdl-32455909

ABSTRACT

This study evaluated the potential of iron oxide nanoparticle-loaded human embryonic stem cell (ESC)-derived spherical neural masses (SNMs) to improve the transportation of stem cells to the brain, ameliorate brain damage from intracerebral hemorrhage (ICH), and recover the functional status after ICH under an external magnetic field of a magnet attached to a helmet. At 24 h after induction of ICH, rats were randomly separated into three experimental groups: ICH with injection of phosphate-buffered saline (PBS group), ICH with intravenous injection of magnetosome-like ferrimagnetic iron oxide nanocubes (FION)-labeled SNMs (SNMs* group), and ICH with intravenous injection of FION-labeled SNMs followed by three days of external magnetic field exposure for targeted delivery by a magnet-embedded helmet (SNMs*+Helmet group). On day 3 after ICH induction, an increased Prussian blue-stained area and decreased swelling volume were observed in the SNMs*+Helmet group compared with that of the other groups. A significantly decreased recruitment of macrophages and neutrophils and a downregulation of pro-inflammatory cytokines followed by improved neurological function three days after ICH were observed in the SNMs*+Helmet group. Hemispheric atrophy at six weeks after ICH was significantly decreased in the SNMs*+Helmet group compared with that of the PBS group. In conclusion, we have developed a targeted delivery system using FION tagged to stem cells and a magnet-embedded helmet. The targeted delivery of SNMs might have the potential for developing novel therapeutic strategies for ICH.


Subject(s)
Brain/drug effects , Cerebral Hemorrhage/drug therapy , Human Embryonic Stem Cells/metabolism , Magnetic Field Therapy/methods , Magnetic Iron Oxide Nanoparticles/chemistry , Recovery of Function/drug effects , Animals , Behavior Rating Scale , Brain/pathology , Brain/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cerebral Hemorrhage/radiotherapy , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Humans , Inflammation/drug therapy , Inflammation/radiotherapy , Injections, Intravenous , Male , Neural Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , Spheroids, Cellular/metabolism
5.
Tissue Eng Part C Methods ; 25(9): 532-542, 2019 09.
Article in English | MEDLINE | ID: mdl-31418341

ABSTRACT

IMPACT STATEMENT: This study describes the methods and results of superparamagnetic iron oxide nanoparticle (SPION) labeling and magnetic resonance imaging (MRI) tracking of human embryonic stem cell-derived photoreceptor precursors transplanted into the subretinal space of Royal College of Surgeons rats. SPION labeling and MRI tracking provide information about the biodistribution of transplanted photoreceptor precursors, which is necessary for improving the functional benefits of cell therapy for degenerative retinal diseases.


Subject(s)
Cell Tracking , Contrast Media , Human Embryonic Stem Cells/metabolism , Magnetic Resonance Imaging , Magnetite Nanoparticles/chemistry , Photoreceptor Cells, Vertebrate , Animals , Cell Line , Contrast Media/chemistry , Contrast Media/pharmacology , Heterografts , Humans , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/transplantation , Rats
6.
Pediatr Gastroenterol Hepatol Nutr ; 22(3): 291-297, 2019 May.
Article in English | MEDLINE | ID: mdl-31110962

ABSTRACT

Bowel ischemia is a life-threatening surgical emergency. We report a case of rapidly progressive bowel necrosis in a previously healthy child without proven mechanical small bowel obstruction. The definite diagnosis was established at the time of an exploratory operation. Of note, imaging studies and even a laparotomy did not reveal any evidence of acute appendicitis or mechanical obstruction such as intussusception or Meckel's diverticulum. During hospitalization, since we could not rule out surgical abdomen after inconclusive image findings, we closely followed the patient and repeated physical examinations carefully. Eventually surgical exploration was performed based on changes in clinical condition, which proved to be the right decision for the patient. We propose that in children with suspected strangulation of small bowel obstruction, especially when imaging findings do not provide a conclusive diagnosis, the timely exploratory surgical approach ought to be chosen based on carefully observed clinical findings and other evaluations.

7.
Int J Stem Cells ; 12(2): 340-346, 2019 Jul 31.
Article in English | MEDLINE | ID: mdl-31023000

ABSTRACT

The concept of cellular reprogramming was developed to generate induced neural precursor cells (iNPCs)/dopaminergic (iDA) neurons using diverse approaches. Here, we investigated the effects of various nanoscale scaffolds (fiber, dot, and line) on iNPC/iDA differentiation by direct reprogramming. The generation and maturation of iDA neurons (microtubule-associated protein 2-positive and tyrosine hydroxylase-positive) and iNPCs (NESTIN-positive and SOX2-positive) increased on fiber and dot scaffolds as compared to that of the flat (control) scaffold. This study demonstrates that nanotopographical environments are suitable for direct differentiation methods and may improve the differentiation efficiency.

8.
J Anesth ; 31(6): 821-828, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28913662

ABSTRACT

PURPOSE: Data from animal experiments suggest that exposure to general anesthetics in early life inhibits neurogenesis and causes long-term memory deficit. Considering short operating times and the popularity of sevoflurane in pediatric anesthesia, it is important to verify the effects of short-period exposure to sevoflurane on the developing brain. METHODS: We measured the effects of short-term exposure (2 h) to 3%, 6%, or 8% sevoflurane, the most commonly used anesthetic, on neural precursor cells derived from human embryonic stem cells, SNUhES32. Cell survival, proliferation, apoptosis, and differentiation on days 1, 3, 5, and 7 post treatment were analyzed. RESULTS: Treatment with 6% sevoflurane increased cell viability (P = 0.046) and decreased apoptosis (P = 0.014) on day 5, but the effect did not persist on day 7. Survival and apoptosis were not affected by 3% and 8% sevoflurane; there was no effect of proliferation at any of the tested concentrations. The differentiation of cells exposed to 6% or 8% sevoflurane decreased on day 1 (P = 0.033 and P = 0.036 for 6% and 8% sevoflurane, respectively) but was again normalized on days 3-7. CONCLUSION: Clinically relevant treatment with sevoflurane for 2 h induces no significant changes in the survival, proliferation, apoptosis, and differentiation of human neural precursor cells, although supraclinical doses of sevoflurane do alter human neurogenesis transiently.


Subject(s)
Anesthetics, Inhalation/administration & dosage , Cell Survival/drug effects , Human Embryonic Stem Cells/drug effects , Methyl Ethers/administration & dosage , Anesthetics, Inhalation/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Humans , Neural Stem Cells/drug effects , Neurons/drug effects , Sevoflurane
9.
J Vis Exp ; (125)2017 07 22.
Article in English | MEDLINE | ID: mdl-28784973

ABSTRACT

Spinal cord ischemia is a fatal complication following thoracoabdominal aortic aneurysm surgery. Researchers can investigate the strategies for preventing and treating this complication using experimental models of spinal cord ischemia. The model described here demonstrates varying degrees of paraplegia that relate to the length of occlusion following thoracic aortic occlusion in a rat spinal cord ischemia model. A 2-Fr. balloon-tipped catheter was advanced through the femoral artery into the descending thoracic aorta until the catheter tip was placed at the left subclavian artery in anesthetized male Sprague-Dawley rats. Spinal cord ischemia was induced by inflating the catheter balloon. After a set period of occlusion (9, 10, or 11 min), the balloon was deflated. Neurologic assessment was performed using the motor deficit index at 24 h after surgery, and the spinal cord was harvested for histopathological examination. Rats that underwent 9 min of aortic occlusion showed mild and reversible motor impairment in the hind limb. Rats subjected to 10 min of aortic occlusion presented with moderate but reversible motor impairment. Rats subjected to 11 min of aortic occlusion displayed complete and persistent paralysis. The motor neurons in the spinal cord sections were more preserved in rats subjected to shorter duration of aortic occlusion. Researchers can achieve a reproducible hind limb motor deficit following thoracic aortic occlusion using this spinal cord ischemia model.


Subject(s)
Hindlimb/physiopathology , Motor Neurons/pathology , Spinal Cord Ischemia/etiology , Animals , Aorta, Thoracic/physiopathology , Arterial Occlusive Diseases/complications , Arterial Occlusive Diseases/physiopathology , Coronary Occlusion/etiology , Disease Models, Animal , Male , Paraplegia/etiology , Rats, Sprague-Dawley , Spinal Cord Ischemia/pathology , Vascular Access Devices
10.
Mol Ther ; 25(9): 2028-2037, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28705346

ABSTRACT

Generation of functional dopamine (DA) neurons is an essential step for the development of effective cell therapy for Parkinson's disease (PD). The generation of DA neurons can be accomplished by overexpression of DA-inducible genes using virus- or DNA-based gene delivery methods. However, these gene delivery methods often cause chromosomal anomalies. In contrast, mRNA-based gene delivery avoids this problem and therefore is considered safe to use in the development of cell-based therapy. Thus, we used mRNA-based gene delivery method to generate safe DA neurons. In this study, we generated transformation-free DA neurons by transfection of mRNA encoding DA-inducible genes Nurr1 and FoxA2. The delivery of mRNA encoding dopaminergic fate inducing genes proved sufficient to induce naive rat forebrain precursor cells to differentiate into neurons exhibiting the biochemical, electrophysiological, and functional properties of DA neurons in vitro. Additionally, the generation efficiency of DA neurons was improved by the addition of small molecules, db-cAMP, and the adjustment of transfection timing. The successful generation of DA neurons using an mRNA-based method offers the possibility of developing clinical-grade cell sources for neuronal cell replacement treatment for PD.


Subject(s)
Dopaminergic Neurons/metabolism , RNA, Messenger/chemical synthesis , RNA, Messenger/genetics , Transcription Factors/genetics , Animals , Cell Line , Dopaminergic Neurons/cytology , Gene Expression , Gene Expression Regulation , Gene Order , Genes, Reporter , Genetic Vectors/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Humans , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Rats , Transfection , Tyrosine 3-Monooxygenase/genetics
11.
Int J Clin Pharmacol Ther ; 54(12): 992-1003, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27668695

ABSTRACT

OBJECTIVE: This study evaluated the possible pharmacokinetic interactions between rosuvastatin and fimasartan, an angiotensin II type 1 (AT1) receptor blocker (ARB), approved in Korea for the treatment of mild to moderate hypertension. METHODS: In this open-label, multiple-dose, two-period, single-sequence study, the enrolled subjects were randomized into two separate parts (A and B). In part A, subjects received 120 mg of fimasartan alone for 7 days during period I, and 120 mg fimasartan with 20 mg rosuvastatin for 7 days during period II. In Part B, subjects received rosuvastatin alone, followed by concomitant administration of fimasartan, with the same doses used as in Part A. There was a 7-day washout between periods I and II. Serial blood samples were collected for up to 48 hours for fimasartan and for up to 72 hours for rosuvastatin after the last dose of each period to determine the steady-state pharmacokinetics of both drugs. RESULTS: The mean Cmax,ss and AUCτ,ss values of fimasartan were 258.03 ± 176.75 ng/mL and 746.52 ± 273.49 ng×h/mL for fimasartan alone, and 289.40 ± 231.44 ng/mL and 848.43 ± 267.45 ng×h/mL for fimasartan and rosuvastatin coadministration, respectively (p-values for Cmax,ss and AUCτ,ss, 0. 513 and 0.006, respectively). The mean Cmax,ss and AUCτ,ss values of rosuvastatin were 9.94 ± 4.48 ng/mL and 85.29 ± 36.25 ng×h/mL for rosuvastatin alone and 11.94 ± 8.47 ng/mL and 77.33 ± 38.71 ng×h/mL for fimasartan and rosuvastatin coadministration, respectively (p-values for Cmax,ss and AUCτ,ss, 0.066 and 0.009, respectively). The geometric mean ratio (GMR) and 90% confidence intervals (CI) for the Cmax,ss and AUCτ,ss of fimasartan (with/without rosuvastatin) were 1.109 (0.813 - 1.511) and 1.159 (1.061 - 1.265), respectively. The GMR and 90% CI for the Cmax,ss and AUCτ,ss of rosuvastatin (with/without fimasartan) were 1.090 (0.979 - 1.213) and 0.870 (0.804 - 0.940), respectively. CONCLUSIONS: These results suggest that fimasartan and rosuvastatin have no relevant pharmacokinetic drug-drug interactions. All treatments were well tolerated during this study, with no serious adverse effects.
.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Biphenyl Compounds/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Rosuvastatin Calcium/pharmacokinetics , Tetrazoles/pharmacokinetics , Adult , Area Under Curve , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacology , Drug Interactions , Healthy Volunteers , Humans , Male , Middle Aged , Pyrimidines/adverse effects , Pyrimidines/pharmacology , Rosuvastatin Calcium/adverse effects , Rosuvastatin Calcium/pharmacology , Tetrazoles/adverse effects , Tetrazoles/pharmacology
12.
Phytother Res ; 30(12): 2036-2043, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27573551

ABSTRACT

Estrogen receptor (ER)α-positive breast cancer cells regulate the expression of estrogen-responsive genes, which are involved in cell proliferation, differentiation, and cell cycle progression. Clinically, the inhibition of ERα-mediated gene expression in breast cancer cells has long been considered an effective way to prevent the development and progression of cancer. Germacrone, a terpenoid compound isolated from Rhizoma curcuma, has been known to have antitumor activity in various human cancer cell lines. However, the mechanism by which germacrone inhibits the proliferation of breast cancer cells is still unclear. Here, we demonstrated that germacrone inhibits ERα-mediated gene expression at the transcriptional level in MCF-7 cells. Germacrone inhibits the recruitment of ERα to the estrogen response element on chromatin and consequently compromises the binding of switch/sucrose non-fermentable chromatin remodeling complex and RNA polymerase II to target gene promoter, thereby inhibiting the estrogen-induced chromatin accessibility. In addition, germacrone efficiently potentiates the antitumor activity of methotrexate and 5-fluorouracil. Our results not only provide substantial molecular mechanism of germacrone on ERα-mediated signaling in breast cancer cells but also demonstrate the benefits of germacrone as a combination therapy with other drugs for the treatment of breast cancer. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Sesquiterpenes, Germacrane/chemistry , Cell Line, Tumor , Cell Proliferation , Drugs, Chinese Herbal/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Sesquiterpenes, Germacrane/pharmacology , Signal Transduction
13.
Clin Drug Investig ; 36(8): 613-23, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27206575

ABSTRACT

BACKGROUND AND OBJECTIVE: Combination therapy is recommended for the effective management of hypertension according to most treatment guidelines, including those of the US Joint National Committee. Therefore, pharmacokinetic drug interactions are an important issue in combination therapy for hypertension. In this study, the pharmacokinetic properties of telmisartan and chlorthalidone were evaluated to investigate their pharmacokinetic interactions in healthy subjects. METHODS: Two separate, randomized, multiple-dose, two-period, one-sequence studies were conducted. In study A, 43 participants received 80 mg of telmisartan orally for 7 days, and were then administered oral chlorthalidone 25 mg for 14 days (days 8-21), coadministered with 80 mg of telmisartan from day 15. In study B, 14 participants received oral chlorthalidone (25 mg) for 13 days, followed by coadministration with 80 mg of telmisartan orally for 7 days. RESULTS: The geometric mean ratios (GMRs) (90 % confidence intervals [CIs]) of the maximum plasma concentration (C max,ss) and area under the concentration-time curve for the dosing interval at steady state (AUCτ,ss) of telmisartan (with and without chlorthalidone) were 1.018 (0.861-1.203) and 1.099 (1.015-1.190), respectively. For chlorthalidone (with/without telmisartan), the GMRs (90 % CIs) for C max,ss and AUCτ,ss were 0.996 (0.922-1.075) and 0.992 (0.925-1.064), respectively. The GMRs and 90 % CIs for telmisartan and chlorthalidone were all within the 0.80-1.25 range. CONCLUSION: Thus, in this study, there was no significant pharmacokinetic interaction between telmisartan and chlorthalidone. CLINICALTRIAL. GOV IDENTIFIER: NCT01806363.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Benzimidazoles/pharmacokinetics , Benzoates/pharmacokinetics , Chlorthalidone/pharmacokinetics , Diuretics/pharmacokinetics , Adult , Angiotensin II Type 1 Receptor Blockers/adverse effects , Antihypertensive Agents/adverse effects , Area Under Curve , Benzimidazoles/adverse effects , Benzoates/adverse effects , Chlorthalidone/adverse effects , Diuretics/adverse effects , Drug Interactions , Healthy Volunteers , Humans , Male , Middle Aged , Telmisartan , Young Adult
14.
Cell Signal ; 28(9): 1137-1144, 2016 09.
Article in English | MEDLINE | ID: mdl-27234130

ABSTRACT

Follistatin-like 1 (FSTL1) functions as a pivotal modulator of inflammation and is implicated in many inflammatory diseases such as rheumatoid arthritis. Here, we report that FSTL1 is strongly upregulated and secreted during osteoclast differentiation of bone marrow-derived macrophages (BMMs) and that FSTL1 positively regulates osteoclast formation induced by RANKL and M-CSF. The overexpression of FSTL1 or treatment with recombinant FSTL1 (rFSTL1) in BMMs enhances the formation of multinuclear osteoclasts and the induction of c-Fos and NFATc1, transcription factors important for osteoclastogenesis. Conversely, knockdown of FSTL1 using a small hairpin RNA suppresses osteoclast formation and the expression of these transcription factors. While FSTL1 does not affect RANKL-stimulated activation of p38 MAPK, phosphorylation of IκBα, JNK, and ERK were increased by overexpression or addition of rFSTL1. Furthermore, rFSTL1 increased RANKL-induced NF-κB transcriptional activity in a dose-dependent manner. In addition to its role in osteoclastogenesis, FSTL1 promotes proliferation of osteoclast precursors by increasing M-CSF-induced ERK activation, which in turn leads to accelerated osteoclast formation. Together, our findings demonstrate that FSTL1 is a secreted osteoclastogenic factor that plays a critical role in osteoclast formation via the NF-κB and MAPKs signaling pathways.


Subject(s)
Follistatin-Related Proteins/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , NF-kappa B/metabolism , Osteoclasts/metabolism , RANK Ligand/pharmacology , Stem Cells/cytology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Follistatin-Related Proteins/pharmacology , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Osteoclasts/drug effects , Osteogenesis/drug effects , RAW 264.7 Cells , Recombinant Proteins/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Up-Regulation/drug effects
15.
J Bone Metab ; 23(1): 8-15, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26981515

ABSTRACT

BACKGROUND: Lipocalin-2 (LCN2), a small glycoprotein, has a pivotal role in diverse biological processes such as cellular proliferation and differentiation. We previously reported that LCN2 is implicated in osteoclast formation induced by receptor activator of nuclear factor-kappa B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF). In the present study, we used a knockout mouse model to further investigate the role of LCN2 in osteoclast development. METHODS: Osteoclastogenesis was assessed using primary bone marrow-derived macrophages. RANKL and M-CSF signaling was determined by immunoblotting, cell proliferation by bromodeoxyuridine (BrdU) enzyme-linked immunosorbent assay (ELISA), and apoptosis by cell death detection ELISA. Bone morphometric parameters were determined using a micro-computed tomography system. RESULTS: Our results showed that LCN2 deficiency increases tartrate-resistant acid phosphatase (TRAP)-positive multinucleated osteoclast formation in vitro, a finding that reflects enhanced proliferation and differentiation of osteoclast lineage cells. LCN2 deficiency promotes M-CSF-induced proliferation of bone marrow macrophages (BMMs), osteoclast precursors, without altering their survival. The accelerated proliferation of LCN2-deficient precursors is associated with enhanced expression and activation of the M-CSF receptor, c-Fms. Furthermore, LCN2 deficiency stimulates the induction of c-Fos and nuclear factor of activated T cells c1 (NFATc1), key transcription factors for osteoclastogenesis, and promotes RANKL-induced inhibitor of kappa B (IκBα) phosphorylation. Interestingly, LCN2 deficiency does not affect basal osteoclast formation in vivo, suggesting that LCN2 might play a role in the enhanced osteoclast development that occurs under some pathological conditions. CONCLUSIONS: Our study establishes LCN2 as a negative modulator of osteoclast formation, results that are in accordance with our previous findings.

16.
J Cell Physiol ; 231(4): 844-51, 2016 04.
Article in English | MEDLINE | ID: mdl-26280807

ABSTRACT

G protein-coupled receptor 120 (GPR120) plays an important role in the regulation of inflammation and lipid metabolism. In this study, we investigated the role of GPR120 in osteoclast development and found that GPR120 regulates osteoclast differentiation, survival and function. We observed that GPR120 was highly expressed in osteoclasts compared to their precursors, bone marrow-derived macrophages (BMMs). Activation of GPR120 by its ligand GW9508 suppressed receptor activator of NF- κB ligand (RANKL)-induced osteoclast differentiation and the expression of nuclear factor of activated T cells c1 (NFATc1), a key modulator of osteoclastogenesis. GPR120 activation further inhibited the RANKL-stimulated phosphorylation of IκBα and JNK. In addition to osteoclast differentiation, GPR120 activation increased the apoptosis of mature osteoclasts by inducing caspase-3 and Bim expression. Activation of GPR120 also interfered with cell spreading and actin cytoskeletal organization mediated by M-CSF but not by RANKL. Coincident with the impaired cytoskeletal organization, GPR120 activation blocked osteoclast bone resorbing activity. Furthermore, knockdown of GPR120 using small hairpin RNA abrogated all these inhibitory effects on osteoclast differentiation, survival, and function. Together, our findings identify GPR120 as a negative modulator of osteoclast development that may be an attractive therapeutic target for bone-destructive diseases. J. Cell. Physiol. 231: 844-851, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Cell Differentiation , Osteoclasts/cytology , Osteoclasts/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Apoptosis/drug effects , Apoptosis/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation/drug effects , Cell Survival/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Osteogenesis/drug effects , RANK Ligand/pharmacology , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
17.
Stem Cells ; 34(4): 888-901, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26701067

ABSTRACT

Neurogenesis occurs spontaneously in the subventricular zone (SVZ) of the lateral ventricle in adult rodent brain, but it has long been debated whether there is sufficient adult neurogenesis in human SVZ. Subcallosal zone (SCZ), a posterior continuum of SVZ closely associated with posterior regions of cortical white matter, has also been reported to contain adult neural stem cells (aNSCs) in both rodents and humans. However, little is known whether SCZ-derived aNSC (SCZ-aNSCs) can produce cortical neurons following brain injury. We found that SCZ-aNSCs exhibited limited neuronal differentiation potential in culture and after transplantation in mice. Neuroblasts derived from SCZ initially migrated toward injured cortex regions following brain injury, but later exhibited apoptosis. Overexpression of anti-apoptotic bcl-xL in the SCZ by retroviral infection rescued neuroblasts from cell death in the injured cortex, but neuronal maturation was still limited, resulting in atrophy. In combination with Bcl-xL, infusion of brain-derived neurotropic factor rescued atrophy, and importantly, a subset of such SCZ-aNSCs differentiated and attained morphological and physiological characteristics of mature, excitatory neurons. These results suggest that the combination of anti-apoptotic and neurotrophic factors might enable the use of aNSCs derived from the SCZ in cortical neurogenesis for neural replacement therapy.


Subject(s)
Brain Injuries/therapy , Cell Differentiation/genetics , Neural Stem Cells/transplantation , Neurogenesis/genetics , Adult Stem Cells/transplantation , Animals , Apoptosis , Brain Injuries/pathology , Cell Proliferation/genetics , Humans , Mice , Neurons/pathology , Prefrontal Cortex
18.
Tissue Eng Regen Med ; 13(5): 554-559, 2016 Oct.
Article in English | MEDLINE | ID: mdl-30603436

ABSTRACT

The direct lineage conversion of fibroblasts into neuronal or neural precursor cells (NPCs) has become a hot issue in recent years as an attractive approach in the field of stem cell regenerative medicine. In this study, we adopted the stromal feeder co-culture method during the early conversion period to enhance conversion efficiency. Stromal cells are often used in directed differentiation of dopaminergic (DA) neurons from pluripotent stem cells. We co-cultured rat embryonic fibroblasts (REFs) on γ-irradiated sonic hedgehog-overexpressing MS5 stromal (MS5-SHH) cells after transduction with Brn2, Ascl1, Myt1L, and BclxL-GFP (BAMXGFP) transcription factors to REFs. One week after co-culture, transduced cells (GFP+ cells) that proliferated on MS5-SHH cells were separated from MS5-SHH cells through a 40 µm cell strainer. Subsequently, the converted cells (GFP+ cells) were expanded on fibronectin-coated culture plates in NPC expansion medium. The induced NPCs (iNPCs) expressed NPC potential (NESTIN+/SOX2+) earlier than seen with non-co-culture methods and were efficiently differentiated into DA neurons by overexpression of Nurr1 and Foxa2 genes, which are specific transcription factors for midbrain DA neuron development. These observations indicated that direct conversion to NPCs using an MS5 stromal cells co-culture method is a suitable technique for efficient generation of iNPC/DA neurons from fibroblasts.

19.
Int J Stem Cells ; 8(2): 228-34, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26634071

ABSTRACT

Induced neural precursor cells (iNPCs) are one source of transplantable dopaminergic neurons used in cell therapy for Parkinson's disease. In the present study, we demonstrate that iNPCs can be generated by transducing Brn2, Ascl1, Myt1L and Bcl-xL in a culture supplemented with several mitogens and subsequently can be differentiated to dopaminergic neurons (DA). However, studies have shown that iDA and/or iNPC-derived DA neurons using various conversion protocols have low efficiency. Here, we show that early exposure of FGF8 to fibroblasts efficiently improves differentiation of DA neurons. So our study demonstrates that FGF8 is a critical factor for generation of iNPC-derived DA neurons.

20.
PLoS One ; 10(9): e0138460, 2015.
Article in English | MEDLINE | ID: mdl-26383864

ABSTRACT

Directed methods for differentiating human embryonic stem cells (hESCs) into dopaminergic (DA) precursor cells using stromal cells co-culture systems are already well established. However, not all of the hESCs differentiate into DA precursors using these methods. HSF6, H1, H7, and H9 cells differentiate well into DA precursors, but CHA13 and CHA15 cells hardly differentiate. To overcome this problem, we modified the differentiation system to include a co-culturing step that exposes the cells to noggin early in the differentiation process. This was done using γ-irradiated noggin-overexpressing CF1-mouse embryonic fibroblasts (MEF-noggin) and MS5 stromal cells (MS5-noggin and MS5-sonic hedgehog). After directed differentiation, RT-PCR analyses revealed that engrailed-1 (En-1), Lmx1b, and Nurr1, which are midbrain DA markers, were expressed regardless of differentiation stage. Moreover, tyrosine hydroxylase (Th) and an A9 midbrain-specific DA marker (Girk2) were expressed during differentiation, whereas levels of Oct3/4, an undifferentiated marker, decreased. Immunocytochemical analyses revealed that protein levels of the neuronal markers TH and TuJ1 increased during the final differentiation stage. These results demonstrate that early noggin exposure may play a specific role in the directed differentiation of DA cells from human embryonic stem cells.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation/genetics , Dopaminergic Neurons/metabolism , Fibroblasts/metabolism , Human Embryonic Stem Cells/metabolism , Stromal Cells/metabolism , Animals , Carrier Proteins/genetics , Coculture Techniques , Dopaminergic Neurons/cytology , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Fibroblasts/cytology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Human Embryonic Stem Cells/cytology , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Stromal Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...