Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Neurotoxicol Teratol ; 83: 106943, 2021.
Article in English | MEDLINE | ID: mdl-33221301

ABSTRACT

Prenatal ethanol exposure can result in loss of neural stem cells (NSCs) and decreased brain growth. Here, we assessed whether a noncoding RNA (ncRNA) related to the NSC self-renewal factor Oct4/Pou5f1, and transcribed from a processed pseudogene locus on mouse chromosome 9 (mOct4pg9), contributed to the loss of NSCs due to ethanol. Mouse fetal cortical-derived NSCs, cultured ex vivo to mimic the early neurogenic environment of the fetal telencephalon, expressed mOct4pg9 ncRNA at significantly higher levels than the parent Oct4/Pou5f1 mRNA. Ethanol exposure increased expression of mOct4pg9 ncRNA, but decreased expression of Oct4/Pou5f1. Gain- and loss-of-function analyses indicated that mOct4pg9 overexpression generally mimicked effects of ethanol exposure, resulting in increased proliferation and expression of transcripts associated with neural maturation. Moreover, mOct4pg9 associated with Ago2 and with miRNAs, including the anti-proliferative miR-328-3p, whose levels were reduced following mOct4pg9 overexpression. Finally, mOct4pg9 inhibited Oct4/Pou5f1-3'UTR-dependent protein translation. Consistent with these observations, data from single-cell transcriptome analysis showed that mOct4pg9-expressing progenitors lack Oct4/Pou5f1, but instead overexpress transcripts for increased mitosis, suggesting initiation of transit amplification. Collectively, these data suggest that the inhibitory effects of ethanol on brain development are explained, in part, by a novel ncRNA which promotes loss of NSC identity and maturation.


Subject(s)
Ethanol/toxicity , Fetal Stem Cells/drug effects , Neural Stem Cells/drug effects , Octamer Transcription Factor-3/genetics , RNA, Untranslated/genetics , Animals , Argonaute Proteins/metabolism , Brain/drug effects , Brain/embryology , Brain/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Disease Models, Animal , Female , Fetal Alcohol Spectrum Disorders/genetics , Fetal Alcohol Spectrum Disorders/metabolism , Fetal Alcohol Spectrum Disorders/pathology , Fetal Stem Cells/metabolism , Fetal Stem Cells/pathology , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Models, Neurological , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/drug effects , Neurogenesis/genetics , Octamer Transcription Factor-3/antagonists & inhibitors , Octamer Transcription Factor-3/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Pseudogenes , RNA, Untranslated/metabolism , Single-Cell Analysis
2.
J Pathol ; 248(2): 155-163, 2019 06.
Article in English | MEDLINE | ID: mdl-30680725

ABSTRACT

The cellular mechanisms by which hepatitis C virus (HCV) replication might mediate cytopathic effects are controversial and not entirely clear. In this study, we found that blood-borne HCV (bbHCV) infection could lead to endoplasmic reticulum (ER)-stress and mitochondria-related/caspase-dependent apoptosis at the early stages of infection based on use of the highly efficient bbHCV cell culture model established previously. Sections of bbHCV-infected human fetal liver stem cells (hFLSCs) revealed convolution and nonlinear ER, cell vacuolization, swelling of mitochondria, and numerous double membrane vesicles (DMVs). The percentage of apoptotic hFLSCs infected by bbHCV reached 29.8% at 16 h postinfection, and the amount of cytochrome c increased remarkably in the cytosolic protein fraction. However, over time, apoptosis was inhibited due to the activation of NF-κB. The expression of NF-κB-p65, Bcl-xL, XIAP, and c-FLIPL in hFLSCs was increased significantly 24 h after in infection by bbHCV. The accelerated cell death cycles involving apoptosis, regeneration and repair by bbHCV infection might give rise to the development of cirrhosis, and ultimately to hepatocellular carcinogenesis. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Fetal Stem Cells/pathology , Hepacivirus/growth & development , Hepatitis C, Chronic/virology , Liver/pathology , Virus Replication , Apoptosis Regulatory Proteins/metabolism , Cell Line , Fetal Stem Cells/metabolism , Fetal Stem Cells/virology , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/pathology , Humans , Liver/metabolism , Liver/virology , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Mitochondria, Liver/virology , Oxidative Stress , Signal Transduction
3.
Stem Cell Rev Rep ; 14(5): 632-641, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29948753

ABSTRACT

Chimerism occurs naturaly throughout gestation and can also occur as a consequence of transfusion and transplantation therapy. It consists of the acquisition and long-term persistence of a genetically distinct population of allogenic cells inside another organism. Previous reports have suggested that feto-maternal microchimerism could exert a beneficial effect on the treatment of hematological and solid tumors in patients treated by PBSCT. In this review we report the mechanism of transplacental fetal stem cell trafficking during pregnancy and the effect of their long-term persistence on autoimmunity, GVHD, PBSCT, cancer and stem cell treatment.


Subject(s)
Autoimmune Diseases/physiopathology , Chimerism , Fetal Stem Cells/physiology , Female , Fetal Stem Cells/pathology , Fetal Stem Cells/transplantation , Fetus/cytology , Fetus/physiology , Humans , Placenta/cytology , Placenta/physiology , Pregnancy
4.
Nat Cell Biol ; 19(6): 614-625, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28504706

ABSTRACT

Adult and fetal haematopoietic stem cells (HSCs) display a glycolytic phenotype, which is required for maintenance of stemness; however, whether mitochondrial respiration is required to maintain HSC function is not known. Here we report that loss of the mitochondrial complex III subunit Rieske iron-sulfur protein (RISP) in fetal mouse HSCs allows them to proliferate but impairs their differentiation, resulting in anaemia and prenatal death. RISP-null fetal HSCs displayed impaired respiration resulting in a decreased NAD+/NADH ratio. RISP-null fetal HSCs and progenitors exhibited an increase in both DNA and histone methylation associated with increases in 2-hydroxyglutarate (2HG), a metabolite known to inhibit DNA and histone demethylases. RISP inactivation in adult HSCs also impaired respiration resulting in loss of quiescence concomitant with severe pancytopenia and lethality. Thus, respiration is dispensable for adult or fetal HSC proliferation, but essential for fetal HSC differentiation and maintenance of adult HSC quiescence.


Subject(s)
Adult Stem Cells/metabolism , Cell Proliferation , Electron Transport Complex III/metabolism , Energy Metabolism , Fetal Stem Cells/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Mitochondria/metabolism , Adult Stem Cells/pathology , Anemia/blood , Anemia/genetics , Animals , Cell Death , Cells, Cultured , Cellular Senescence , Electron Transport , Electron Transport Complex III/deficiency , Electron Transport Complex III/genetics , Epigenesis, Genetic , Female , Fetal Stem Cells/pathology , Genotype , Glutarates/metabolism , Hematopoietic Stem Cells/pathology , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/pathology , NAD/metabolism , Phenotype , Pregnancy , Signal Transduction , Time Factors
5.
Alcohol ; 60: 149-158, 2017 05.
Article in English | MEDLINE | ID: mdl-28438527

ABSTRACT

Fetal alcohol spectrum disorders are a leading cause of intellectual disability worldwide. Previous studies have shown that developmental ethanol exposure results in loss of microRNAs (miRNAs), including miR-9, and loss of these miRNAs, in turn, mediates some of ethanol's teratogenic effects in the developing brain. We previously found that ethanol increased methylation at the miR-9-2 encoding gene locus in mouse fetal neural stem cells (NSC), advancing a mechanism for epigenetic silencing of this locus and consequently, miR-9 loss in NSCs. Therefore, we assessed the role of the BAF (BRG1/BRM-Associated Factor) complex, which disassembles nucleosomes to facilitate access to chromatin, as an epigenetic mediator of ethanol's effects on miR-9. Chromatin immunoprecipitation and DNAse I-hypersensitivity analyses showed that the BAF complex was associated with both transcriptionally accessible and heterochromatic regions of the miR-9-2 locus, and that disintegration of the BAF complex by combined knockdown of BAF170 and BAF155 resulted in a significant decrease in miR-9. We hypothesized that ethanol exposure would result in loss of BAF-complex function at the miR-9-2 locus. However, ethanol exposure significantly increased mRNA transcripts for maturation-associated BAF-complex members BAF170, SS18, ARID2, BAF60a, BRM/BAF190b, and BAF53b. Ethanol also significantly increased BAF-complex binding within an intron containing a CpG island and in the terminal exon encoding precursor (pre)-miR-9-2. These data suggest that the BAF complex may adaptively respond to ethanol exposure to protect against a complete loss of miR-9-2 in fetal NSCs. Chromatin remodeling factors may adapt to the presence of a teratogen, to maintain transcription of critical miRNA regulatory pathways.


Subject(s)
Chromatin Assembly and Disassembly/drug effects , DNA Helicases/metabolism , Ethanol/toxicity , Fetal Stem Cells/drug effects , Gene Expression Regulation, Developmental/drug effects , MicroRNAs/metabolism , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription, Genetic/drug effects , Animals , Cells, Cultured , DNA Helicases/genetics , Fetal Alcohol Spectrum Disorders/genetics , Fetal Alcohol Spectrum Disorders/metabolism , Fetal Alcohol Spectrum Disorders/pathology , Fetal Stem Cells/metabolism , Fetal Stem Cells/pathology , Mice , MicroRNAs/genetics , Multiprotein Complexes , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/genetics , Nuclear Proteins/genetics , RNA Interference , Time Factors , Transcription Factors/metabolism , Transfection
6.
Exp Clin Transplant ; 15(2): 231-234, 2017 04.
Article in English | MEDLINE | ID: mdl-26788941

ABSTRACT

Diabetes mellitus is a disease with no definite cure. In recent years, stem cell transplant has led to treatment of various diseases including diabetes. We sought to report a type 1 diabetic patient with a brain mass, diagnosed as transitional meningioma, after a fetal hematopoietic stem cell transplant. A 57-year-old woman with type 1 diabetes who previously had undergone a fetal hematopoietic stem cell transplant, attended the clinic with a history of progressive bifrontal headaches accompanied by nausea, vomiting, and visual disturbances over the previous 8 months. Investigations revealed a 2-cm mass in the right temporal region. The patient underwent a craniotomy, and the lesion was removed and sent for pathological and genetic investigations. The results indicated transitional meningioma with the origin of transplanted fetal hematopoietic stem cells. To our knowledge, this is the first report of transitional meningioma as a result of stem cell transplant. Despite all unanswered questions about the safety of stem cell transplant, this novel therapy provides hope for patients with type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Fetal Stem Cells/transplantation , Hematopoietic Stem Cell Transplantation/adverse effects , Meningeal Neoplasms/etiology , Meningioma/etiology , Craniotomy , Diabetes Mellitus, Type 1/diagnosis , Female , Fetal Stem Cells/pathology , Humans , Magnetic Resonance Imaging , Meningeal Neoplasms/diagnosis , Meningeal Neoplasms/surgery , Meningioma/diagnosis , Meningioma/surgery , Middle Aged , Treatment Outcome , Tumor Burden
7.
Placenta ; 36(4): 410-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25684656

ABSTRACT

INTRODUCTION: Gestational diabetes (GDM) is associated with long-term cardiovascular and metabolic diseases in offspring. However, the mechanisms are not well understood. We explored whether fetal exposure to a diabetic environment is associated with fetal endothelial progenitor cell dysfunction, and whether vitamin D can reverse the impairment. METHODS: Nineteen women with uncomplicated pregnancies and 18 women with GDM were recruited before delivery. Time to first appearance of endothelial colony forming cell (ECFC) colonies and number of ECFC colonies formed from culture of cord peripheral blood mononuclear cells were determined. Angiogenesis-related functions of ECFCs in vitro were tested in the presence or absence of vitamin D. RESULTS: Fetal ECFCs from GDM pregnancies formed fewer colonies in culture (P = 0.04) and displayed reduced proliferation (P = 0.02), migration (P = 0.04) and tubule formation (P = 0.03) compared to uncomplicated pregnancies. Fetal ECFCs exposed to hyperglycemia in vitro exhibited less migration (P < 0.05) and less tubule formation (P < 0.05) than normoglycemic control. Vitamin D significantly improved the dysfunction of fetal ECFCs from pregnancies complicated by GDM or after exposure of healthy ECFCs to hyperglycemia. DISCUSSION: Fetal ECFCs from GDM pregnancies or ECFCs exposed to hyperglycemia in vitro exhibit reduced quantity and impaired angiogenesis-related functions. Vitamin D significantly rescues these functions. These findings may have implications for vascular function of infants exposed to a diabetic intrauterine environment.


Subject(s)
Calcitriol/metabolism , Diabetes, Gestational/metabolism , Diabetic Angiopathies/etiology , Endothelium, Vascular/metabolism , Fetal Stem Cells/metabolism , Neovascularization, Pathologic/etiology , Systemic Vasculitis/etiology , Adult , Cell Movement , Cell Proliferation , Cells, Cultured , Colony-Forming Units Assay , Diabetes, Gestational/immunology , Diabetes, Gestational/pathology , Diabetes, Gestational/physiopathology , Diabetic Angiopathies/prevention & control , Dietary Supplements , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Female , Fetal Blood , Fetal Stem Cells/immunology , Fetal Stem Cells/pathology , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Neovascularization, Pathologic/prevention & control , Pregnancy , RNA Interference , Receptors, Calcitriol/agonists , Receptors, Calcitriol/antagonists & inhibitors , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Retrospective Studies , Systemic Vasculitis/prevention & control , Vitamin D/metabolism , Vitamin D/therapeutic use
8.
Carcinogenesis ; 36(1): 94-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25398884

ABSTRACT

Women born from a preeclamptic (PE) pregnancy are associated with a lower risk of breast cancer. Prenatal and early-life exposures are hypothesized to influence breast cancer susceptibility through their effect on stem cells. We examined stem cell populations in umbilical cord blood from PE pregnancies and compared with those from pregnancies without this condition. We isolated mononuclear cells from 58 PE and 197 normotensive (non-PE) umbilical cord blood samples and examined the different stem cell populations. Hematopoietic (CD34(+) and CD34(+)CD38(-)), endothelial (CD34(+)CD133(+), CD34(+)VEGFR2(+), CD133(+)VEGFR2(+) and CD34(+)CD133(+)VEGFR2(+)), and putative breast (EpCAM(+), EpCAM(+)CD49f(+), EpCAM(+)CD49f(+)CD117(+), CD49f(+)CD24(+), CD24(+)CD29(+) and CD24(+)CD29(+)CD49f(+)) stem/progenitor cell subpopulations were quantified by flow cytometry and compared between PE and non-PE samples. Hematopoietic CD34(+) cell counts were significantly lowered in PE compared with non-PE samples (P = 0.039, Kruskal-Wallis test). Levels of CD34(+)CD133(+) endothelial progenitor cells were also lower in PE samples (P = 0.032, multiple regression analysis). EpCAM(+) and EpCAM(+)CD49f(+) putative breast stem cell levels were significantly lowered in PE subjects (multiple regression analysis: P = 0.038 and 0.007, respectively). Stratifying by newborn gender, EpCAM(+) and EpCAM(+)CD49f(+) stem cells were significantly lowered in PE samples of female, but not male, newborns. Umbilical cord blood samples from pregnancies complicated by preeclampsia thus had significantly lower levels of hematopoietic, endothelial, and putative breast stem cells than non-PE controls. With a lowered breast cancer risk for offspring of a PE pregnancy, our findings provide support to the hypothesis that susceptibility to breast oncogenesis may be affected by conditions and processes during the prenatal period.


Subject(s)
Breast Neoplasms/pathology , Fetal Blood/cytology , Fetal Stem Cells/pathology , Pre-Eclampsia/physiopathology , Adolescent , Adult , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cells, Cultured , Disease Susceptibility , Female , Fetal Blood/metabolism , Fetal Stem Cells/metabolism , Flow Cytometry , Humans , Infant, Newborn , Male , Pregnancy , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
9.
J Hepatol ; 62(5): 1085-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25529619

ABSTRACT

BACKGROUND & AIMS: During pregnancy, acetaminophen is one of the very few medications recommended by physicians to treat fever or pain. Recent insights from epidemiological studies suggest an association between prenatal acetaminophen medication and an increased risk for development of asthma in children later in life. The underlying pathogenesis of such association is still unknown. METHODS: We aimed to develop a mouse model to provide insights into the effect of prenatal acetaminophen on maternal, fetal and adult offspring's health. The toxic effect of acetaminophen was studied in mice on 1) maternal liver; mirrored by biomarkers of liver injury, centrilobular necrosis, and infiltration of granulocytes; 2) fetal liver; reflected by the frequency of hematopoietic stem cells, and 3) postnatal health; evaluated by the severity of allergic airway inflammation among offspring. RESULTS: We observed an increased susceptibility towards acetaminophen-induced liver damage in pregnant mice compared to virgins. Moreover, hematopoietic stem cell frequency in fetal liver declined in response to acetaminophen. Furthermore, a greater severity of airway inflammation was observed in offspring of dams upon prenatal acetaminophen treatment, identified lung infiltration by leukocytes and eosinophil infiltration into the airways. CONCLUSION: Our newly developed mouse model on prenatal use of acetaminophen reflects findings from epidemiological studies in humans. The availability of this model will allow improvement in our understanding of how acetaminophen-related hepatotoxicity is operational in pregnant individuals and how an increased risk for allergic diseases in response to prenatal acetaminophen is mediated. Such insights, once available, may change the recommendations for prenatal acetaminophen use.


Subject(s)
Acetaminophen , Asthma , Chemical and Drug Induced Liver Injury , Fetal Stem Cells , Prenatal Exposure Delayed Effects , Acetaminophen/administration & dosage , Acetaminophen/adverse effects , Adult , Adult Children , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/adverse effects , Animals , Asthma/etiology , Asthma/physiopathology , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/physiopathology , Chemical and Drug Induced Liver Injury/prevention & control , Disease Models, Animal , Female , Fetal Stem Cells/drug effects , Fetal Stem Cells/pathology , Humans , Inflammation/etiology , Inflammation/physiopathology , Liver/drug effects , Liver/pathology , Male , Mice , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/physiopathology , Prenatal Exposure Delayed Effects/prevention & control , Severity of Illness Index
10.
Endocr J ; 61(4): 311-20, 2014.
Article in English | MEDLINE | ID: mdl-24452016

ABSTRACT

Thyroid cancer cells were believed to be generated by multi-step carcinogenesis, in which cancer cells are derived from thyrocytes, via multiple incidences of damage to their genome, especially in oncogenes or anti-oncogenes that accelerate proliferation or foster malignant phenotypes, such as the ability to invade the surrounding tissue or metastasize to distant organs, until a new hypothesis, fetal cell carcinogenesis, was presented. In fetal cell carcinogenesis, thyroid tumor cells are assumed to be derived from three types of fetal thyroid cell which only exist in fetuses or young children, namely, thyroid stem cells (TSCs), thyroblasts and prothyrocytes, by proliferation without differentiation. Genomic alternations, such as RET/PTC and PAX8-PPARγ1 rearrangements and a mutation in the BRAF gene, play an oncogenic role by preventing thyroid fetal cells from differentiating. Fetal cell carcinogenesis effectively explains recent molecular and clinical evidence regarding thyroid cancer, including thyroid cancer initiating cells (TCICs), and it underscores the importance of identifying a stem cells and clarifying the molecular mechanism of organ development in cancer research. It introduces three important concepts, the reverse approach, stem cell crisis and mature and immature cancers. Further, it implies that analysis of a small population of cells in a cancer tissue will be a key technique in establishing future laboratory tests. In the contrary, mass analysis such as gene expression profiling, whole genomic scan, and proteomics analysis may have definite limitations since they can only provide information based on many cells.


Subject(s)
Adult Stem Cells/pathology , Carcinogenesis , Fetal Stem Cells/pathology , Models, Biological , Neoplastic Stem Cells/pathology , Thyroid Gland/pathology , Thyroid Neoplasms/pathology , Animals , Cell Proliferation , Humans , Organogenesis , Thyroid Gland/embryology , Thyroid Neoplasms/physiopathology
11.
J Clin Endocrinol Metab ; 98(12): 4953-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24106289

ABSTRACT

CONTEXT: Endothelial colony-forming cells (ECFCs) are the only putative endothelial progenitor cells capable of vasculogenesis, and their dysfunction may represent a risk factor for cardiovascular disease. Intrauterine growth restriction (IUGR) is a pregnancy-related disorder associated with long-term cardiovascular risk. OBJECTIVE: Our objective was to determine whether ECFCs derived from pregnancies complicated by IUGR exhibit altered vasculogenic potential. DESIGN AND SETTING: This was a prospective cohort study; patients were recruited at St. Mary's Hospital, Manchester, United Kingdom. PARTICIPANTS: Twenty-three women with normal pregnancies and 13 women with IUGR-complicated pregnancies at gestational ages above 37 weeks were included. MAIN OUTCOME MEASURES: Vasculogenic capacity of rigorously characterized ECFCs was investigated in vivo by measuring blood vessel formation in collagen/fibronectin gels implanted in mice; proliferative, migratory, and chemotactic abilities were assessed in cell culture. Placental uptake of fetal ECFCs, assessed by differences in arterial and venous cord blood content, was determined by flow cytometry. RESULTS: In vivo, IUGR ECFCs formed fewer blood vessels (P < .001) and capillaries (P = .001) compared with normal pregnancy-derived ECFCs. In culture conditions, IUGR ECFCs had reduced proliferation (P = .01) and migration (P = .007) and diminished chemotactic abilities to stromal cell-derived factor 1 (P = .007) coupled with reduced hypoxia-induced matrix metalloproteinase-2 release (P = .02). Finally, in IUGR pregnancies, the number of ECFCs was lower in arterial cord blood (P = .002) and placental uptake of cells was reduced (P < .001). CONCLUSIONS: ECFCs derived from IUGR cord blood are rarefied and dysfunctional, resulting in diminished vasculogenic potential; this could be a cause of placental dysfunction in IUGR, with long-term postnatal implications for cardiovascular function in offspring.


Subject(s)
Endothelium, Vascular/pathology , Fetal Growth Retardation/pathology , Fetal Stem Cells/pathology , Neovascularization, Pathologic/pathology , Capillaries/enzymology , Capillaries/pathology , Capillaries/physiopathology , Cardiovascular Diseases/etiology , Cell Count , Cell Movement , Cell Proliferation , Cells, Cultured , Chemotaxis , Cohort Studies , Down-Regulation , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Female , Fetal Blood , Fetal Growth Retardation/blood , Fetal Growth Retardation/enzymology , Fetal Growth Retardation/physiopathology , Fetal Stem Cells/enzymology , Humans , Matrix Metalloproteinase 2/metabolism , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/physiopathology , Placenta/blood supply , Placenta/pathology , Pregnancy , Prospective Studies
12.
Anesthesiology ; 118(3): 537-49, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23314167

ABSTRACT

BACKGROUND: Previous studies have demonstrated that isoflurane can provide both neuroprotection and neurotoxicity in various tissue culture models and in rodent developing brains. The cellular and molecular mechanisms mediating these dual effects are not clear, but the exposure level and duration of isoflurane appear to be determinant factors. METHODS: Using the ReNcell CX (Millipore, Billerica, MA) human neural progenitor cell line, the authors investigated the impact of prolonged exposure to varying isoflurane concentrations on cell survival and neurogenesis. In addition, the authors assessed the impact of short isoflurane preconditioning on elevation of cytosolic Ca concentration and cytotoxic effects mediated by prolonged isoflurane exposures and the contribution of inositol-1,4,5-trisphosphate or ryanodine receptor activation to these processes. RESULTS: Short exposures to low isoflurane concentrations promote proliferation and differentiation of ReNcell CX cells, with no cell damage. However, prolonged exposures to high isoflurane concentrations induced significant ReNcell CX cell damage and inhibited cell proliferation. These prolonged exposures suppressed neuronal cell fate and promoted glial cell fate. Preconditioning of ReNcell CX cultures with short exposures to low concentrations of isoflurane ameliorated the effects of prolonged exposures to isoflurane. Pretreatment of ReNcell cultures with inositol-1,4,5-trisphosphate or ryanodine receptor antagonists mostly prevented isoflurane-mediated effects on survival, proliferation, and differentiation. Finally, isoflurane-preconditioned cultures showed significantly less isoflurane-evoked changes in calcium concentration. CONCLUSION: The commonly used general anesthetic isoflurane exerts dual effects on neuronal stem cell survival, proliferation, and differentiation, which may be attributed to differential regulation of calcium release through activation of endoplasmic reticulum localized inositol-1,4,5-trisphosphate and/or ryanodine receptors.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Fetal Stem Cells/drug effects , Isoflurane/pharmacology , Neural Stem Cells/drug effects , Calcium/metabolism , Cell Differentiation/physiology , Cell Line, Transformed , Cell Survival/physiology , Fetal Stem Cells/metabolism , Fetal Stem Cells/pathology , Humans , Isoflurane/toxicity , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Time Factors
13.
J Matern Fetal Neonatal Med ; 25(9): 1769-73, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22339619

ABSTRACT

OBJECTIVE: The aim of our study was to investigate the change of count and the status of cellular senescence in fetal endothelial progenitor cells (EPCs) obtained from the umbilical cord blood of women with fetal growth restriction (FGR). METHODS: Fetal EPCs were obtained from thirty five normal and thirty pregnant women with FGR. Each EPC was characterized and counted. EPC differentiation time and outgrowth endothelial cell (OEC) colony formation assay, senescence-associated ß-galactosidase (SA-ß-gal) activity assay, and telomerase activity assay were performed. RESULTS: Fetal EPC counts were significantly decreased in the FGR group compared with normal controls. In the FGR group, the EPC differentiation time was prolonged, OEC colonies were much less formed, the staining intensity of SA-ß-gal was relatively increased and the telomerase activity of EPCs was significantly decreased, compared with normal pregnancy (p < 0.001 for all). CONCLUSIONS: The fetal EPCs in FGR pregnancies were decreased, functionally impaired and senescently altered.


Subject(s)
Cellular Senescence/physiology , Endothelial Cells/pathology , Endothelial Cells/physiology , Fetal Growth Retardation/pathology , Fetal Stem Cells/physiology , Adult , Case-Control Studies , Cell Differentiation/physiology , Cell Proliferation , Cell Separation , Cells, Cultured , Endothelial Cells/cytology , Female , Fetal Growth Retardation/diagnostic imaging , Fetal Growth Retardation/physiopathology , Fetal Stem Cells/cytology , Fetal Stem Cells/pathology , Humans , Infant, Newborn , Pregnancy , Ultrasonography
14.
Stem Cells Dev ; 21(2): 321-30, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-21521032

ABSTRACT

Although maternal intake of folic acid (FA) prevents neural tube defects in 70% of the population, the exact mechanism of prevention has not been elucidated. We hypothesized that FA affects neural stem cell (NSC) proliferation and differentiation. This hypothesis was examined in a folate-responsive spina bifida mouse model, Splotch (Sp(-/-)), which has a homozygous loss-of-function mutation in the Pax3 gene. Neurospheres were generated with NSCs from the lower lumbar neural tube of E10.5 wild-type (WT) and Sp(-/-) embryos, in the presence and absence of FA. In the absence of FA, the number of neurospheres generated from Sp(-/-) embryos compared with WT was minimal (P<0.05). Addition of FA to Sp(-/-) cultures increased the expression of a Pax3 downstream target, fgfr4, and rescued NSC proliferative potential, as demonstrated by a significant increase in neurosphere formation (P<0.01). To ascertain if FA affected cell differentiation, FA-stimulated Sp(-/-) neurospheres were allowed to differentiate in the continued presence or absence of FA. Neurospheres from both conditions expressed multi-potent stem cell characteristics and the same differentiation potential as WT. Further, multiple neurospheres from both WT and FA-stimulated Sp(-/-) cell cultures formed extensive synaptic connections. On the whole, FA-mediated rescue of neural tube defects in Sp(-/-) embryos promotes NSC proliferation at an early embryonic stage. FA-stimulated Sp(-/-) neurospheres differentiate and form synaptic connections, comparable to WT.


Subject(s)
Fetal Stem Cells/drug effects , Folic Acid/pharmacology , Gene Expression Regulation, Developmental , Neural Stem Cells/drug effects , Neural Tube/drug effects , Spinal Dysraphism/drug therapy , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Embryo, Mammalian , Fetal Stem Cells/pathology , Fetus , Gene Expression Regulation, Developmental/drug effects , Gene Knockout Techniques , Homozygote , Mice , Mice, Knockout , Neural Stem Cells/pathology , Neural Tube/embryology , Neural Tube/pathology , Neurons/drug effects , Neurons/pathology , PAX3 Transcription Factor , Paired Box Transcription Factors/deficiency , Paired Box Transcription Factors/genetics , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Spinal Dysraphism/embryology , Spinal Dysraphism/genetics , Spinal Dysraphism/pathology , Synapses/drug effects , Synapses/physiology
15.
Stem Cell Rev Rep ; 8(2): 532-45, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21755312

ABSTRACT

Neural stem cells in the brain have been shown to be 'cells of origin' of certain brain cancers, most notably astrocytomas and medulloblastoma. In particular, in a mouse model, the targeting of genetic modifications for astrocytoma-relevant tumor suppressors to neural stem cells causes malignant astrocytoma to arise, thereby suggesting that astrocytoma is derived from neural stem cells. However, it remains to be determined whether this important finding is reproducible in humans. Herein, we generated cancerous neural stem cells by introducing a set of oncogenes to human fetal neural stem cells (hfNSCs). Serial genetic modification with v-myc for immortalization and consequent H-Ras for oncogenic stimulation with viral gene delivery proved sufficient to induce the transformation of hfNSCs. The resultant F3.Ras cells evidenced a variety of the hallmarks of brain cancer stem cells and most importantly were tumorigenic, forming brain cancers consisting of both a large number of differentiated and a very few undifferentiated populations of cells in an in vivo mouse model. On the contrary, oligodendrocytes derived from the v-myc expressing parent neural stem cells were not transformed by H-Ras, which suggests that neural stem cells may be more susceptible to cancerous transformation by a combination of oncogenes. We also determined that v-myc expressing fetal neural stem cells were defective in p53 response upon the introduction of H-Ras; this finding suggests that an insufficient p53-dependent tumor suppressive mechanism would be associated with high oncogenic susceptibility to H-Ras introduction.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Neoplastic Stem Cells/pathology , Neural Stem Cells/pathology , Oncogenes/genetics , Animals , Biomarkers/metabolism , Brain Neoplasms/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Disease Susceptibility , Fetal Stem Cells/pathology , Genes, myc , Genes, ras , Glioma/metabolism , Humans , Mice , Mice, SCID , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism
16.
Histol Histopathol ; 25(4): 485-93, 2010 04.
Article in English | MEDLINE | ID: mdl-20183801

ABSTRACT

Etoposide (VP-16), an anti-tumor agent, is a topoisomerase II inhibitor that causes DNA damage. In our previous studies, it was shown that VP-16 induces S-phase accumulation and G2/M arrest, eventually resulting in apoptosis, through p53-related pathway in the mouse fetal brain. We injected 4 mg/kg of VP-16 into pregnant mice on day 12 of gestation, and the fetuses were investigated for the cell cycle checkpoint and mechanism of apoptosis. The transition of the neural progenitor cells in the fetuses was delayed as compared to that in the control, and most of the apoptotic cells were BrdU positive. VP-16-induced S-phase accumulation was brought about by the acceleration of G1/S transition rather than by the inhibition of S-phase progression. Phosphorylation of ataxia telangiectasia-mutated kinase (ATM) at Ser1981 and gammaH2AX after VP-16 treatment showed DNA damage. p53 was phosphorylated at Ser15 and 20 and increased after activation of the ATM kinase pathway. Cdc25A degradation might induce the inhibition of S-phase progression. It is supposed that an increase in cyclin A might accelerate G1/S progression. It is also indicated that VP-16-induced G2/M arrest is caused by p21, which inactivates cyclin B-Cdc2 complex and eventually prevents mitotic entry. In p53-deficient fetal brains, G2/M and apoptosis were almost abrogated, although S-phase accumulation still occurred. It is suggested that VP-16 induced p53-independent S-phase accumulation, and p53-dependent G2/M arrest and apoptosis of the neural progenitor cells in fetal mouse brain.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Cycle Proteins/metabolism , Cell Cycle/drug effects , DNA Damage , DNA-Binding Proteins/metabolism , Etoposide/pharmacology , Fetal Stem Cells/pathology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Bromodeoxyuridine/metabolism , Cell Division/drug effects , Female , Fetal Stem Cells/drug effects , Fetus/drug effects , Fetus/metabolism , G2 Phase/drug effects , Maternal Exposure , Mice , Mice, Inbred ICR , Mice, Knockout , Neurons/drug effects , Neurons/pathology , Pregnancy , Telencephalon/drug effects , Telencephalon/pathology
17.
Toxicol Sci ; 113(1): 207-15, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19812361

ABSTRACT

It is hypothesized that the increasing incidence of childhood leukemia may be due to in utero exposure to environmental pollutants, such as benzene, but the mechanisms involved remain unknown. We hypothesize that reactive oxygen species (ROS) contribute to the deregulation of fetal hematopoiesis caused by in utero benzene exposure. To evaluate this hypothesis, pregnant C57Bl/6N mice were exposed to benzene or polyethylene glycol-conjugated catalase (PEG-catalase) (antioxidative enzyme) and benzene. Colony formation assays on fetal liver cells were performed to measure erythroid and myeloid progenitor cell growth potential. The presence of ROS in CD117(+) fetal liver cells was measured by flow cytometric analysis. Oxidative cellular damage was assessed by Western blot analysis of 4-hydroxynonenol (4-HNE) and nitrotyrosine products, as well as reduced to oxidized glutathione ratios. Alterations in the redox-sensitive signaling pathway nuclear factor-kappa-light-chain-enhancer of activated B cells (NF-kappaB) were measured by Western blot analysis of Inhibitor of NF-kB-alpha (IkappaB-alpha) protein levels in fetal liver tissue. In utero exposure to benzene caused a significant increase in ROS production and significantly altered fetal liver erythroid and myeloid colony numbers but did not increase the levels of 4-HNE or nitrotyrosine products or alter reduced to oxidized glutathione ratios. However, in utero exposure to benzene did cause a significant decrease in fetal liver IkappaB-alpha protein levels, suggesting activation of the NF-kappaB pathway. Benzene-induced ROS formation, abnormal colony growth, and decreased IkappaB-alpha levels were all abrogated by pretreatment with PEG-catalase. These results suggest that ROS play a key role in the development of in utero-initiated benzene toxicity potentially through disruption of hematopoietic cell signaling pathways.


Subject(s)
Benzene/toxicity , Cell Proliferation/drug effects , Environmental Pollutants/toxicity , Fetal Stem Cells/drug effects , Hematopoietic Stem Cells/drug effects , Liver/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Aldehydes/metabolism , Animals , Antioxidants/pharmacology , Blotting, Western , Catalase/pharmacology , Cells, Cultured , Colony-Forming Units Assay , Female , Fetal Stem Cells/metabolism , Fetal Stem Cells/pathology , Flow Cytometry , Gestational Age , Glutathione Disulfide/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , I-kappa B Proteins/metabolism , Liver/embryology , Liver/metabolism , Liver/pathology , Maternal Exposure , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Polyethylene Glycols/pharmacology , Pregnancy , Proto-Oncogene Proteins c-kit/analysis , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism
18.
Hepatology ; 50(4): 1194-203, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19637284

ABSTRACT

UNLABELLED: Tracking stem/progenitor cells through noninvasive imaging is a helpful means of assessing the targeting of transplanted cells to specific organs. We performed in vitro and in vivo studies wherein adult human hepatocytes and human fetal liver stem/progenitor cells were labeled with indium-111 ((111)In)-oxine and technetium-99m ((99m)Tc)-Ultratag or (99m)Tc-Ceretec. The labeling efficiency and viability of cells was analyzed in vitro, and organ biodistribution of cells was analyzed in vivo after transplantation in xenotolerant nonobese diabetic/severe combined immunodeficiency mice through intrasplenic or intraportal routes. We found that adult hepatocytes and fetal liver stem/progenitor cells incorporated (111)In but not (99m)Tc labels. After radiolabeling, cell viability was unchanged. Transplanted adult hepatocytes or fetal liver stem/progenitor cells were targeted to the liver more effectively by the intraportal rather than the intrasplenic route. Transplanted cells were retained in the liver after intraportal injection and in the liver and spleen after intrasplenic injection, without translocations into pulmonary or systemic circulations. Compared with fetal liver stem/progenitor cells, fewer adult hepatocytes were retained in the spleen after intrasplenic transplantation. The distribution of transplanted cells in organs was substantiated by genetic assays, including polymerase chain reaction amplification of DNA sequences from a primate-specific Charcot-Marie-Tooth element, and in situ hybridization for primate alphoid satellite sequences ubiquitous in all centromeres. CONCLUSION: (111)In labeling of human fetal liver stem/progenitor cells and adult hepatocytes was effective for noninvasive localization of transplanted cells. This should facilitate continued development of cell therapies through further animal and clinical studies.


Subject(s)
Fetal Stem Cells/pathology , Hepatocytes/pathology , Liver/pathology , Stem Cell Transplantation , Stem Cells/pathology , Animals , Cell Movement , Cell Survival , Female , Fetal Stem Cells/transplantation , Humans , Indium Radioisotopes , Mice , Mice, Inbred NOD , Mice, SCID , Pregnancy , Technetium
19.
Pak J Pharm Sci ; 22(2): 126-30, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19339220

ABSTRACT

Fenugreek has a wide range of medical applications and its medicinal use has been clear in several studies, however, few studies are available on effects on haematopoietic stem cell of bone marrow. The goal of the present study was to investigate the effect of Fenugreek on fetal macroscopic diameters and microscopic bone marrow cell histological changes in its teratogenic dosages. Fenugreek decoction was dissolved in 1.5 milliliter distilled water and injected intraperitoneumly in three dosages of 0.8 g/kg, 1.6 g/kg, and 3.2 g/kg for three groups of Wistar female rats mated by Wistar male. For another group (as control group) only 1.5 milliliter distilled water was injected. Bone marrow tissue was prepared from rat fetus and was cut using a microtome and stained with hematoxylin and eosin. Sections were evaluated for changes using light microscope. LD(50) for the measurement of teratogenic dosage of fenugreek was 4.1 and 3.5 g/kg in female and male rat, respectively. There was a positive relation between the injected drug dosage and fetal mortality rate. Among all fetal diameters, ear to ear diameter was decreased in groups received Fenugreek decoction. The severity of stem cell histological changes caused by 3.2 g/kg drug injection was lower than distilled water injection and in evaluation of other cells, differences in the severity of histological changes across three groups with different drug dosages and control group was detected. Fenugreek in teratogenic dosages can decrease the severity of bone marrow cell proliferation and increase fetal mortality rate.


Subject(s)
Cell Proliferation/drug effects , Fetal Death/chemically induced , Fetal Stem Cells/drug effects , Hematopoietic Stem Cells/drug effects , Plant Extracts/toxicity , Teratogens/toxicity , Trigonella , Animals , Dose-Response Relationship, Drug , Female , Fetal Stem Cells/pathology , Hematopoietic Stem Cells/pathology , Injections, Intraperitoneal , Lethal Dose 50 , Male , Plant Extracts/administration & dosage , Pregnancy , Rats , Rats, Wistar
20.
Mol Cell ; 33(5): 627-38, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19285945

ABSTRACT

Mutations in PINK1 cause autosomal recessive Parkinson's disease. PINK1 is a mitochondrial kinase of unknown function. We investigated calcium homeostasis and mitochondrial function in PINK1-deficient mammalian neurons. We demonstrate physiologically that PINK1 regulates calcium efflux from the mitochondria via the mitochondrial Na(+)/Ca(2+) exchanger. PINK1 deficiency causes mitochondrial accumulation of calcium, resulting in mitochondrial calcium overload. We show that calcium overload stimulates reactive oxygen species (ROS) production via NADPH oxidase. ROS production inhibits the glucose transporter, reducing substrate delivery and causing impaired respiration. We demonstrate that impaired respiration may be restored by provision of mitochondrial complex I and II substrates. Taken together, reduced mitochondrial calcium capacity and increased ROS lower the threshold of opening of the mitochondrial permeability transition pore (mPTP) such that physiological calcium stimuli become sufficient to induce mPTP opening in PINK1-deficient cells. Our findings propose a mechanism by which PINK1 dysfunction renders neurons vulnerable to cell death.


Subject(s)
Apoptosis , Calcium/metabolism , Fetal Stem Cells/enzymology , Mitochondria/enzymology , Neurons/enzymology , Parkinsonian Disorders/enzymology , Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cells, Cultured , Cytosol/metabolism , Energy Metabolism , Fetal Stem Cells/drug effects , Fetal Stem Cells/pathology , Fetal Stem Cells/radiation effects , Glucose Transport Proteins, Facilitative/metabolism , Homeostasis , Humans , Membrane Potential, Mitochondrial , Mesencephalon/embryology , Mesencephalon/enzymology , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/pathology , Mitochondria/radiation effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , NADPH Oxidases/metabolism , Neurons/drug effects , Neurons/pathology , Neurons/radiation effects , Oxidation-Reduction , Oxidative Stress , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Protein Kinases/deficiency , Protein Kinases/genetics , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Sodium-Calcium Exchanger/metabolism , Time Factors , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...