Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Proc Natl Acad Sci U S A ; 120(20): e2210058120, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37155858

ABSTRACT

Notch signaling is essential for the emergence of definitive hematopoietic stem cells (HSCs) in the embryo and their development in the fetal liver niche. However, how Notch signaling is activated and which fetal liver cell type provides the ligand for receptor activation in HSCs is unknown. Here we provide evidence that endothelial Jagged1 (Jag1) has a critical early role in fetal liver vascular development but is not required for hematopoietic function during fetal HSC expansion. We demonstrate that Jag1 is expressed in many hematopoietic cells in the fetal liver, including HSCs, and that its expression is lost in adult bone marrow HSCs. Deletion of hematopoietic Jag1 does not affect fetal liver development; however, Jag1-deficient fetal liver HSCs exhibit a significant transplantation defect. Bulk and single-cell transcriptomic analysis of HSCs during peak expansion in the fetal liver indicates that loss of hematopoietic Jag1 leads to the downregulation of critical hematopoietic factors such as GATA2, Mllt3, and HoxA7, but does not perturb Notch receptor expression. Ex vivo activation of Notch signaling in Jag1-deficient fetal HSCs partially rescues the functional defect in a transplant setting. These findings indicate a new fetal-specific niche that is based on juxtracrine hematopoietic Notch signaling and reveal Jag1 as a fetal-specific niche factor essential for HSC function.


Subject(s)
Fetus , Hematopoietic Stem Cells , Adult , Humans , Endothelium/metabolism , Fetus/metabolism , Hematopoietic Stem Cells/metabolism , Liver/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism
2.
Proc Natl Acad Sci U S A ; 119(35): e2121251119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994670

ABSTRACT

GCN2 (general control nonderepressible 2) is a serine/threonine-protein kinase that controls messenger RNA translation in response to amino acid availability and ribosome stalling. Here, we show that GCN2 controls erythrocyte clearance and iron recycling during stress. Our data highlight the importance of liver macrophages as the primary cell type mediating these effects. During different stress conditions, such as hemolysis, amino acid deficiency or hypoxia, GCN2 knockout (GCN2-/-) mice displayed resistance to anemia compared with wild-type (GCN2+/+) mice. GCN2-/- liver macrophages exhibited defective erythrophagocytosis and lysosome maturation. Molecular analysis of GCN2-/- cells demonstrated that the ATF4-NRF2 pathway is a critical downstream mediator of GCN2 in regulating red blood cell clearance and iron recycling.


Subject(s)
Amino Acids , Erythrocytes , Iron , Liver , Macrophages , Protein Serine-Threonine Kinases , Activating Transcription Factor 4/metabolism , Amino Acids/deficiency , Amino Acids/metabolism , Anemia/metabolism , Animals , Cytophagocytosis , Erythrocytes/metabolism , Gene Deletion , Hemolysis , Hypoxia/metabolism , Iron/metabolism , Liver/cytology , Lysosomes/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Stress, Physiological
3.
J Biol Chem ; 299(2): 102841, 2023 02.
Article in English | MEDLINE | ID: mdl-36574841

ABSTRACT

Hem1 (hematopoietic protein 1), a hematopoietic cell-specific member of the Hem family of cytoplasmic adaptor proteins, is essential for lymphopoiesis and innate immunity as well as for the transition of hematopoiesis from the fetal liver to the bone marrow. However, the role of Hem1 in bone cell differentiation and bone remodeling is unknown. Here, we show that deletion of Hem1 resulted in a markedly increase in bone mass because of defective bone resorption in mice of both sexes. Hem1-deficient osteoclast progenitors were able to differentiate into osteoclasts, but the osteoclasts exhibited impaired osteoclast fusion and decreased bone-resorption activity, potentially because of decreased mitogen-activated protein kinase and tyrosine kinase c-Abl activity. Transplantation of bone marrow hematopoietic stem and progenitor cells from wildtype into Hem1 knockout mice increased bone resorption and normalized bone mass. These findings indicate that Hem1 plays a pivotal role in the maintenance of normal bone mass.


Subject(s)
Adaptor Proteins, Signal Transducing , Bone Resorption , Osteoclasts , Animals , Female , Male , Mice , Bone Resorption/genetics , Bone Resorption/metabolism , Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Mice, Knockout , Osteoclasts/metabolism , Adaptor Proteins, Signal Transducing/metabolism
4.
Blood ; 137(6): 775-787, 2021 02 11.
Article in English | MEDLINE | ID: mdl-32881992

ABSTRACT

Hematopoietic and nervous systems are linked via innervation of bone marrow (BM) niche cells. Hematopoietic stem/progenitor cells (HSPCs) express neurotransmitter receptors, such as the γ-aminobutyric acid (GABA) type B receptor subunit 1 (GABBR1), suggesting that HSPCs could be directly regulated by neurotransmitters like GABA that directly bind to GABBR1. We performed imaging mass spectrometry and found that the endogenous GABA molecule is regionally localized and concentrated near the endosteum of the BM niche. To better understand the role of GABBR1 in regulating HSPCs, we generated a constitutive Gabbr1-knockout mouse model. Analysis revealed that HSPC numbers were significantly reduced in the BM compared with wild-type littermates. Moreover, Gabbr1-null hematopoietic stem cells had diminished capacity to reconstitute irradiated recipients in a competitive transplantation model. Gabbr1-null HSPCs were less proliferative under steady-state conditions and upon stress. Colony-forming unit assays demonstrated that almost all Gabbr1-null HSPCs were in a slow or noncycling state. In vitro differentiation of Gabbr1-null HSPCs in cocultures produced fewer overall cell numbers with significant defects in differentiation and expansion of the B-cell lineage. To determine whether a GABBR1 agonist could stimulate human umbilical cord blood (UCB) HSPCs, we performed brief ex vivo treatment prior to transplant into immunodeficient mice, with significant increases in long-term engraftment of HSPCs compared with GABBR1 antagonist or vehicle treatments. Our results indicate a direct role for GABBR1 in HSPC proliferation, and identify a potential target to improve HSPC engraftment in clinical transplantation.


Subject(s)
Hematopoietic Stem Cells/cytology , Receptors, GABA-B/physiology , Animals , B-Lymphocytes/pathology , Baclofen/analogs & derivatives , Baclofen/pharmacology , Bone Marrow/innervation , Bone Marrow/metabolism , Bone Marrow Transplantation , Cell Division , Cell Lineage , Female , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Lymphopenia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Radiation Chimera , Receptors, GABA-B/deficiency , Receptors, GABA-B/genetics , Stem Cell Niche
5.
Int J Mol Sci ; 24(13)2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37446049

ABSTRACT

Coronavirus disease 2019 (COVID-19) threatens public health all over the world. It is well-accepted that the immune cells in peripheral blood are widely involved in the pathological process of COVID-19. However, hematopoietic stem and progenitor cells (HSPCs), as the main source of peripheral immune cells, have not been well studied during COVID-19 infection. We comprehensively revealed the transcriptome changes of peripheral blood HSPCs after COVID-19 infection and vaccination by single-cell RNA-seq. Compared with healthy individuals, the proportion of HSPCs in COVID-19 patients significantly increased. The increase in the proportion of HSPCs might be partly attributed to the enhancement of the HSPCs proliferation upon COVID-19 infection. However, the stemness damage of HSPCs is reflected by the decrease of differentiation signal, which can be used as a potential specific indicator of the severity and duration of COVID-19 infection. Type I interferon (IFN-I) and translation signals in HSPCs were mostly activated and inhibited after COVID-19 infection, respectively. In addition, the response of COVID-19 vaccination to the body is mild, while the secondary vaccination strengthens the immune response of primary vaccination. In conclusion, our study provides new insights into understanding the immune mechanism of COVID-19 infection.


Subject(s)
COVID-19 , Transcriptome , Humans , COVID-19 Vaccines , Single-Cell Gene Expression Analysis , Hematopoietic Stem Cells
6.
Exp Cell Res ; 388(2): 111819, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31917964

ABSTRACT

Intestinal stem cells (ISCs) play a crucial role in maintaining intestinal homeostasis upon chemotherapy and radiotherapy. It has been documented that prostaglandin E2 (PGE2) treatment improved hematopoietic stem cell function in vitro and in vivo, while the relationship between PGE2 and intestinal stem cells remains unclear. Presently, mice were exposed to PGE1, dmPGE2 and indomethacin. Numbers and function of ISCs were assessed by analyzing Olfm4+ ISCs. Intestinal protection of dmPGE2 was investigated on a 5-fluorouracil (5FU)-induced intestinal damage mouse model. The results showed that dmPGE2 treatment, but not PGE1, increased numbers of Olfm4+ ISCs in dose- and time-dependent manners. Indomethacin treatment decreased numbers of Olfm4+ ISCs. The beneficial effects of short-term dmPGE2 treatment on intestine were supported in a 5FU-induced intestinal damage model. Our data showed that 5FU treatment significantly decreased numbers of Olfm4+ ISCs and goblet cells in intestine, which could be ameliorated by dmPGE2 treatment. dmPGE2 treatment accelerated the recovery of 5FU-induced ISC injury via increasing expression of cyclin D1 and D2 in intestine. Furthermore, dmPGE2 treatment-induced expression of cyclin D1 and D2 might be mediated by up-regulation of FOXM1 expression in intestine. These findings feature PGE2 as an effective protector against chemotherapy-induced intestinal damage.


Subject(s)
Cyclin D/metabolism , Dinoprostone/pharmacology , Fluorouracil/pharmacology , Gene Expression Regulation/drug effects , Intestinal Mucosa/drug effects , Stem Cells/drug effects , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Cell Proliferation , Cyclin D/genetics , Humans , Intestinal Mucosa/injuries , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Oxytocics/pharmacology , Stem Cells/metabolism , Stem Cells/pathology , Tumor Cells, Cultured
7.
J Cell Physiol ; 235(4): 3646-3656, 2020 04.
Article in English | MEDLINE | ID: mdl-31559639

ABSTRACT

It is well known that exposure of double-stranded RNA (dsRNA) to intestine immediately induces villus damage with severe diarrhea, which is mediated by toll-like receptor 3 signaling activation. However, the role of intestinal stem cells (ISCs) remains obscure during the pathology. In the present study, polyinosinic-polycytidylic acid (poly[I:C]), mimicking viral dsRNA, was used to establish intestinal damage model. Mice were acutely and chronically exposed to poly(I:C), and ISCs in jejunum were analyzed. The results showed that the height of villus was shorter 48 hr after acute poly(I:C) exposure compared with that of controls, while chronic poly(I:C) treatment increased both villus height and crypt depth in jejunum compared with control animals. The numbers of ISCs in jejunum were significantly increased after acute and chronic poly(I:C) exposure. Poly (I:C)-stimulated ISCs have stronger capacities to differentiate into intestine endocrine cells. Mechanistically, poly(I:C) treatment increased expression of Stat1 and Axin2 in the intestinal crypt, which was along with increased expression of Myc, Bcl2, and ISC proliferation. These findings suggest that dsRNA exposure could induce ISC proliferation to ameliorate dsRNA-induced intestinal injury.


Subject(s)
Intestinal Mucosa/growth & development , Poly I-C/pharmacology , Proto-Oncogene Proteins c-myc/genetics , Stem Cells/drug effects , Animals , Apoptosis/drug effects , Axin Protein/genetics , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Jejunum/drug effects , Jejunum/growth & development , Mice , RNA, Double-Stranded/drug effects , STAT1 Transcription Factor/genetics , Signal Transduction , Toll-Like Receptor 3/genetics
8.
Ecotoxicol Environ Saf ; 202: 110900, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32593095

ABSTRACT

Heavy metals such as lead (Pb), cadmium (Cd), and mercury (Hg) are representative neurotoxicological contaminants that can evoke cognitive dysfunctions. Low levels of these contaminants can be detected simultaneously in the human blood. In our previous study, behavioral performances were markedly impaired by exposure to these heavy metal mixtures (MM) at low levels. However, the aspects of cognitive functions involved are not well understood. Here, we further analyzed search strategies using a new algorithm named Morris water maze-unbiased strategy classification (MUST-C). Rat pups were co-exposed to low doses of Pb, Cd, and Hg during the embryonic and lactation stage. MM exposure at low doses, similar to those found in the general population, impaired search strategies even though their latency and path length were not affected in the Morris water maze task. MM-exposed rats preferred to use more directionless repetition strategies and less target orientation strategies than did vehicle-exposed animals in a dose-dependent manner. In addition, thionine staining and electron microscopy further revealed that MM exposure induced dose-dependent search strategy related place cell injures in the hippocampal CA1 and CA3 regions. These results demonstrate that the use of suboptimal search strategies underlies the early cognitive deficits in rats exposed to low doses of MM. The current study determined that search strategy analysis might be a novel sensitive assessment method for evaluating in the neurobehavioral toxicity.


Subject(s)
Cognitive Dysfunction/chemically induced , Environmental Pollutants/toxicity , Metals, Heavy/toxicity , Animals , Cadmium/toxicity , Cognition , Female , Hippocampus , Humans , Juvenile Hormones , Lactation , Male , Mercury/toxicity , Rats
9.
Haematologica ; 104(11): 2164-2177, 2019 11.
Article in English | MEDLINE | ID: mdl-30923091

ABSTRACT

Loss-of-function studies have determined that Notch signaling is essential for hematopoietic and endothelial development. By deleting a single allele of the Notch1 transcriptional activation domain we generated viable, post-natal mice exhibiting hypomorphic Notch signaling. These heterozygous mice, which lack only one copy of the transcriptional activation domain, appear normal and have no endothelial or hematopoietic phenotype, apart from an inherent, cell-autonomous defect in T-cell lineage development. Following chemotherapy, these hypomorphs exhibited severe pancytopenia, weight loss and morbidity. This phenotype was confirmed in an endothelial-specific, loss-of-function Notch1 model system. Ang1, secreted by hematopoietic progenitors after damage, activated endothelial Tie2 signaling, which in turn enhanced expression of Notch ligands and potentiated Notch1 receptor activation. In our heterozygous, hypomorphic model system, the mutant protein that lacks the Notch1 transcriptional activation domain accumulated in endothelial cells and interfered with optimal activity of the wildtype Notch1 transcriptional complex. Failure of the hypomorphic mutant to efficiently drive transcription of key gene targets such as Hes1 and Myc prolonged apoptosis and limited regeneration of the bone marrow niche. Thus, basal Notch1 signaling is sufficient for niche development, but robust Notch activity is required for regeneration of the bone marrow endothelial niche and hematopoietic recovery.


Subject(s)
Cellular Microenvironment , Endothelial Cells/physiology , Receptor, Notch1/metabolism , Receptor, TIE-2/metabolism , Regeneration , Signal Transduction , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Cellular Microenvironment/drug effects , Endothelial Cells/drug effects , Fluorouracil/pharmacology , Gamma Rays/adverse effects , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Knockout , Pancytopenia/etiology , Pancytopenia/metabolism , Pancytopenia/pathology , Signal Transduction/drug effects
10.
Blood ; 127(15): 1912-22, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-26764354

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is an aggressive pediatric mixed myelodysplastic/myeloproliferative neoplasm (MDS/MPN). JMML leukemogenesis is linked to a hyperactivated RAS pathway, with driver mutations in the KRAS, NRAS, NF1, PTPN11, or CBL genes. Previous murine models demonstrated how those genes contributed to the selective hypersensitivity of JMML cells to granulocyte macrophage-colony-stimulating factor (GM-CSF), a unifying characteristic in the disease. However, it is unclear what causes the early death in children with JMML, because transformation to acute leukemia is rare. Here, we demonstrate that loss of Pten (phosphatase and tensin homolog) protein at postnatal day 8 in mice harboring Nf1 haploinsufficiency results in an aggressive MPN with death at a murine prepubertal age of 20 to 35 days (equivalent to an early juvenile age in JMML patients). The death in the mice was due to organ infiltration with monocytes/macrophages. There were elevated activities of protein kinase B (Akt) and mitogen-activated protein kinase (MAPK) in cells at physiological concentrations of GM-CSF. These were more pronounced in mice with Nf1 haploinsufficiency than in littermates with wild-type Nf1,but this model is insufficient to cause cells to be GM-CSF hypersensitive. This new model represents a murine MPN model with features of a pediatric unclassifiable mixed MDS/MPN and mimics many clinical manifestations of JMML in terms of age of onset, aggressiveness, and organ infiltration with monocytes/macrophages. Our data suggest that the timing of the loss of PTEN protein plays a critical role in determining the disease severity in myeloid malignancies. This model may be useful for studying the pathogenesis of pediatric diseases with alterations in the Ras pathway.


Subject(s)
Myeloproliferative Disorders/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Animals , Bone Marrow Transplantation , Cell Movement , Cell Separation , Disease Models, Animal , Female , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , MAP Kinase Signaling System , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/metabolism , Myeloproliferative Disorders/metabolism , Neurofibromin 1/genetics , Proto-Oncogene Proteins c-akt/metabolism , Stem Cells/cytology , Time Factors , ras Proteins/metabolism
11.
Cell Mol Biol Lett ; 23: 34, 2018.
Article in English | MEDLINE | ID: mdl-30065760

ABSTRACT

Upregulation of histone acetylation plays a critical role in the dysregulation of transcription. It alters the structure of chromatin, which leads to the onset of cancer. Histone deacetylase inhibitors may therefore be a promising way to limit cancer progression. In this study, we examined the effects of droxinostat on the growth of HT-29 colon cancer cells. Our results show that droxinostat effectively inhibited cell growth and colony-forming ability by inducing cellular apoptosis and ROS production in HT-29 cells. Notably, the apoptotic inhibitor Z-VAD-FMK significantly decreased the levels of cellular apoptosis and the antioxidant γ-tocotrienol (GT3) significantly decreased ROS production induced by droxinostat treatment. Z-VAD-FMK and GT3 also partially reversed the negative growth effects of droxinstat on HT-29 cells. GT3 treatment decreased cellular apoptosis and increased colony-forming ability upon droxinostat administration. Z-VAD-FMK treatment also partially decreased droxinostat-induced ROS production. Our findings suggest that the effects of droxinostat on colon cancer cells are mediated by the induction of oxidative stress and apoptotic cell death.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Oxidative Stress/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Colonic Neoplasms/metabolism , HT29 Cells , Humans , Reactive Oxygen Species/metabolism
12.
PLoS Genet ; 11(12): e1005675, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26684013

ABSTRACT

Replication fork stalling and collapse is a major source of genome instability leading to neoplastic transformation or cell death. Such stressed replication forks can be conservatively repaired and restarted using homologous recombination (HR) or non-conservatively repaired using micro-homology mediated end joining (MMEJ). HR repair of stressed forks is initiated by 5' end resection near the fork junction, which permits 3' single strand invasion of a homologous template for fork restart. This 5' end resection also prevents classical non-homologous end-joining (cNHEJ), a competing pathway for DNA double-strand break (DSB) repair. Unopposed NHEJ can cause genome instability during replication stress by abnormally fusing free double strand ends that occur as unstable replication fork repair intermediates. We show here that the previously uncharacterized Exonuclease/Endonuclease/Phosphatase Domain-1 (EEPD1) protein is required for initiating repair and restart of stalled forks. EEPD1 is recruited to stalled forks, enhances 5' DNA end resection, and promotes restart of stalled forks. Interestingly, EEPD1 directs DSB repair away from cNHEJ, and also away from MMEJ, which requires limited end resection for initiation. EEPD1 is also required for proper ATR and CHK1 phosphorylation, and formation of gamma-H2AX, RAD51 and phospho-RPA32 foci. Consistent with a direct role in stalled replication fork cleavage, EEPD1 is a 5' overhang nuclease in an obligate complex with the end resection nuclease Exo1 and BLM. EEPD1 depletion causes nuclear and cytogenetic defects, which are made worse by replication stress. Depleting 53BP1, which slows cNHEJ, fully rescues the nuclear and cytogenetic abnormalities seen with EEPD1 depletion. These data demonstrate that genome stability during replication stress is maintained by EEPD1, which initiates HR and inhibits cNHEJ and MMEJ.


Subject(s)
DNA Helicases/genetics , Endodeoxyribonucleases/genetics , Genomic Instability , Homologous Recombination/genetics , Intracellular Signaling Peptides and Proteins/genetics , Recombinational DNA Repair/genetics , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA End-Joining Repair/genetics , Escherichia coli Proteins/genetics , Gene Expression Regulation , HEK293 Cells , Histones/genetics , Humans , Tumor Suppressor p53-Binding Protein 1
13.
Int J Mol Sci ; 18(7)2017 Jul 04.
Article in English | MEDLINE | ID: mdl-28677663

ABSTRACT

Long Interspersed Nuclear Element 1 (LINE-1) retrotransposons are the major repetitive elements in mammalian genomes. LINE-1s are well-accepted as driving forces of evolution and critical regulators of the expression of genetic information. Alterations in LINE-1 DNA methylation may lead to its aberrant activity and are reported in virtually all human cancers and in experimental carcinogenesis. In this study, we investigated the endogenous DNA methylation status of the 5' untranslated region (UTR) of LINE-1 elements in the bone marrow hematopoietic stem cells (HSCs), hematopoietic progenitor cells (HPCs), and mononuclear cells (MNCs) in radioresistant C57BL/6J and radiosensitive CBA/J mice and in response to ionizing radiation (IR). We demonstrated that basal levels of DNA methylation within the 5'-UTRs of LINE-1 elements did not differ significantly between the two mouse strains and were negatively correlated with the evolutionary age of LINE-1 elements. Meanwhile, the expression of LINE-1 elements was higher in CBA/J mice. At two months after irradiation to 0.1 or 1 Gy of 137Cs (dose rate 1.21 Gy/min), significant decreases in LINE-1 DNA methylation in HSCs were observed in prone to radiation-induced carcinogenesis CBA/J, but not C57BL/6J mice. At the same time, no residual DNA damage, increased ROS, or changes in the cell cycle were detected in HSCs of CBA/J mice. These results suggest that epigenetic alterations may potentially serve as driving forces of radiation-induced carcinogenesis; however, future studies are needed to demonstrate the direct link between the LINE-1 DNA hypomethylation and radiation carcinogenesis.


Subject(s)
DNA Methylation/radiation effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Long Interspersed Nucleotide Elements , Radiation, Ionizing , Animals , DNA Damage , Dose-Response Relationship, Radiation , Gene Expression Regulation/radiation effects , Hematopoiesis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Retroelements , Species Specificity
14.
Blood ; 123(20): 3105-15, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24622326

ABSTRACT

Exposure to total body irradiation (TBI) induces not only acute hematopoietic radiation syndrome but also long-term or residual bone marrow (BM) injury. This residual BM injury is mainly attributed to permanent damage to hematopoietic stem cells (HSCs), including impaired self-renewal, decreased long-term repopulating capacity, and myeloid skewing. These HSC defects were associated with significant increases in production of reactive oxygen species (ROS), expression of p16(Ink4a) (p16) and Arf mRNA, and senescence-associated ß-galacotosidase (SA-ß-gal) activity, but not with telomere shortening or increased apoptosis, suggesting that TBI induces residual BM injury via induction of HSC premature senescence. This suggestion is supported by the finding that SA-ß-gal(+) HSC-enriched LSK cells showed more pronounced defects in clonogenic activity in vitro and long-term engraftment after transplantation than SA-ß-gal(-) LSK cells isolated from irradiated mice. However, genetic deletion of p16 and/or Arf had no effect on TBI-induced residual BM suppression and HSC senescence, because HSCs from irradiated p16 and/or Arf knockout (KO) mice exhibited changes similar to those seen in HSCs from wild-type mice after exposure to TBI. These findings provide important new insights into the mechanism by which TBI causes long-term BM suppression (eg, via induction of premature senescence of HSCs in a p16-Arf-independent manner).


Subject(s)
Bone Marrow/pathology , Bone Marrow/radiation effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/radiation effects , Animals , Bone Marrow/metabolism , Cells, Cultured , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Knockout Techniques , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Telomere/metabolism , Telomere/pathology , Telomere/radiation effects , Whole-Body Irradiation
15.
Tetrahedron ; 72(27-28): 4001-4006, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27773949

ABSTRACT

A group of side chain partially saturated tocotrienol analogues, namely tocoflexols, have been previously designed in an effort to improve the pharmacokinetic properties of tocotrienols. (2R,8'S,3'E,11'E)-δ-Tocodienol (1) was predicted to be a high value tocoflexol for further pharmacological evaluation. We now report here an efficient 8-step synthetic route to compound 1 utilizing naturally-occurring δ-tocotrienol as a starting material (24% total yield). The key step in the synthesis is oxidative olefin cleavage of δ-tocotrienol to afford the chroman core of 1 with retention of chirality at the C-2 stereocenter.

16.
Blood ; 121(21): 4359-65, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23568489

ABSTRACT

Chromosomal translocations are common contributors to malignancy, yet little is known about the precise molecular mechanisms by which they are generated. Sequencing translocation junctions in acute leukemias revealed that the translocations were likely mediated by a DNA double-strand break repair pathway termed nonhomologous end-joining (NHEJ). There are major 2 types of NHEJ: (1) the classical pathway initiated by the Ku complex, and (2) the alternative pathway initiated by poly ADP-ribose polymerase 1 (PARP1). Recent reports suggest that classical NHEJ repair components repress translocations, whereas alternative NHEJ components were required for translocations. The rate-limiting step for initiation of alternative NHEJ is the displacement of the Ku complex by PARP1. Therefore, we asked whether PARP1 inhibition could prevent chromosomal translocations in 3 translocation reporter systems. We found that 2 PARP1 inhibitors or repression of PARP1 protein expression strongly repressed chromosomal translocations, implying that PARP1 is essential for this process. Finally, PARP1 inhibition also reduced both ionizing radiation-generated and VP16-generated translocations in 2 cell lines. These data define PARP1 as a critical mediator of chromosomal translocations and raise the possibility that oncogenic translocations occurring after high-dose chemotherapy or radiation could be prevented by treatment with a clinically available PARP1 inhibitor.


Subject(s)
Leukemia/genetics , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/physiology , Translocation, Genetic/genetics , Translocation, Genetic/physiology , Acute Disease , Cells, Cultured , DNA Breaks, Double-Stranded , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Indoles/pharmacology , Leukemia/drug therapy , Leukemia/prevention & control , Phthalazines/pharmacology , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , RNA, Small Interfering/genetics , Translocation, Genetic/drug effects
17.
Front Immunol ; 15: 1339977, 2024.
Article in English | MEDLINE | ID: mdl-38524139

ABSTRACT

With the continuous development of nuclear technology, the radiation exposure caused by radiation therapy is a serious health hazard. It is of great significance to further develop effective radiation countermeasures. B cells easily succumb to irradiation exposure along with immunosuppressive response. The approach to ameliorate radiation-induced B cell damage is rarely studied, implying that the underlying mechanisms of B cell damage after exposure are eager to be revealed. Recent studies suggest that Notch signaling plays an important role in B cell-mediated immune response. Notch signaling is a critical regulator for B cells to maintain immune function. Although accumulating studies reported that Notch signaling contributes to the functionality of hematopoietic stem cells and T cells, its role in B cells is scarcely appreciated. Presently, we discussed the regulation of Notch signaling on B cells under radiation exposure to provide a scientific basis to prevent radiation-induced B cell damage.


Subject(s)
Biological Phenomena , Radiation Exposure , Cell Differentiation/physiology , Hematopoietic Stem Cells/physiology , Signal Transduction/physiology
18.
Andrology ; 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39287511

ABSTRACT

BACKGROUND: Previous studies have shown that the activation of p38MAPK signaling plays a crucial role in regulating gonadal cell fate decisions in both mouse and human. Excessive activation of p38MAPK by radiation significantly causes testicular damage and negatively affects the male reproductive function. Therefore, fine-tuned regulation of p38MAPK signaling is critical in both physiological and pathological conditions. RESULT: This review summarizes the impact of p38MAPK signaling on testicular germ cells and microenvironment under normal condition. The relationship between radiation, reactive oxygen species (ROS), and p38MAPK is summarized. In conclusion, radiation exposure triggers the overactivation of p38MAPK, which is regulated by ROS, resulting in testicular damage. Various p38MAPK-targeting agents are discussed, providing guidance for developing new strategies.

19.
Andrology ; 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39132925

ABSTRACT

BACKGROUND: Physical, chemical, and biological factors in the environment constantly influence in vivo and in vitro biological processes, including diverse histone modifications involved in cancer and metabolism. However, the intricate mechanisms of acetylation regulation remain poorly elucidated. In mammalian spermatogenesis, acetylation plays a crucial role in repairing double-strand DNA breaks, regulating gene transcription, and modulating various signaling pathways. RESULTS: This review summarizes the histone acetylation sites in the mouse testis and provides a comprehensive overview of how histone acetylation is involved in different stages of spermatogenesis under the regulation by histone deacetylases. The regulatory functions of various class histone deacetylases during spermatogenesis and the crossroad between histone acetylation and other histone modifications are highlighted. It is imperative to understand the mechanisms of histone acetylation regulated by histone deacetylases in spermatogenesis, which facilitates to prevent and treat infertility-related diseases.

20.
Br J Pharmacol ; 181(3): 393-412, 2024 02.
Article in English | MEDLINE | ID: mdl-37580308

ABSTRACT

BACKGROUND AND PURPOSE: Damage to the testis following exposure to ionizing radiation has become an urgent problem to be solved. Here we have investigated if inhibition of p38 mitogen-activated protein kinase (p38MAPK) signalling could alleviate radiation-induced testicular damage. EXPERIMENTAL APPROACH: In mice exposed to whole body radiation (2-6 Gy), morphological changes of the epididymis and testis was measured by histochemical staining. immunohistochemical and immunofluorescence procedures and western blotting were used to monitor expression and cellular location of proteins. Expression of genes was assessed by qPCR and RNA-Seq was used to profile gene expression. KEY RESULTS: Exposure to ionizing radiation induced dose-dependent damage to mouse testis. The sperm quality decreased at 6 and 8 weeks after 6 Gy X-ray radiation. Radiation decreased PLZF+ cells and increased SOX9+ cells, and affected the expression of 969 genes, compared with data from non-irradiated mice. Expression of genes related to p38MAPK were enriched by GO analysis and were increased in the irradiated testis, and confirmed by qPCR. Levels of phospho-p38MAPK protein increased at 28 days after irradiation. In irradiated mice, SB203580 treatment increased spermatozoa, SOX9+ cells, the area and diameter of seminiferous tubules, sperm movement rate and density. Furthermore, SB203580 treatment increased SCP3+ cells, accelerating the process of spermatogenesis. CONCLUSION AND IMPLICATIONS: Exposure to ionizing radiation clearly changed gene expression in mouse testis, involving activation of p38MAPK signalling pathways. Inhibition of p38MAPK by SB203580 partly alleviated the testicular damage caused by radiation and accelerated the recovery of sperms through promoting spermatogenesis.


Subject(s)
Semen , Testis , Male , Mice , Animals , Testis/anatomy & histology , Testis/metabolism , Spermatogenesis/radiation effects , Spermatozoa/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL