Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Syst Biol Reprod Med ; 59(6): 312-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23947923

ABSTRACT

A sub-acute toxicity test was performed to investigate the effects of molybdenum (Mo) on ovarian function. ICR adult female mice were exposed to Mo by free access to distilled water containing the Mo at 5, 10, 20, and 40 mg/L for 14 days. Compared to the control group, M II oocyte morphology, ovary index, and ovulation improved within the 5 mg/L Mo group, but were negatively affected by Mo at 40 mg/L. Morphologically abnormal ovarian mitochondria were observed at ≥ 20 mg/L. These alterations accompanied the changes in superoxide dismutase (SOD), glutathione peroxidise (GPx), and malondialdehyde (MDA) levels in ovaries. In conclusion, Mo affects oocyte quality possibly through regulating ovarian oxidative stress in a dose-dependent manner. It appears that Mo may improve ovarian function at a suitable concentration, which might be a candidate for the treatment of female infertility.


Subject(s)
Molybdenum/pharmacology , Oocytes/drug effects , Ovary/drug effects , Oxidative Stress/drug effects , Animals , Female , Glutathione Peroxidase/metabolism , Malondialdehyde/metabolism , Mice , Mice, Inbred ICR , Microscopy, Electron, Transmission , Oocytes/ultrastructure , Ovary/cytology , Ovary/enzymology , Ovary/metabolism , Superoxide Dismutase/metabolism
2.
Syst Biol Reprod Med ; 59(5): 251-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23651332

ABSTRACT

In order to investigate the effects of molybdenum (Mo) on sperm parameters and testicular oxidative stress, the ICR strain of adult mice were exposed to different doses of molybdenum for a sub-acute toxicity test. Compared to the control, our results showed that the sperm parameters, including the epididymis index, sperm motility, sperm count, and morphology, increased by a moderate dose of Mo (25 mg/L), but were negatively affected at high doses (≥ 100 mg/L). In addition, the changes of sperm parameters were accompanied with changes of the superoxide dismutase (SOD) activities, the glutathione peroxidase (GPx) activities, and the malondialdehyde (MDA) levels in testes. In conclusion, Mo affects the sperm quality through regulating the testicular oxidative stress in a complex manner.


Subject(s)
Molybdenum/toxicity , Oxidative Stress/drug effects , Sperm Motility/drug effects , Testis/drug effects , Animals , Epididymis/drug effects , Glutathione Peroxidase/metabolism , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred ICR , Semen Analysis , Sperm Count , Superoxide Dismutase/metabolism
3.
Genes Cancer ; 1(1): 26-39, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20981276

ABSTRACT

Chromosomal instability during cell division frequently causes cell death or malignant transformation. Orderly chromosome congression at the metaphase plate, a paramount process to vertebrate mitosis and meiosis, is controlled by a number of molecular regulators, including kinesins. Kinesin-8 (Kif18A) functions to control mitotic chromosome alignment at the mid-zone by negative regulation of kinetochore oscillation. Here the authors report that disrupting Kif18a function results in complete sterility in male but not in female mice. Histological examination reveals that Kif18a(-/-) testes exhibit severe developmental impairment of seminiferous tubules. Testis atrophy in Kif18a(-/-) mice is caused by perturbation of microtubule dynamics and spindle pole integrity, leading to chromosome congression defects during mitosis and meiosis. Depletion of KIF18A via RNAi causes mitotic arrest accompanied by unaligned chromosomes and increased microtubule nucleating centers in both GC-1 and HeLa cells. Prolonged depletion of KIF18A causes apoptosis due to perturbed microtubule dynamics. Further studies reveal that KIF18A silencing results in degradation of CENP-E and BubR1, which is accompanied by premature sister chromatid separation. KIF18A physically interacts with BubR1 and CENP-E, and this interaction is modulated during mitosis. Combined, the studies indicate that KIF18A is essential for normal chromosome congression during cell division and that the absence of KIF18A function causes severe defects in microtubule dynamics, spindle integrity, and checkpoint activation, leading to germinal cell aplasia in mice.

4.
PLoS One ; 4(9): e6916, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19730728

ABSTRACT

Palladin is an actin cytoskeleton-associated protein which is crucial for cell morphogenesis and motility. Previous studies have shown that palladin is localized to the axonal growth cone in neurons and may play an important role in axonal extension. Previously, we have generated palladin knockout mice which display cranial neural tube closure defect and embryonic lethality before embryonic day 15.5 (E15.5). To further study the role of palladin in the developing nervous system, we examined the innervation of palladin-deficient mouse embryos since the 200 kd, 140 kd, 90-92 kd and 50 kd palladin isoforms were undetectable in the mutant mouse embryo brain. Contrary to the results of previous studies, we found no inhibition of the axonal extension in palladin-deficient mouse embryos. The cortical neurons derived from palladin-deficient mice also showed no significant difference in neurite outgrowth as compared with those from wild-type mice. Moreover, no difference was found in neurite outgrowth of neural stem cell derived-neurons between palladin-deficient mice and wild-type mice. In conclusion, these results suggest that palladin is dispensable for normal neurite outgrowth in mice.


Subject(s)
Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Gene Expression Regulation, Developmental , Neurites/metabolism , Phosphoproteins/metabolism , Animals , Brain/embryology , Cell Proliferation , Mice , Mice, Knockout , Models, Biological , Nervous System/embryology , Neurons/metabolism , Protein Isoforms , Stem Cells/metabolism , Time Factors
5.
Lab Invest ; 89(9): 1043-52, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19564844

ABSTRACT

Liver regeneration is a very complex and well-orchestrated process associated with signaling cascades involving cytokines, growth factors, and metabolic pathways. Adiponectin is an adipocytokine secreted by mature adipocytes, and its receptors are widely distributed in many tissues, including the liver. Adiponectin has direct actions in the liver with prominent roles to improve hepatic insulin sensitivity, increase fatty acid oxidation, and decrease inflammation. To test the hypothesis that adiponectin is required for normal progress of liver regeneration, 2/3 partial hepatectomy (PH) was performed on wild-type and adiponectin-null mice. Compared to wild-type mice, adiponectin-null mice displayed decreased liver mass regrowth, impeded hepatocyte proliferation, and increased hepatic lipid accumulation. Gene expression analysis revealed that adiponectin regulated the gene transcription related to lipid metabolism. Furthermore, the suppressed hepatocyte proliferation was accompanied with reduced signal transducer and activator of transcription protein 3 (STAT3) activity and enhanced suppressor of cytokine signaling 3 (Socs3) transcription. In conclusion, adiponectin-null mice exhibit impaired liver regeneration and increased hepatic steatosis. Increased expression of Socs3 and subsequently reduced activation of STAT3 in adiponectin-null mice may contribute to the alteration of the liver regeneration capability and hepatic lipid metabolism after PH.


Subject(s)
Adiponectin/deficiency , Liver Regeneration/physiology , STAT3 Transcription Factor/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Animals , Cell Proliferation , Disease Models, Animal , Hepatectomy , Hepatocytes/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Liver/pathology , Liver/surgery , Male , Mice , Mice, Knockout , Phosphorylation , STAT3 Transcription Factor/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription, Genetic , Transcriptional Activation
6.
Blood ; 110(3): 870-6, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17431131

ABSTRACT

Palladin was originally found up-regulated with NB4 cell differentiation induced by all-trans retinoic acid. Disruption of palladin results in neural tube closure defects, liver herniation, and embryonic lethality. Here we further report that Palld(-/-) embryos exhibit a significant defect in erythropoiesis characterized by a dramatic reduction in definitive erythrocytes derived from fetal liver but not primitive erythrocytes from yolk sac. The reduction of erythrocytes is accompanied by increased apoptosis of erythroblasts and partial blockage of erythroid differentiation. However, colony-forming assay shows no differences between wild-type (wt) and mutant fetal liver or yolk sac in the number and size of colonies tested. In addition, Palld(-/-) fetal liver cells can reconstitute hematopoiesis in lethally irradiated mice. These data strongly suggest that deficient erythropoiesis in Palld(-/-) fetal liver is mainly due to a compromised erythropoietic microenvironment. As expected, erythroblastic island in Palld(-/-) fetal liver was found disorganized. Palld(-/-) fetal liver cells fail to form erythroblastic island in vitro. Interestingly, wt macrophages can form such units with either wt or mutant erythroblasts, while mutant macrophages lose their ability to bind wt or mutant erythroblasts. These data demonstrate that palladin is crucial for definitive erythropoiesis and erythroblastic island formation and, especially, required for normal function of macrophages in fetal liver.


Subject(s)
Cytoskeletal Proteins/deficiency , Erythroblasts/metabolism , Erythropoiesis/genetics , Liver/embryology , Phosphoproteins/deficiency , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Colony-Forming Units Assay , Embryo Loss/genetics , Embryo Loss/metabolism , Embryo Loss/pathology , Erythroblasts/pathology , Fetus/metabolism , Fetus/pathology , Hematopoiesis, Extramedullary/genetics , Liver/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Neural Tube Defects/pathology , Yolk Sac/embryology , Yolk Sac/metabolism
7.
Biochem Biophys Res Commun ; 355(2): 312-7, 2007 Apr 06.
Article in English | MEDLINE | ID: mdl-17300748

ABSTRACT

Triphalangeal thumb-polysyndactyly syndrome (TPT-PS) is a type of human hand-foot malformation. In this study, we collected data from a Chinese family with TPT-PS and mapped the disease region to chromosome 7q36. By using a fine mapping study and a haplotype analysis, we narrowed the affected region to 1.7cM between markers D7S2465 and D7S2423, which contains four candidate genes: HLXB9, LMBR1, NOM1, and RNF32. By sequence analysis, we found no sequence alterations, which are specific to the patients in the transcribed regions and in the intron-exon boundaries among the four genes. After closely examining intron 5 of the LMBR1 gene, we discovered a single C to T transition in the affected TPT-PS individuals of the Chinese subject family. The position of this C to T transition is located close to other sequence alterations involved in several preaxial polydactyly (PPD) families, supporting the notion that intron 5 of LMBR1 contains a cis-acting regulator of limb-specific Sonic Hedgehog (SHH). We postulate that the disruption of this cis-regulator via a single C to T transition results in the dysregulation of SHH, which leads to the TPT-PS found in this case.


Subject(s)
Fingers/abnormalities , Introns , Membrane Proteins/genetics , Syndactyly/genetics , Thumb/abnormalities , Female , Hedgehog Proteins/genetics , Humans , Male , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL
...