Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Genome Res ; 31(11): 1983-1993, 2021 11.
Article in English | MEDLINE | ID: mdl-34675070

ABSTRACT

Long terminal repeat (LTR) retrotransposons are widely distributed across the human genome. They have accumulated through retroviral integration into germline DNA and are latent genetic modules. Active LTR promoters are observed in germline cells; however, little is known about the mechanisms underlying their active transcription in somatic tissues. Here, by integrating our previous transcriptome data set with publicly available data sets, we show that the LTR families MLT2A1 and MLT2A2 are primarily expressed in human four-cell and eight-cell embryos and are also activated in some adult somatic tissues, particularly pineal gland. Three MLT2A elements function as the promoters and first exons of the protein-coding genes ABCE1, COL5A1, and GALNT13 specifically in the pineal gland of humans but not in that of macaques, suggesting that the exaptation of these LTRs as promoters occurred during recent primate evolution. This analysis provides insight into the possible transition from germline insertion to somatic expression of LTR retrotransposons.


Subject(s)
Retroelements , Terminal Repeat Sequences , Animals , Primates/genetics , Promoter Regions, Genetic , Retroelements/genetics , Terminal Repeat Sequences/genetics
2.
Genome Med ; 13(1): 40, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33726816

ABSTRACT

BACKGROUND: We report the findings from 4437 individuals (3219 patients and 1218 relatives) who have been analyzed by whole genome sequencing (WGS) at the Genomic Medicine Center Karolinska-Rare Diseases (GMCK-RD) since mid-2015. GMCK-RD represents a long-term collaborative initiative between Karolinska University Hospital and Science for Life Laboratory to establish advanced, genomics-based diagnostics in the Stockholm healthcare setting. METHODS: Our analysis covers detection and interpretation of SNVs, INDELs, uniparental disomy, CNVs, balanced structural variants, and short tandem repeat expansions. Visualization of results for clinical interpretation is carried out in Scout-a custom-developed decision support system. Results from both singleton (84%) and trio/family (16%) analyses are reported. Variant interpretation is done by 15 expert teams at the hospital involving staff from three clinics. For patients with complex phenotypes, data is shared between the teams. RESULTS: Overall, 40% of the patients received a molecular diagnosis ranging from 19 to 54% for specific disease groups. There was heterogeneity regarding causative genes (n = 754) with some of the most common ones being COL2A1 (n = 12; skeletal dysplasia), SCN1A (n = 8; epilepsy), and TNFRSF13B (n = 4; inborn errors of immunity). Some causative variants were recurrent, including previously known founder mutations, some novel mutations, and recurrent de novo mutations. Overall, GMCK-RD has resulted in a large number of patients receiving specific molecular diagnoses. Furthermore, negative cases have been included in research studies that have resulted in the discovery of 17 published, novel disease-causing genes. To facilitate the discovery of new disease genes, GMCK-RD has joined international data sharing initiatives, including ClinVar, UDNI, Beacon, and MatchMaker Exchange. CONCLUSIONS: Clinical WGS at GMCK-RD has provided molecular diagnoses to over 1200 individuals with a broad range of rare diseases. Consolidation and spread of this clinical-academic partnership will enable large-scale national collaboration.


Subject(s)
Delivery of Health Care , Rare Diseases/diagnosis , Rare Diseases/genetics , Whole Genome Sequencing , Cohort Studies , DNA Copy Number Variations/genetics , Genetic Heterogeneity , Genomics , High-Throughput Nucleotide Sequencing , Humans , Information Dissemination , Inheritance Patterns/genetics , Microsatellite Repeats/genetics , Mutation/genetics , Sweden , Uniparental Disomy/genetics
3.
Neuroendocrinology ; 111(1-2): 27-44, 2021.
Article in English | MEDLINE | ID: mdl-31991411

ABSTRACT

BACKGROUND: Male estrogen receptor beta (ERß) knockout (BERKO) mice display anxiety and aggression linked to, among others, altered serotonergic signaling in the basolateral amygdala and dorsal raphe, impaired cortical radial glia migration, and reduced GABAergic signaling. The effects on primary motor cortex (M1 cortex) and locomotor activity as a consequence of ERß loss have not been investigated. OBJECTIVE: The aim of this study was to determine whether locomotor activity is altered as a consequence of the changes in the M1 cortex. METHODS: The locomotor activity of male wild-type (WT) and BERKO mice was evaluated using the open-field and rotarod tests. Molecular changes in the M1 cortex were analyzed by RNA sequencing, electron microscopy, electrophysiology, and immunohistological techniques. In addition, we established oligodendrocyte (OL) cultures from WT and BERKO mouse embryonic stem cells to evaluate OL function. RESULTS: Locomotor profiling revealed that BERKO mice were more active than WT mice but had impaired motor coordination. Analysis of the M1 cortex pointed out differences in synapse function and myelination. There was a reduction in GABAergic signaling resulting in imbalanced excitatory and inhibitory neurotransmission as well as a defective OL differentiation accompanied by myelin defects. The effects of ERß loss on OL differentiation were confirmed in vitro. CONCLUSION: ERß is an important regulator of GABAergic interneurons and OL differentiation, which impacts on adult M1 cortex function and may be linked to increased locomotor activity and decreased motor coordination in BERKO mice.


Subject(s)
Estrogen Receptor beta/genetics , Locomotion/genetics , Motor Cortex/physiopathology , Myelin Sheath/physiology , Psychomotor Performance , Synaptic Transmission , Animals , Gene Expression , Male , Mice , Mice, Knockout , Motor Cortex/metabolism , Oligodendroglia/pathology
4.
Proc Natl Acad Sci U S A ; 114(19): E3816-E3822, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28439009

ABSTRACT

As estrogen receptor ß-/- (ERß-/-) mice age, the ventral prostate (VP) develops increased numbers of hyperplastic, fibroplastic lesions and inflammatory cells. To identify genes involved in these changes, we used RNA sequencing and immunohistochemistry to compare gene expression profiles in the VP of young (2-mo-old) and aging (18-mo-old) ERß-/- mice and their WT littermates. We also treated young and old WT mice with an ERß-selective agonist and evaluated protein expression. The most significant findings were that ERß down-regulates androgen receptor (AR) signaling and up-regulates the tumor suppressor phosphatase and tensin homolog (PTEN). ERß agonist increased expression of the AR corepressor dachshund family (DACH1/2), T-cadherin, stromal caveolin-1, and nuclear PTEN and decreased expression of RAR-related orphan receptor c, Bcl2, inducible nitric oxide synthase, and IL-6. In the ERß-/- mouse VP, RNA sequencing revealed that the following genes were up-regulated more than fivefold: Bcl2, clusterin, the cytokines CXCL16 and -17, and a marker of basal/intermediate cells (prostate stem cell antigen) and cytokeratins 4, 5, and 17. The most down-regulated genes were the following: the antioxidant gene glutathione peroxidase 3; protease inhibitors WAP four-disulfide core domain 3 (WFDC3); the tumor-suppressive genes T-cadherin and caveolin-1; the regulator of transforming growth factor ß signaling SMAD7; and the PTEN ubiquitin ligase NEDD4. The role of ERß in opposing AR signaling, proliferation, and inflammation suggests that ERß-selective agonists may be used to prevent progression of prostate cancer, prevent fibrosis and development of benign prostatic hyperplasia, and treat prostatitis.


Subject(s)
Aging/metabolism , Down-Regulation , Estrogen Receptor beta/metabolism , Prostate/metabolism , Receptors, Androgen/biosynthesis , Signal Transduction , Aging/genetics , Aging/pathology , Androgens/metabolism , Animals , Chemokine CXCL16/biosynthesis , Chemokine CXCL16/genetics , Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Clusterin/biosynthesis , Clusterin/genetics , Estrogen Receptor beta/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Keratins/biosynthesis , Keratins/genetics , Male , Mice , Mice, Knockout , Nedd4 Ubiquitin Protein Ligases/biosynthesis , Nedd4 Ubiquitin Protein Ligases/genetics , PTEN Phosphohydrolase/biosynthesis , PTEN Phosphohydrolase/genetics , Prostate/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Androgen/genetics , Smad7 Protein/biosynthesis , Smad7 Protein/genetics
6.
Development ; 143(19): 3459-3469, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27578796

ABSTRACT

Leucine twenty homeobox (LEUTX) is a paired (PRD)-like homeobox gene that is expressed almost exclusively in human embryos during preimplantation development. We previously identified a novel transcription start site for the predicted human LEUTX gene based on the transcriptional analysis of human preimplantation embryos. The novel variant encodes a protein with a complete homeodomain. Here, we provide a detailed description of the molecular cloning of the complete homeodomain-containing LEUTX Using a human embryonic stem cell overexpression model we show that the complete homeodomain isoform is functional and sufficient to activate the transcription of a large proportion of the genes that are upregulated in human embryo genome activation (EGA), whereas the previously predicted partial homeodomain isoform is largely inactive. Another PRD-like transcription factor, DPRX, is then upregulated as a powerful repressor of transcription. We propose a two-stage model of human EGA in which LEUTX acts as a transcriptional activator at the 4-cell stage, and DPRX as a balancing repressor at the 8-cell stage. We conclude that LEUTX is a candidate regulator of human EGA.


Subject(s)
Blastocyst/metabolism , Embryonic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Protein Isoforms/metabolism , Animals , Cell Line , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Humans , Mice , Polymerase Chain Reaction , Protein Isoforms/genetics
7.
Sci Rep ; 6: 28995, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27412763

ABSTRACT

PAIRED (PRD)-like homeobox genes belong to a class of predicted transcription factor genes. Several of these PRD-like homeobox genes have been predicted in silico from genomic sequence but until recently had no evidence of transcript expression. We found recently that nine PRD-like homeobox genes, ARGFX, CPHX1, CPHX2, DPRX, DUXA, DUXB, NOBOX, TPRX1 and TPRX2, were expressed in human preimplantation embryos. In the current study we characterized these PRD-like homeobox genes in depth and studied their functions as transcription factors. We cloned multiple transcript variants from human embryos and showed that the expression of these genes is specific to embryos and pluripotent stem cells. Overexpression of the genes in human embryonic stem cells confirmed their roles as transcription factors as either activators (CPHX1, CPHX2, ARGFX) or repressors (DPRX, DUXA, TPRX2) with distinct targets that could be explained by the amino acid sequence in homeodomain. Some PRD-like homeodomain transcription factors had high concordance of target genes and showed enrichment for both developmentally important gene sets and a 36 bp DNA recognition motif implicated in Embryo Genome Activation (EGA). Our data implicate a role for these previously uncharacterized PRD-like homeodomain proteins in the regulation of human embryo genome activation and preimplantation embryo development.


Subject(s)
Blastocyst/metabolism , Fetal Proteins/genetics , Gene Expression Regulation, Developmental , Genes, Homeobox , Homeodomain Proteins/genetics , Transcription Factors/genetics , Amino Acid Motifs , Amino Acid Sequence , Cloning, Molecular , Consensus Sequence , DNA, Complementary/genetics , Fetal Proteins/biosynthesis , Gene Expression Profiling , Gene Library , Homeodomain Proteins/biosynthesis , Human Embryonic Stem Cells/metabolism , Humans , Multigene Family , Organ Specificity , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors/biosynthesis
8.
Reproduction ; 151(3): 253-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26700939

ABSTRACT

Estrogen receptor beta (ERß/ESR2) has a central role in mouse ovaries, as ERß knockout (BERKO) mice are subfertile due to an increase in fibrosis around the maturing follicle and a decrease in blood supply. This has a consequence that these follicles rarely rupture to release the mature oocyte. Matrix metalloproteinases (MMPs) are modulators of the extracellular matrix, and the expression of one specific MMP, MMP-19, is normally increased in granulosa cells during their maturation until ovulation. In this study, we demonstrate that MMP-19 levels are downregulated in BERKO mouse ovaries. Using human MCF-7 cells that overexpress ERß, we could identify MMP-19 to be a transcriptional target of ligand-bound activated ERß acting on a specificity protein-1 binding site. These data provide a molecular explanation for the observed follicle rupture defect that contributes to the subfertility of female BERKO mice.


Subject(s)
Estrogen Receptor beta/metabolism , Matrix Metalloproteinases, Secreted/metabolism , Ovary/metabolism , Animals , Chorionic Gonadotropin , Down-Regulation , Estrogen Receptor alpha/metabolism , Female , Humans , MCF-7 Cells , Matrix Metalloproteinases, Secreted/genetics , Mice, Inbred C57BL , Mice, Knockout , Ovulation , Sp1 Transcription Factor/metabolism
9.
Nat Commun ; 6: 8207, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26360614

ABSTRACT

Transcriptional program that drives human preimplantation development is largely unknown. Here, by using single-cell RNA sequencing of 348 oocytes, zygotes and single blastomeres from 2- to 3-day-old embryos, we provide a detailed analysis of the human preimplantation transcriptome. By quantifying transcript far 5'-ends (TFEs), we include in our analysis transcripts that derive from alternative promoters. We show that 32 and 129 genes are transcribed during the transition from oocyte to four-cell stage and from four- to eight-cell stage, respectively. A number of identified transcripts originates from previously unannotated genes that include the PRD-like homeobox genes ARGFX, CPHX1, CPHX2, DPRX, DUXA, DUXB and LEUTX. Employing de novo promoter motif extraction on sequences surrounding TFEs, we identify significantly enriched gene regulatory motifs that often overlap with Alu elements. Our high-resolution analysis of the human transcriptome during preimplantation development may have important implications on future studies of human pluripotent stem cells and cell reprograming.


Subject(s)
Blastocyst/metabolism , Blastomeres/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Oocytes/metabolism , RNA, Messenger/metabolism , Retroelements/genetics , Transcription Factors/genetics , Zygote/metabolism , 5' Untranslated Regions , Gene Expression Profiling , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Sequence Analysis, RNA , Transcription Factors/metabolism
10.
Stem Cells ; 33(5): 1434-46, 2015 May.
Article in English | MEDLINE | ID: mdl-25645121

ABSTRACT

Congenital heart defects (CHD) occur in approximately 50% of patients with Down syndrome (DS); the mechanisms for this occurrence however remain unknown. In order to understand how these defects evolve in early development in DS, we focused on the earliest stages of cardiogenesis to ascertain perturbations in development leading to CHD. Using a trisomy 21 (T21) sibling human embryonic stem cell (hESC) model of DS, we show that T21-hESC display many significant differences in expression of genes and cell populations associated with mesodermal, and more notably, secondary heart field (SHF) development, in particular a reduced number of ISL1(+) progenitor cells. Furthermore, we provide evidence for two candidate genes located on chromosome 21, ETS2 and ERG, whose overexpression during cardiac commitment likely account for the disruption of SHF development, as revealed by downregulation or overexpression experiments. Additionally, we uncover an abnormal electrophysiological phenotype in functional T21 cardiomyocytes, a result further supported by mRNA expression data acquired using RNA-Seq. These data, in combination, revealed a cardiomyocyte-specific phenotype in T21 cardiomyocytes, likely due to the overexpression of genes such as RYR2, NCX, and L-type Ca(2+) channel. These results contribute to the understanding of the mechanisms involved in the development of CHD. Stem Cells 2015;33:1434-1446.


Subject(s)
Down Syndrome/pathology , Down Syndrome/physiopathology , Heart/embryology , Heart/physiopathology , Human Embryonic Stem Cells/metabolism , Myocytes, Cardiac/pathology , Action Potentials , Cell Differentiation , Cell Line , Chromosomes, Human, Pair 21/genetics , Down Syndrome/genetics , Gene Expression Regulation, Developmental , Genetic Association Studies , Heart Defects, Congenital/genetics , Humans , Models, Biological , Myocytes, Cardiac/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, RNA , Transcriptome/genetics
11.
PLoS One ; 9(8): e102949, 2014.
Article in English | MEDLINE | ID: mdl-25089626

ABSTRACT

Infertility is a worldwide concern that can be treated with in vitro fertilization (IVF). Improvements in IVF and infertility treatment depend largely on better understanding of the molecular mechanisms for human preimplantation development. Several large-scale studies have been conducted to identify gene expression patterns for the first five days of human development, and many functional studies utilize mouse as a model system. We have identified genes of possible importance for this time period by analyzing human microarray data and available data from online databases. We selected 70 candidate genes for human preimplantation development and investigated their expression in the early mouse development from oocyte to the 8-cell stage. Maternally loaded genes expectedly decreased in expression during development both in human and mouse. We discovered that 25 significantly upregulated genes after fertilization in human included 13 genes whose orthologs in mouse behaved differently and mimicked the expression profile of maternally expressed genes. Our findings highlight many significant differences in gene expression patterns during mouse and human preimplantation development. We also describe four cancer-testis antigen families that are also highly expressed in human embryos: PRAME, SSX, GAGE and MAGEA.


Subject(s)
Embryo, Mammalian/metabolism , Embryonic Development/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Blastocyst/metabolism , Cluster Analysis , Female , Humans , Male , Mice , Multigene Family
12.
Reproduction ; 147(6): 875-83, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24625359

ABSTRACT

The coxsackievirus and adenovirus receptor (CXADR (CAR)) is a cell adhesion molecule expressed mainly in epithelial cells. Numerous evidence indicate that CXADR has an important role in testis development and function of the blood-testis barrier (BTB) in vitro. The role of CXADR in testis physiology in vivo has, however, not been addressed. We therefore constructed a conditional CXADR knockout (cKO) mouse model in which CXADR can be depleted at any chosen timepoint by the administration of tamoxifen. We report for the first time that testicular depletion of CXADR in adult and pubertal mice does not alter BTB permeability or germ cell migration across the BTB during spermatogenesis. Adult cKO mice display normal junctional ultra-structure and localization of the junctional proteins claudin-3, occludin, junction-associated molecule-A (JAM-A), and ZO1. The BTB was intact with no leakage of biotin and lanthanum tracers into the tubular lumen. Adult CXADR cKO mice were fertile with normal sperm parameters and litter size. Breeding experiments and genotyping of the pups demonstrated that CXADR-negative sperm could fertilize WT eggs. In addition, knocking down CXADR from postnatal day 9 (P9) does not affect testicular development and BTB formation. These cKO mice were analyzed at P49 and P90 and display an intact barrier and uncompromised fertility. We conclude that CXADR possesses no direct role in testicular physiology in vivo.


Subject(s)
Blood-Testis Barrier/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/deficiency , Spermatogenesis , Spermatozoa/metabolism , Age Factors , Animals , Blood-Testis Barrier/ultrastructure , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Female , Fertility , Intercellular Junctions/metabolism , Litter Size , Male , Mice, Inbred C57BL , Mice, Knockout , Permeability , Pregnancy , Sexual Maturation , Tight Junction Proteins/metabolism
13.
Bioinformatics ; 29(22): 2943-5, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23995393

ABSTRACT

MOTIVATION: Recent transcriptome studies have revealed that total transcript numbers vary by cell type and condition; therefore, the statistical assumptions for single-cell transcriptome studies must be revisited. SAMstrt is an extension code for SAMseq, which is a statistical method for differential expression, to enable spike-in normalization and statistical testing based on the estimated absolute number of transcripts per cell for single-cell RNA-seq methods. AVAILABILITY AND IMPLEMENTATION: SAMstrt is implemented on R and available in github (https://github.com/shka/R-SAMstrt). CONTACT: shintaro.katayama@ki.se


Subject(s)
Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Data Interpretation, Statistical , Humans , Single-Cell Analysis
14.
Biomed Res Int ; 2013: 489032, 2013.
Article in English | MEDLINE | ID: mdl-23956986

ABSTRACT

The aim of this study was to evaluate mitochondrial alteration and ATP content of germinal vesicle (GV) oocytes isolated from fresh and vitrified ovaries. After superovulation, the ovaries from adult mice were collected and divided into control and vitrified groups. GV oocytes were isolated mechanically from each group. Half were cultured for 24 hours and their maturation was assessed. Metaphase II oocytes were collected and submitted to in vitro fertilization and their fertilization rates and development to the blastocyst stage were evaluated. In the remaining GV oocytes, ATP levels were quantified, and mitochondrial distribution, mitochondrial membrane potential, and intracellular free calcium were detected with rhodamine 123, JC-1 and Flou-4 AM staining, using laser-scanning confocal microscopy. Maturation and fertilization rates of GV oocytes and the developmental rates of subsequent embryos were significantly lower in vitrified samples (P < 0.05). The ATP content and Ca(2+) levels differed significantly in fresh and vitrified GV oocytes (P < 0.05). Most mitochondria were seen as large and homogenous aggregates (66.6%) in fresh GV oocytes compared to vitrified oocytes (50%). No significant differences in mitochondrial membrane potential were found between the groups. The lower maturation and fertilization rates of GV oocytes from vitrified ovaries may be due to changes in their mitochondrial function and distribution.


Subject(s)
Adenosine Triphosphate/metabolism , Cryopreservation , Oocytes/physiology , Ovary/physiology , Animals , Blastocyst/cytology , Cell Survival/physiology , Female , Fertilization/physiology , Fertilization in Vitro , Membrane Potential, Mitochondrial/physiology , Mice , Mitochondria/metabolism , Mitochondria/physiology , Oocytes/metabolism , Ovary/cytology , Vitrification
15.
Int J Dev Biol ; 55(3): 313-9, 2011.
Article in English | MEDLINE | ID: mdl-21710437

ABSTRACT

During development there is a multitude of signaling events governing the assembly of the developing organism. Receptors for signaling molecules such as fibroblast growth factor receptor 2 (FGFR2) enable the embryo to communicate with the surrounding environment and activate downstream pathways. The neural cell adhesion molecule (NCAM) was first characterized as a cell adhesion molecule highly expressed in the nervous system, but recent studies have shown that it is also a signaling receptor. Using a novel single oocyte adaptation of the proximity ligation assay, we here show a close association between NCAM and FGFR2 in mouse oocytes and 2-cell embryos. Real-time PCR analyses revealed the presence of messenger RNA encoding key proteins in downstream signaling pathways in oocytes and early mouse embryos. In summary these findings show a co-localization of NCAM and FGFR2 in early vertebrate development with intracellular signaling pathways present to enable a cellular response.


Subject(s)
CD56 Antigen/metabolism , Embryo, Mammalian/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Animals , CD56 Antigen/genetics , Cell Differentiation , Cell Membrane/metabolism , Embryo, Mammalian/cytology , Female , Gene Expression Regulation, Developmental , Mice , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Oocytes , Polymerase Chain Reaction , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , Signal Transduction
16.
Evol Dev ; 12(3): 329-42, 2010.
Article in English | MEDLINE | ID: mdl-20565543

ABSTRACT

The cystatin family comprises a group of generally broadly expressed protease inhibitors. The Cres/Testatin subgroup (CTES) genes within the type 2 cystatins differs from the classical type 2 cystatins in having a strikingly reproductive tissue-specific expression, and putative functions in reproduction have therefore been discussed. We have performed evolutionary studies of the CTES genes based on gene searches in genomes from 11 species. Ancestors of the cystatin family can be traced back to plants. We have localized the evolutionary origin of the CTES genes to the split of marsupial and placental mammals. A model for the evolution of these genes illustrates that they constitute a dynamic group of genes, which has undergone several gene expansions and we find indications of a high degree of positive selection, in striking contrast to what is seen for the classical cystatin C. We show with phylogenetic relations that the CTES genes are clustered into three original groups, a testatin, a Cres, and a CstL1 group. We have further characterized the expression patterns of all human members of the subfamily. Of a total of nine identified human genes, four express putative functional transcripts with a predominant expression in the male reproductive system. Our results are compatible with a function of this gene family in reproduction.


Subject(s)
Cystatins/genetics , Evolution, Molecular , Base Sequence , DNA Primers , DNA, Complementary , Humans , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
17.
PLoS One ; 5(4): e10263, 2010 Apr 23.
Article in English | MEDLINE | ID: mdl-20428235

ABSTRACT

The first Swiss human embryonic stem cell (hESC) line, CH-ES1, has shown features of a malignant cell line. It originated from the only single blastomere that survived cryopreservation of an embryo, and it more closely resembles teratocarcinoma lines than other hESC lines with respect to its abnormal karyotype and its formation of invasive tumors when injected into SCID mice. The aim of this study was to characterize the molecular basis of the oncogenicity of CH-ES1 cells, we looked for abnormal chromosomal copy number (by array Comparative Genomic Hybridization, aCGH) and single nucleotide polymorphisms (SNPs). To see how unique these changes were, we compared these results to data collected from the 2102Ep teratocarcinoma line and four hESC lines (H1, HS293, HS401 and SIVF-02) which displayed normal G-banding result. We identified genomic gains and losses in CH-ES1, including gains in areas containing several oncogenes. These features are similar to those observed in teratocarcinomas, and this explains the high malignancy. The CH-ES1 line was trisomic for chromosomes 1, 9, 12, 17, 19, 20 and X. Also the karyotypically (based on G-banding) normal hESC lines were also found to have several genomic changes that involved genes with known roles in cancer. The largest changes were found in the H1 line at passage number 56, when large 5 Mb duplications in chromosomes 1q32.2 and 22q12.2 were detected, but the losses and gains were seen already at passage 22. These changes found in the other lines highlight the importance of assessing the acquisition of genetic changes by hESCs before their use in regenerative medicine applications. They also point to the possibility that the acquisition of genetic changes by ESCs in culture may be used to explore certain aspects of the mechanisms regulating oncogenesis.


Subject(s)
Comparative Genomic Hybridization , Embryonic Stem Cells/pathology , Oncogenes/genetics , Polymorphism, Single Nucleotide , Teratocarcinoma/pathology , Aneuploidy , Cell Line , Chromosome Aberrations , Genome, Human , Humans
18.
Fertil Steril ; 93(8): 2621-6, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20138270

ABSTRACT

OBJECTIVE: To study the role of a synthetic insulin-like growth factor-I receptor (IGF-IR) antagonist, picropodophyllin, for mouse preimplantation embryo development in vivo and in vitro. DESIGN: In vitro and in vivo study. SETTING: Hospital-based research unit. ANIMALS: FVB/N mice and mouse embryos. INTERVENTION(S): The effect of picropodophyllin in mouse embryo development in vivo and in vitro, immunohistochemistry, ELISA, polymerase chain reaction. MAIN OUTCOME MEASURE(S): Embryo development, presence of IGF-IR, messenger RNA expression, IGF-I synthesis. RESULT(S): The effect of picropodophyllin on embryo development in vitro and in vivo was not reversible. Mice treated with picropodophyllin 1 to 3 days after mating had a reduced number of blastocysts, 40.5% versus 78.8%, and a higher number of embryos with delayed development, 48.6% versus 11.5%. Insulin-like growth factor-IR protein is present in both phosphorylated and nonphosphorylated form at all stages of embryo development. The relative IGF-IR messenger RNA expression was highest in the oocyte and reduced during development to blastocyst stage. Insulin-like growth factor-I in culture media was reduced after picropodophyllin treatment. CONCLUSION(S): We conclude that IGF-I has an important role in normal mouse embryo development and that its receptor plays an essential role in the embryonic genome activation process.


Subject(s)
Embryonic Development/drug effects , Insulin-Like Growth Factor I/pharmacology , Podophyllotoxin/analogs & derivatives , Animals , Mice , Podophyllotoxin/pharmacology , RNA, Messenger/metabolism , Receptor, IGF Type 1/metabolism
19.
N Engl J Med ; 361(5): 489-95, 2009 Jul 30.
Article in English | MEDLINE | ID: mdl-19641205

ABSTRACT

The mitochondrial aspartate-glutamate carrier isoform 1 (AGC1), specific to neurons and muscle, supplies aspartate to the cytosol and, as a component of the malate-aspartate shuttle, enables mitochondrial oxidation of cytosolic NADH, thought to be important in providing energy for neurons in the central nervous system. We describe AGC1 deficiency, a novel syndrome characterized by arrested psychomotor development, hypotonia, and seizures in a child with a homozygous missense mutation in the solute carrier family 25, member 12, gene SLC25A12, which encodes the AGC1 protein. Functional analysis of the mutant AGC1 protein showed abolished activity. The child had global hypomyelination in the cerebral hemispheres, suggesting that impaired efflux of aspartate from neuronal mitochondria prevents normal myelin formation.


Subject(s)
Amino Acid Transport Systems, Acidic/deficiency , Antiporters/deficiency , Cerebrum/pathology , Epilepsy/genetics , Hereditary Central Nervous System Demyelinating Diseases/genetics , Mitochondrial Membrane Transport Proteins/genetics , Mutation, Missense , Psychomotor Disorders/genetics , Aspartic Acid/metabolism , Child, Preschool , Female , Homozygote , Humans , Magnetic Resonance Imaging , Mitochondria/metabolism , Muscle Hypotonia/genetics , Protein Isoforms , Sequence Analysis, DNA , Syndrome
20.
Hum Mol Genet ; 18(15): 2813-24, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19423553

ABSTRACT

The molecular mechanisms that control reproductive aging and menopausal age in females are poorly understood. Here, we provide genetic evidence that 3-phosphoinositide-dependent protein kinase-1 (PDK1) signaling in oocytes preserves reproductive lifespan by maintaining the survival of ovarian primordial follicles. In mice lacking the PDK1-encoding gene Pdk1 in oocytes, the majority of primordial follicles are depleted around the onset of sexual maturity, causing premature ovarian failure (POF) during early adulthood. We further showed that suppressed PDK1-Akt-p70 S6 kinase 1 (S6K1)-ribosomal protein S6 (rpS6) signaling in oocytes appears to be responsible for the loss of primordial follicles, and mice lacking the Rps6 gene in oocytes show POF similar to that in Pdk1-deficient mice. In combination with our earlier finding that phosphatase and tensin homolog deleted on chromosome 10 (PTEN) in oocytes suppresses follicular activation, we have now pinpointed the molecular network involving phosphatidylinositol 3 kinase (PI3K)/PTEN-PDK1 signaling in oocytes that controls the survival, loss and activation of primordial follicles, which together determine reproductive aging and the length of reproductive life in females. Underactivation or overactivation of this signaling pathway in oocytes is shown to cause pathological conditions in the ovary, including POF and infertility.


Subject(s)
Aging/metabolism , Oocytes/metabolism , Ovarian Follicle/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Female , Humans , Life Expectancy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/cytology , Oocytes/growth & development , Ovarian Follicle/cytology , Ovarian Follicle/growth & development , Protein Serine-Threonine Kinases/genetics , Ribosomal Protein S6/genetics , Ribosomal Protein S6/metabolism , Ribosomal Protein S6 Kinases/genetics , Ribosomal Protein S6 Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...