Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 411
Filter
1.
Exp Hematol ; 133: 104205, 2024 May.
Article in English | MEDLINE | ID: mdl-38490577

ABSTRACT

Protein phosphatase 6 (PP6) is a serine/threonine (Ser/Thr) protein phosphatase, and its catalytic subunit is Ppp6c. PP6 forms the PP2A subfamily with PP2A and PP4. The diverse phenotypes observed following small interfering RNA (siRNA)-based knockdown of Ppp6c in cultured mammalian cells suggest that PP6 plays roles in cell growth and DNA repair. There is also evidence that PP6 regulates nuclear factor kappa B (NF-κB) signaling and mitogen-activated protein kinases and inactivates transforming growth factor-ß-activated kinase 1 (TAK1). Loss of Ppp6c causes several abnormalities, including those of T cell and regulatory T cell function, neurogenesis, oogenesis, and spermatogenesis. PP2A has been reported to play an important role in erythropoiesis. However, the roles of PP6 in other hematopoietic cells have not been investigated. We generated Ppp6cfl/fl;Tie2-Cre (Ppp6cTKO) mice, in which Ppp6c was specifically deleted in hematopoietic and vascular endothelial cells. Ppp6cTKO mice displayed embryonic lethality. Ppp6c deficiency increased the number of dead cells and decreased the percentages of erythroid and monocytic cells during fetal hematopoiesis. By contrast, the number of Lin-Sca-1+c-Kit+ cells, which give rise to all hematopoietic cells, was slightly increased, but their colony-forming cell activity was markedly decreased. Ppp6c deficiency also increased phosphorylation of extracellular signal-regulated kinase 1/2 and c-Jun amino (N)-terminal kinase in fetal liver hematopoietic cells.


Subject(s)
Hematopoiesis , Mice, Knockout , Phosphoprotein Phosphatases , Animals , Mice , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/deficiency , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hematopoietic Stem Cells/metabolism , Embryo Loss/genetics , Embryo Loss/pathology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Female
3.
Mediators Inflamm ; 2023: 8215567, 2023.
Article in English | MEDLINE | ID: mdl-37035756

ABSTRACT

This study explored the role of T cell subsets and the expression of related microRNAs in patients with recurrent early pregnancy loss (EPL). Fifty patients with EPL loss between May 2018 and May 2021 were randomly selected as the EPL group, and 50 pregnant women with normal pregnancies or normal delivery outcomes were randomly selected as the control group. The expression levels of T cell subset-related markers and T cell subset-related miRNAs, in addition to the frequencies of T cell subsets, in peripheral blood of the two groups were analyzed. In terms of T cell-related markers, the results showed that the expression levels of the transcriptional regulator TBX-21 (T-bet) and interferon regulatory factor 4 (IRF4) were significantly upregulated in peripheral blood of the patients in the EPL group (P < 0.05), whereas the expression levels of GATA binding protein 3 (GATA3) and glucocorticoid-induced tumor necrosis factor receptor (GITR) were significantly downregulated (P < 0.05). In the EPL group, the expression of mir-106b, mir-93, and mir-25 was upregulated (1.51 ± 0.129, 1.43 ± 0.132, and 1.73 ± 0.156, respectively) in regulatory T (Treg) cell-related T cell subsets, whereas the expression of miR-146a and miR-155 was downregulated (P < 0.05). The frequencies of Treg and exhausted T cells in the EPL group were significantly lower than those in the control group (P < 0.05). The cell frequencies of T helper 17 (Th17) cells and exhausted Treg cells in the EPL group were significantly higher than those in the control group (P < 0.05). In conclusion, immune cells and associated miRNA profiles can be used as prognostic biomarkers for the treatment of human reproductive disorders, such as EPL.


Subject(s)
Abortion, Habitual , Embryo Loss , MicroRNAs , T-Lymphocyte Subsets , Female , Humans , Pregnancy , Abortion, Habitual/genetics , Abortion, Habitual/immunology , Embryo Loss/genetics , Embryo Loss/immunology , Gene Expression , MicroRNAs/genetics , MicroRNAs/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
4.
Nature ; 613(7943): 365-374, 2023 01.
Article in English | MEDLINE | ID: mdl-36544019

ABSTRACT

How paternal exposure to ionizing radiation affects genetic inheritance and disease risk in the offspring has been a long-standing question in radiation biology. In humans, nearly 80% of transmitted mutations arise in the paternal germline1, but the transgenerational effects of ionizing radiation exposure has remained controversial and the mechanisms are unknown. Here we show that in sex-separated Caenorhabditis elegans strains, paternal, but not maternal, exposure to ionizing radiation leads to transgenerational embryonic lethality. The offspring of irradiated males displayed various genome instability phenotypes, including DNA fragmentation, chromosomal rearrangement and aneuploidy. Paternal DNA double strand breaks were repaired by maternally provided error-prone polymerase theta-mediated end joining. Mechanistically, we show that depletion of an orthologue of human histone H1.0, HIS-24, or the heterochromatin protein HPL-1, could significantly reverse the transgenerational embryonic lethality. Removal of HIS-24 or HPL-1 reduced histone 3 lysine 9 dimethylation and enabled error-free homologous recombination repair in the germline of the F1 generation from ionizing radiation-treated P0 males, consequently improving the viability of the F2 generation. This work establishes the mechanistic underpinnings of the heritable consequences of paternal radiation exposure on the health of offspring, which may lead to congenital disorders and cancer in humans.


Subject(s)
Caenorhabditis elegans , DNA Damage , DNA Repair , Histones , Animals , Humans , Male , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/radiation effects , DNA Damage/radiation effects , Genomic Instability/radiation effects , Histones/metabolism , Mutation , Radiation, Ionizing , Embryo Loss/genetics , Female , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair , DNA Polymerase theta
5.
Science ; 378(6625): 1201-1207, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36520901

ABSTRACT

Cell death induced by tumor necrosis factor (TNF) can be beneficial during infection by helping to mount proper immune responses. However, TNF-induced death can also drive a variety of inflammatory pathologies. Protectives brakes, or cell-death checkpoints, normally repress TNF cytotoxicity to protect the organism from its potential detrimental consequences. Thus, although TNF can kill, this only occurs when one of the checkpoints is inactivated. Here, we describe a checkpoint that prevents apoptosis through the detoxification of the cytotoxic complex IIa that forms upon TNF sensing. We found that autophagy-related 9A (ATG9A) and 200kD FAK family kinase-interacting protein (FIP200) promote the degradation of this complex through a light chain 3 (LC3)-independent lysosomal targeting pathway. This detoxification mechanism was found to counteract TNF receptor 1 (TNFR1)-mediated embryonic lethality and inflammatory skin disease in mouse models.


Subject(s)
Apoptosis , Autophagy-Related Proteins , Membrane Proteins , Tumor Necrosis Factor-alpha , Vesicular Transport Proteins , Animals , Mice , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Dermatitis/genetics , Dermatitis/metabolism , Dermatitis/pathology , Disease Models, Animal , Embryo Loss/genetics , Embryo Loss/metabolism , Embryo Loss/pathology , Lysosomes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
6.
Int J Mol Sci ; 23(15)2022 Jul 24.
Article in English | MEDLINE | ID: mdl-35897733

ABSTRACT

Litter size is an important indicator to measure the production capacity of commercial pigs. Spontaneous embryo loss is an essential factor in determining sow litter size. In early pregnancy, spontaneous embryo loss in porcine is as high as 20-30% during embryo implantation. However, the specific molecular mechanism underlying spontaneous embryo loss at the end of embryo implantation remains unknown. Therefore, we comprehensively used small RNA sequencing technology, bioinformatics analysis, and molecular experiments to determine the microRNA (miRNA) expression profile in the healthy and arresting embryo implantation site of porcine endometrium on day of gestation (DG) 28. A total of 464 miRNAs were identified in arresting endometrium (AE) and healthy endometrium (HE), and 139 differentially expressed miRNAs (DEMs) were screened. We combined the mRNA sequencing dataset from the SRA database to predict the target genes of these miRNAs. A quantitative real-time PCR assay identified the expression levels of miRNAs and mRNAs. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed on differentially expressed target genes of DEMs, mainly enriched in epithelial development and amino acids metabolism-related pathways. We performed fluorescence in situ hybridization (FISH) and the dual-luciferase report gene assay to confirm miRNA and predicted target gene binding. miR-205 may inhibit its expression by combining 3'-untranslated regions (3' UTR) of tubulointerstitial nephritis antigen-like 1 (TINAGL1). The resulting inhibition of angiogenesis in the maternal endometrium ultimately leads to the formation of arresting embryos during the implantation period. This study provides a reference for the effect of miRNA on the successful implantation of pig embryos in early gestation.


Subject(s)
Embryo Loss , MicroRNAs , 3' Untranslated Regions , Animals , Embryo Implantation/genetics , Embryo Loss/genetics , Endometrium/metabolism , Female , Humans , In Situ Hybridization, Fluorescence , MicroRNAs/genetics , MicroRNAs/metabolism , Pregnancy , Swine
7.
Int J Mol Sci ; 22(21)2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34769078

ABSTRACT

Selenophosphate synthetase 1 (SEPHS1) plays an essential role in cell growth and survival. However, the underlying molecular mechanisms remain unclear. In the present study, the pathways regulated by SEPHS1 during gastrulation were determined by bioinformatical analyses and experimental verification using systemic knockout mice targeting Sephs1. We found that the coagulation system and retinoic acid signaling were most highly affected by SEPHS1 deficiency throughout gastrulation. Gene expression patterns of altered embryo morphogenesis and inhibition of Wnt signaling were predicted with high probability at E6.5. These predictions were verified by structural abnormalities in the dermal layer of Sephs1-/- embryos. At E7.5, organogenesis and activation of prolactin signaling were predicted to be affected by Sephs1 knockout. Delay of head fold formation was observed in the Sephs1-/- embryos. At E8.5, gene expression associated with organ development and insulin-like growth hormone signaling that regulates organ growth during development was altered. Consistent with these observations, various morphological abnormalities of organs and axial rotation failure were observed. We also found that the gene sets related to redox homeostasis and apoptosis were gradually enriched in a time-dependent manner until E8.5. However, DNA damage and apoptosis markers were detected only when the Sephs1-/- embryos aged to E9.5. Our results suggest that SEPHS1 deficiency causes a gradual increase of oxidative stress which changes signaling pathways during gastrulation, and afterwards leads to apoptosis.


Subject(s)
Gastrulation , Gene Expression Regulation, Developmental , Mice/embryology , Phosphotransferases/genetics , Animals , Embryo Loss/genetics , Embryo Loss/metabolism , Embryo Loss/pathology , Female , Gene Deletion , Mice/genetics , Mice/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phosphotransferases/metabolism , Pregnancy , Signal Transduction
8.
Nat Commun ; 12(1): 5005, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34408140

ABSTRACT

Embryonic aneuploidy from mis-segregation of chromosomes during meiosis causes pregnancy loss. Proper disjunction of homologous chromosomes requires the mismatch repair (MMR) genes MLH1 and MLH3, essential in mice for fertility. Variants in these genes can increase colorectal cancer risk, yet the reproductive impacts are unclear. To determine if MLH1/3 single nucleotide polymorphisms (SNPs) in human populations could cause reproductive abnormalities, we use computational predictions, yeast two-hybrid assays, and MMR and recombination assays in yeast, selecting nine MLH1 and MLH3 variants to model in mice via genome editing. We identify seven alleles causing reproductive defects in mice including female subfertility and male infertility. Remarkably, in females these alleles cause age-dependent decreases in litter size and increased embryo resorption, likely a consequence of fewer chiasmata that increase univalents at meiotic metaphase I. Our data suggest that hypomorphic alleles of meiotic recombination genes can predispose females to increased incidence of pregnancy loss from gamete aneuploidy.


Subject(s)
Abortion, Spontaneous/genetics , Aneuploidy , Embryo Loss/genetics , MutL Protein Homolog 1/genetics , MutL Proteins/genetics , Abortion, Spontaneous/metabolism , Abortion, Spontaneous/physiopathology , Alleles , Animals , Crossing Over, Genetic , DNA Mismatch Repair , Embryo Loss/physiopathology , Female , Homologous Recombination , Humans , Litter Size , Male , Meiosis , Mice , MutL Protein Homolog 1/metabolism , MutL Proteins/metabolism , Pregnancy , Reproduction , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
9.
Cells Dev ; 165: 203663, 2021 03.
Article in English | MEDLINE | ID: mdl-33993984

ABSTRACT

Asb2, ankyrin repeat, and SOCS box protein 2 form an E3 ubiquitin ligase complex. Asb2 ubiquitin ligase activity drives the degradation of filamins, which have essential functions in humans. The placenta is a temporary organ that forms during pregnancy, and normal placentation is important for survival and growth of the fetus. Recent studies have shown that approximately 25-30% of knockout (KO) mice have non-viable offspring, and 68% of knockout lines exhibit placental dysmorphologies. There are very few studies on Asb2, with insufficient research on its role in placental development. Therefore, we generated Asb2 knockout mice and undertook to investigate Asb2 expression during organogenesis, and to identify its role in early embryonic and placental development. The external morphology of KO embryos revealed abnormal phenotypes including growth retardation, pericardial effusion, pale color, and especially heart beat defect from E 9.5. Furthermore, Asb2 expression was observed in the heart from E 9.5, indicating that it is specifically expressed during early heart formation, resulting in embryonic lethality. Histological analysis of E 10.5 KO heart showed malformations such as failure of chamber formation, reduction in trabeculated myocardium length, absence of mesenchymal cells, and destruction of myocardium wall. Moreover, the histological results of Asb2-deficient placenta showed abnormal phenotypes including small labyrinth and reduced vascular complexity, indicating that failure to establish mature circulatory pattern affects the embryonic development and results in early mortality. Collectively, our results demonstrate that Asb2 knockout mice have placental defects, that subsequently result in failure to form a normal cardiac septum, and thereby result in embryo mortality in utero at around E 9.5.


Subject(s)
Embryo Loss/pathology , Heart Defects, Congenital/pathology , Placenta/pathology , Suppressor of Cytokine Signaling Proteins/deficiency , Alleles , Animals , Crosses, Genetic , Embryo Loss/genetics , Embryo, Mammalian/pathology , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental , Gene Targeting , Heart Defects, Congenital/genetics , Male , Mice, Knockout , Phenotype , Pregnancy , Suppressor of Cytokine Signaling Proteins/metabolism
10.
Mol Reprod Dev ; 88(5): 338-348, 2021 05.
Article in English | MEDLINE | ID: mdl-33843124

ABSTRACT

Embryo development requires orchestrated events, finely regulated at the molecular and cellular level by mechanisms which are progressively emerging from animal studies. With progress in genetic technologies-such as genome editing and single-cell RNA analysis-we can now assess embryo gene expression with increased precision and gain new insights into complex processes until recently difficult to explore. Multiple genes and regulative pathways have been identified for each developmental stage. We have learned that embryos with undisturbed and timely gene expression have higher chances of successful development. For example, selected genes are highly expressed during the first stages, being involved in cell adhesion, cell cycle, and regulation of transcription; other genes are instead crucial for lineage specification and therefore expressed at later stages. Due to ethical constraints, studies on human embryos remain scarce, mainly descriptive, and unable to provide functional evidence. This highlights the importance of animal studies as basic knowledge to test and appraise in a clinical context. In this review, we report on preimplantation development with a focus on genes whose impairment leads to developmental arrest. Preconceptional genetic screening could identify loss-of-function mutations of these genes; thereby, novel biomarkers of embryo quality could be adopted to improve diagnosis and treatment of infertility.


Subject(s)
Blastocyst , Embryo Loss/genetics , Embryonic Development/genetics , Animals , Blastocyst/physiology , Cell Lineage , Embryo Implantation/genetics , Female , Gene Expression Regulation, Developmental , Humans , Infertility/genetics , Male , Mice , Mice, Knockout , Morula/physiology , Mutation , Pregnancy , Wnt Signaling Pathway
11.
Sci Rep ; 11(1): 8297, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859300

ABSTRACT

E26 avian leukemia oncogene 2, 3' domain (Ets2) has been implicated in various biological processes. An Ets2 mutant model (Ets2db1/db1), which lacks the DNA-binding domain, was previously reported to exhibit embryonic lethality caused by a trophoblast abnormality. This phenotype could be rescued by tetraploid complementation, resulting in pups with wavy hair and curly whiskers. Here, we generated new Ets2 mutant models with a frame-shift mutation in exon 8 using the CRISPR/Cas9 method. Homozygous mutants could not be obtained by natural mating as embryonic development stopped before E8.5, as previously reported. When we rescued them by tetraploid complementation, these mice did not exhibit wavy hair or curly whisker phenotypes. Our newly generated mice exhibited exon 8 skipping, which led to in-frame mutant mRNA expression in the skin and thymus but not in E7.5 Ets2em1/em1 embryos. This exon 8-skipped Ets2 mRNA was translated into protein, suggesting that this Ets2 mutant protein complemented the Ets2 function in the skin. Our data implies that novel splicing variants incidentally generated after genome editing may complicate the phenotypic analysis but may also give insight into the new mechanisms related to biological gene functions.


Subject(s)
Frameshift Mutation/genetics , Phenotype , Proto-Oncogene Protein c-ets-2/genetics , RNA Splicing/genetics , CRISPR-Cas Systems , DNA-Binding Proteins/genetics , Embryo Loss/genetics , Embryo Loss/pathology , Embryonic Development/genetics , Exons/genetics , Female , Gene Editing/methods , Humans , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skin/metabolism , Trophoblasts/pathology
12.
Mol Hum Reprod ; 27(5)2021 05 08.
Article in English | MEDLINE | ID: mdl-33830236

ABSTRACT

The human endometrium is a dynamic tissue that only is receptive to host the embryo during a brief time in the middle secretory phase, called the window of implantation (WOI). Despite its importance, regulation of the menstrual cycle remains incompletely understood. The aim of this study was to characterize the gene cooperation and regulation of menstrual cycle progression, to dissect the molecular complexity underlying acquisition of endometrial receptivity for a successful pregnancy, and to provide the scientific community with detailed gene co-expression information throughout the menstrual cycle on a user-friendly web-tool database. A retrospective gene co-expression analysis was performed based on the endometrial receptivity array (ERarray) gene signature from 523 human endometrial samples collected across the menstrual cycle, including during the WOI. Gene co-expression analysis revealed the WOI as having the significantly smallest proportion of negative correlations for transcriptional profiles associated with successful pregnancies compared to other cycle stages, pointing to a global transcriptional derepression being involved in acquisition of endometrial receptivity. Regulation was greatest during the transition between proliferative and secretory endometrial phases. Further, we prioritized nuclear hormone receptors as major regulators of this derepression and proved that some genes and transcription factors involved in this process were dysregulated in patients with recurrent implantation failure. We also compiled the wealth of gene co-expression data to stimulate hypothesis-driven single-molecule endometrial studies in a user-friendly database: Menstrual Cycle Gene Co-expression Network (www.menstrualcyclegcn.com). This study revealed a global transcriptional repression across the menstrual cycle, which relaxes when the WOI opens for transcriptional profiles associated with successful pregnancies. These findings suggest that a global transcriptional derepression is needed for embryo implantation and early development.


Subject(s)
Embryo Implantation/genetics , Gene Expression Regulation, Developmental , Menstrual Cycle/genetics , Cohort Studies , Embryo Loss/genetics , Endometrium/physiology , Female , Humans , Pregnancy , Transcription, Genetic , Transcriptome
13.
FEBS Lett ; 595(10): 1462-1472, 2021 05.
Article in English | MEDLINE | ID: mdl-33686659

ABSTRACT

Centrosomal protein FOR20 has been reported to be crucial for essential cellular processes, including ciliogenesis, cell migration, and cell cycle in vertebrates. However, the function of FOR20 during mammalian embryonic development remains unknown. To investigate the in vivo function of the For20 gene in mammals, we generated For20 homozygous knockout mice by gene targeting. Our data reveal that homozygous knockout of For20 results in significant embryonic growth arrest and lethality during gestation, while the heterozygotes show no obvious defects. The absence of For20 leads to impaired left-right patterning of embryos and reduced cilia in the embryonic node. Deletion of For20 also disrupts angiogenesis in yolk sacs and embryos. These results highlight a critical role of For20 in early mammalian embryogenesis.


Subject(s)
Body Patterning/genetics , Embryo Loss/genetics , Embryo, Mammalian/abnormalities , Embryo, Mammalian/pathology , Gene Deletion , Animals , Cilia/pathology , Embryo, Mammalian/blood supply , Embryonic Development , Female , Heterozygote , Homozygote , Male , Mice , Mice, Knockout , Neovascularization, Pathologic , RNA, Messenger/genetics
14.
Reprod Biol Endocrinol ; 19(1): 18, 2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33536035

ABSTRACT

OBJECTIVE: The aim of this study is to investigate the effect of irisin on leukemia inhibitory factor (LIF) and integrin αvß3 in implantation failure uterus. METHODS: Early pregnant rats were randomly divided into normal group (N), mifepristone treated group (M), irisin group (I) and progestin group (P). The implantation failure model was established using mifepristone. Second, we evaluated the average number of embryos and detected the expression of LIF and integrin αvß3 protein and mRNA in endometrium. RESULTS: Compared with group M, the average number of embryos was significantly higher in group N, P and I, the expression of LIF and integrin αvß3 in endometrium was significantly higher in group N, P and I. CONCLUSION: Irisin could improve the poor receptive state of endometrium by promoting LIF and integrin αvß3 secretion to improve blastocyst implantation in rats of implantation failure.


Subject(s)
Embryo Implantation/drug effects , Fibronectins/pharmacology , Integrin alphaVbeta3/genetics , Leukemia Inhibitory Factor/genetics , Animals , Embryo Implantation/genetics , Embryo Loss/chemically induced , Embryo Loss/genetics , Embryo Loss/metabolism , Embryo Loss/pathology , Endometrium/drug effects , Endometrium/metabolism , Female , Fibronectins/administration & dosage , Gene Expression Regulation/drug effects , Injections, Intramuscular , Integrin alphaVbeta3/metabolism , Leukemia Inhibitory Factor/metabolism , Mifepristone/pharmacology , Pregnancy , Progestins/administration & dosage , Progestins/pharmacology , Rats , Rats, Wistar
15.
Blood ; 137(14): 1945-1958, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33512417

ABSTRACT

Although BCL-xL is critical to the survival of mature erythrocytes, it is still unclear whether other antiapoptotic molecules mediate survival during earlier stages of erythropoiesis. Here, we demonstrate that erythroid-specific Mcl1 deletion results in embryonic lethality beyond embryonic day 13.5 as a result of severe anemia caused by a lack of mature red blood cells (RBCs). Mcl1-deleted embryos exhibit stunted growth, ischemic necrosis, and decreased RBCs in the blood. Furthermore, we demonstrate that MCL-1 is only required during early definitive erythropoiesis; during later stages, developing erythrocytes become MCL-1 independent and upregulate the expression of BCL-xL. Functionally, MCL-1 relies upon its ability to prevent apoptosis to promote erythroid development because codeletion of the proapoptotic effectors Bax and Bak can overcome the requirement for MCL-1 expression. Furthermore, ectopic expression of human BCL2 in erythroid progenitors can compensate for Mcl1 deletion, indicating redundancy between these 2 antiapoptotic family members. These data clearly demonstrate a requirement for MCL-1 in promoting survival of early erythroid progenitors.


Subject(s)
Erythropoiesis , Gene Deletion , Gene Expression Regulation, Developmental , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Anemia/genetics , Anemia/pathology , Animals , Apoptosis , Cells, Cultured , Embryo Loss/genetics , Embryo Loss/pathology , Erythrocytes/pathology , Erythroid Cells/pathology , Humans , Mice, Inbred C57BL
16.
Physiol Res ; 70(1): 3-12, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33453719

ABSTRACT

Approximately 35 % of the mouse genes are indispensable for life, thus, global knock-out (KO) of those genes may result in embryonic or early postnatal lethality due to developmental abnormalities. Several KO mouse lines are valuable human disease models, but viable homozygous mutant mice are frequently required to mirror most symptoms of a human disease. The site-specific gene editing systems, the transcription activator-like effector nucleases (TALENs), Zinc-finger nucleases (ZFNs) and the clustered regularly interspaced short palindrome repeat-associated Cas9 nuclease (CRISPR/Cas9) made the generation of KO mice more efficient than before, but the homozygous lethality is still an undesired side-effect in case of many genes. The literature search was conducted using PubMed and Web of Science databases until June 30th, 2020. The following terms were combined to find relevant studies: "lethality", "mice", "knock-out", "deficient", "embryonic", "perinatal", "rescue". Additional manual search was also performed to find the related human diseases in the Online Mendelian Inheritance in Man (OMIM) database and to check the citations of the selected studies for rescuing methods. In this review, the possible solutions for rescuing human disease-relevant homozygous KO mice lethal phenotypes were summarized.


Subject(s)
CRISPR-Cas Systems/genetics , Embryo Loss/prevention & control , Gene Editing/methods , Transcription Activator-Like Effector Nucleases/genetics , Zinc Finger Nucleases/genetics , Animals , Embryo Loss/genetics , Mice , Mice, Knockout , Phenotype
17.
Biol Reprod ; 104(4): 835-849, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33354716

ABSTRACT

Defects in the maternal reproductive system that result in early pregnancy loss are important causes of human female infertility. A wide variety of biological processes are involved in implantation and establishment of a successful pregnancy. Although chromatin remodelers have been shown to play an important role in many biological processes, our understanding of the role of chromatin remodelers in female reproduction remains limited. Here, we demonstrate that female mice mutant for chromatin remodeler Cecr2 are subfertile, with defects detected at the peri-implantation stage or early pregnancy. Using both a less severe hypomorphic mutation (Cecr2GT) and a more severe presumptive null mutation (Cecr2Del), we demonstrate a clear difference in the severity of the phenotype depending on the mutation. Although neither strain shows detectable defects in folliculogenesis, both Cecr2GT/GT and Cecr2GT/Del dams show defects in pregnancy. Cecr2GT/GT females have a normal number of implantation sites at embryonic day 5.5 (E5.5), but significant embryo loss by E10.5 accompanied by the presence of vaginal blood. Cecr2GT/Del females show a more severe phenotype, with significantly fewer detectable implantation sites than wild type at E5.5. Some Cecr2GT/Del females also show premature loss of decidual tissue after artificial decidualization. Together, these results suggest a role for Cecr2 in the establishment of a successful pregnancy.


Subject(s)
Embryo Implantation/genetics , Embryo Loss/genetics , Infertility, Female/genetics , Transcription Factors/genetics , Animals , Embryo, Mammalian , Female , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mutation , Pregnancy , Transcription Factors/physiology
18.
Ultrasound Med Biol ; 47(3): 751-758, 2021 03.
Article in English | MEDLINE | ID: mdl-33293111

ABSTRACT

In vivo micro-imaging of mice is useful in studying the genetic basis of cardiac development in mutant embryos. We examined Phox2b-/- mutant mice, which lack autonomic innervation to the heart and die in utero, and investigated whether this lack of innervation causes cardiac dysfunction during embryogenesis. A VisualSonics Vevo 2100 ultrahigh-frequency linear array ultrasound machine with 30- and 40-MHz probes was used to analyze embryo size, gross characteristics, ventricular contractility and rhythm. Phox2b-/- mutant embryos underwent cessation of heartbeat and death at a greater rate than wild-type controls. We did not observe a hydrops phenotype or congenital heart defects in Phox2b-/- mutants. Analysis of heart rhythm revealed no significant correlation with genotype. Absent these signs of a progressive pathology, we suggest that Phox2b-/- mutant embryos likely die of sudden death secondary to acute arrhythmia. These data provide insight into the role of cardiac autonomic innervation during development.


Subject(s)
Echocardiography/methods , Embryo Loss/genetics , Heart/diagnostic imaging , Heart/embryology , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , Homozygote , Mice , Mice, Inbred C57BL , Phenotype
19.
Mol Cell Probes ; 55: 101688, 2021 02.
Article in English | MEDLINE | ID: mdl-33279530

ABSTRACT

Jersey haplotype (JH) 1, a stop-gain lethal mutation in the CWC15 gene, causes embryonic losses in Jersey cattle. Two PCR based assays using Amplification Refractory Mutation System (T-ARMS-PCR) and restriction fragment length polymorphism (PCR-RFLP) were developed for screening of the JH1 in cattle. During the screening, seven among 30 Indian Jersey bulls were identified as carriers of the mutant JH1 allele, the first time in the country. These PCR assays are economical, rapid and accurate; and can be used separately or in combination for screening and cross-validation of the JH1 carriers in Jersey cattle.


Subject(s)
Cattle/embryology , Cattle/genetics , Embryo Loss/genetics , Haplotypes/genetics , Mutation/genetics , Polymerase Chain Reaction/methods , Animals , Base Sequence , Biological Assay , Polymorphism, Restriction Fragment Length/genetics
20.
Clin Epigenetics ; 12(1): 179, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33213487

ABSTRACT

BACKGROUND: Paternal obesity has been associated with reduced live birth rates. It could lead to inheritance of metabolic disturbances to the offspring through epigenetic mechanisms. However, obesity is a multifactorial disorder with genetic or environmental causes. Earlier we had demonstrated differential effects of high-fat diet-induced obesity (DIO) and genetically inherited obesity (GIO) on metabolic, hormonal profile, male fertility, and spermatogenesis using two rat models. The present study aimed to understand the effect of DIO and GIO on DNA methylation in male germline, and its subsequent effects on the resorbed (post-implantation embryo loss) and normal embryos. First, we assessed the DNA methylation enzymatic machinery in the testis by Real-Time PCR, followed global DNA methylation levels in spermatozoa and testicular cells by ELISA and flow cytometry, respectively. Further, we performed Methylation Sequencing in spermatozoa for both the groups. Sequencing data in spermatozoa from both the groups were validated using Pyrosequencing. Expression of the differentially methylated genes was assessed in the resorbed and normal embryos sired by the DIO group using Real-Time PCR for functional validation. RESULTS: We noted a significant decrease in Dnmt transcript and global DNA methylation levels in the DIO group and an increase in the GIO group. Sequencing analysis showed 16,966 and 9113 differentially methylated regions in the spermatozoa of the DIO and GIO groups, respectively. Upon pathway analysis, we observed genes enriched in pathways involved in embryo growth and development namely Wnt, Hedgehog, TGF-beta, and Notch in spermatozoa for both the groups, the methylation status of which partially correlated with the gene expression pattern in resorbed and normal embryos sired by the DIO group. CONCLUSION: Our study reports the mechanism by which diet-induced and genetically inherited obesity causes differential effects on the DNA methylation in the male germline that could be due to a difference in the white adipose tissue accumulation. These differences could either lead to embryo loss or transmit obesity-related traits to the offspring in adult life.


Subject(s)
Diet, High-Fat/adverse effects , Embryo Loss/genetics , Germ Cells/metabolism , Heredity/genetics , Obesity/genetics , Adipose Tissue, White/metabolism , Animals , Case-Control Studies , DNA Methylation , Embryo Loss/metabolism , Embryonic Development/genetics , Epigenesis, Genetic , Gene Expression , Male , Models, Animal , Obesity/metabolism , Rats , Rats, Wistar , Spermatogenesis/genetics , Spermatozoa/metabolism , Testis/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...