51
|
Human Long Noncoding RNA Regulation of Stem Cell Potency and Differentiation. Stem Cells Int 2017; 2017:6374504. [PMID: 28951743 PMCID: PMC5603141 DOI: 10.1155/2017/6374504] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/12/2017] [Accepted: 08/02/2017] [Indexed: 12/15/2022] Open
Abstract
Because of their capability of differentiation into lineage-specific cells, stem cells are an attractive therapeutic modality in regenerative medicine. To develop an effective stem cell-based therapeutic strategy with predictable results, deeper understanding of the underlying molecular mechanisms of stem cell differentiation and/or pluripotency maintenance is required. Thus, reviewing the key factors involved in the transcriptional and epigenetic regulation of stem cell differentiation and maintenance is important. Accumulating data indicate that long noncoding RNAs (lncRNAs) mediate numerous biological processes, including stem cell differentiation and maintenance. Here, we review recent findings on the human lncRNA regulation of stem cell potency and differentiation. Although the clinical implication of these lncRNAs is only beginning to be elucidated, it is anticipated that lncRNAs will become important therapeutic targets in the near future.
Collapse
|
52
|
Fucic A, Guszak V, Mantovani A. Transplacental exposure to environmental carcinogens: Association with childhood cancer risks and the role of modulating factors. Reprod Toxicol 2017. [PMID: 28624605 DOI: 10.1016/j.reprotox.2017.06.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Biological responses to carcinogens from environmental exposure during adulthood are modulated over years or decades. Conversely, during transplacental exposure, the effects on the human foetus change within weeks, intertwining with developmental mechanisms: even short periods of transplacental exposure may be imprinted in the organism for a lifetime. The pathways leading to childhood and juvenile cancers, such as leukaemias, neuroblastoma/brain tumours, hepatoblastoma, and Willm's tumour involve prenatally-induced genomic, epigenomic and/or non-genomic effects caused by xenobiotics. Pregnant women most often live in complex environmental settings that cause transplacental exposure of the foetus to xenobiotic mixtures. Mother-child biomonitoring should integrate the analysis of chemicals/radiation present in the living and workplace environment with relevant risk modulators related to life style. The interdisciplinary approach for transplacental cancer risk assessment in high-pressure areas should be based on an integrated model for mother-child exposure estimation via profiling the exposure level by water quality analysis, usage of emission grids, and land use maps.
Collapse
Affiliation(s)
- A Fucic
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | - V Guszak
- University Clinical Centre "Zagreb", Zagreb, Croatia
| | | |
Collapse
|
53
|
Rhee C, Edwards M, Dang C, Harris J, Brown M, Kim J, Tucker HO. ARID3A is required for mammalian placenta development. Dev Biol 2017; 422:83-91. [PMID: 27965054 PMCID: PMC5540318 DOI: 10.1016/j.ydbio.2016.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 11/29/2016] [Accepted: 12/01/2016] [Indexed: 11/17/2022]
Abstract
Previous studies in the mouse indicated that ARID3A plays a critical role in the first cell fate decision required for generation of trophectoderm (TE). Here, we demonstrate that ARID3A is widely expressed during mouse and human placentation and essential for early embryonic viability. ARID3A localizes to trophoblast giant cells and other trophoblast-derived cell subtypes in the junctional and labyrinth zones of the placenta. Conventional Arid3a knockout embryos suffer restricted intrauterine growth with severe defects in placental structural organization. Arid3a null placentas show aberrant expression of subtype-specific markers as well as significant alteration in cytokines, chemokines and inflammatory response-related genes, including previously established markers of human placentation disorders. BMP4-mediated induction of trophoblast stem (TS)-like cells from human induced pluripotent stem cells results in ARID3A up-regulation and cytoplasmic to nuclear translocation. Overexpression of ARID3A in BMP4-mediated TS-like cells up-regulates TE markers, whereas pluripotency markers are down-regulated. Our results reveal an essential, conserved function for ARID3A in mammalian placental development through regulation of both intrinsic and extrinsic developmental programs.
Collapse
Affiliation(s)
- Catherine Rhee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States
| | - Melissa Edwards
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States; Cell and Molecular Biology, Colorado State University, Fort Collins, CO 80523, United States
| | - Christine Dang
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States
| | - June Harris
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States
| | - Mark Brown
- Cell and Molecular Biology, Colorado State University, Fort Collins, CO 80523, United States
| | - Jonghwan Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States
| | - Haley O Tucker
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
54
|
Lu S, Du P, Shan C, Wang Y, Ma C, Dong J. Haploinsufficiency of Hand1 improves mice survival after acute myocardial infarction through preventing cardiac rupture. Biochem Biophys Res Commun 2016; 478:1726-31. [PMID: 27601324 DOI: 10.1016/j.bbrc.2016.09.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/02/2016] [Indexed: 10/21/2022]
Abstract
Previous studies have demonstrated a significantly lower level of Hand1 in ischemic cardiomyopathy than in normal heart tissue. The role of decreased Hand1 in myocardial infarction remains unclear. This study was designed to investigate the effects of haploinsufficiency of Hand1 on mouse heart after myocardial infarction. 8-10 weeks old male heterozygous Hand1-deficient (Hand1(+/-)) mice and wild-type littermates (control) were subjected to sham operation or ligation of the left anterior descending coronary artery to induce acute myocardial infarction (AMI). Hand1(+/-) mice have low incidence of left ventricular free wall rupture in the first week after operation than control mice. Then we found lower MMP9 activity and less cardiomyocytes apoptosis in Hand1(+/-) than in control mice. All of these contribute to the protection role of haploinsufficiency of Hand1 after AMI.
Collapse
Affiliation(s)
- Shuangshuang Lu
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Science, The Academy of Medical Science of Zhengzhou University, Zhengzhou, China; Model Animal Research Center of Nanjing University, Nanjing, China.
| | - Pan Du
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Science, The Academy of Medical Science of Zhengzhou University, Zhengzhou, China
| | - Congjia Shan
- Model Animal Research Center of Nanjing University, Nanjing, China
| | - Yaohe Wang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Science, The Academy of Medical Science of Zhengzhou University, Zhengzhou, China
| | - Changsheng Ma
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jianzeng Dong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
55
|
Tunster SJ, McNamara GI, Creeth HDJ, John RM. Increased dosage of the imprinted Ascl2 gene restrains two key endocrine lineages of the mouse Placenta. Dev Biol 2016; 418:55-65. [PMID: 27542691 PMCID: PMC5040514 DOI: 10.1016/j.ydbio.2016.08.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/26/2016] [Accepted: 08/15/2016] [Indexed: 11/25/2022]
Abstract
Imprinted genes are expressed primarily from one parental allele by virtue of a germ line epigenetic process. Achaete-scute complex homolog 2 (Ascl2 aka Mash2) is a maternally expressed imprinted gene that plays a key role in placental and intestinal development. Loss-of-function of Ascl2 results in an expansion of the parietal trophoblast giant cell (P-TGC) lineage, an almost complete loss of Trophoblast specific protein alpha (Tpbpa) positive cells in the ectoplacental cone and embryonic failure by E10.5. Tpbpa expression marks the progenitors of some P-TGCs, two additional trophoblast giant cell lineages (spiral artery and canal), the spongiotrophoblast and the glycogen cell lineage. Using a transgenic model, here we show that elevated expression of Ascl2 reduced the number of P-TGC cells by 40%. Elevated Ascl2 also resulted in a marked loss of the spongiotrophoblast and a substantial mislocalisation of glycogen cells into the labyrinth. In addition, Ascl2-Tg placenta contained considerably more placental glycogen than wild type. Glycogen cells are normally located within the junctional zone in close contact with spongiotrophoblast cells, before migrating through the P-TGC layer into the maternal decidua late in gestation where their stores of glycogen are released. The failure of glycogen cells to release their stores of glycogen may explain both the inappropriate accumulation of glycogen and fetal growth restriction observed late in gestation in this model. In addition, using in a genetic cross we provide evidence that Ascl2 requires the activity of a second maternally expressed imprinted gene, Pleckstrin homology-like domain, family a, member 2 (Phlda2) to limit the expansion of the spongiotrophoblast. This "belts and braces" approach demonstrates the importance of genomic imprinting in regulating the size of the placental endocrine compartment for optimal placental development and fetal growth.
Collapse
Affiliation(s)
- S J Tunster
- Cardiff School of Biosciences, Cardiff University, Cardiff, Wales CF103AX, UK
| | - G I McNamara
- Cardiff School of Biosciences, Cardiff University, Cardiff, Wales CF103AX, UK
| | - H D J Creeth
- Cardiff School of Biosciences, Cardiff University, Cardiff, Wales CF103AX, UK
| | - R M John
- Cardiff School of Biosciences, Cardiff University, Cardiff, Wales CF103AX, UK.
| |
Collapse
|
56
|
Selesniemi K, Albers RE, Brown TL. Id2 Mediates Differentiation of Labyrinthine Placental Progenitor Cell Line, SM10. Stem Cells Dev 2016; 25:959-74. [PMID: 27168216 PMCID: PMC4931356 DOI: 10.1089/scd.2016.0010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/17/2016] [Indexed: 11/12/2022] Open
Abstract
The placenta is an organ that is formed transiently during pregnancy, and appropriate placental development is necessary for fetal survival and growth. Proper differentiation of the labyrinthine layer of the placenta is especially crucial, as it establishes the fetal-maternal interface that is involved in physiological exchange processes. Although previous studies have indicated the importance of inhibitor of differentiation/inhibitor of DNA binding-2 (Id2) helix-loop-helix transcriptional regulator in mediating cell differentiation, the ability of Id2 to regulate differentiation toward the labyrinthine (transport) lineage of the placenta has yet to be determined. In the current study, we have generated labyrinthine trophoblast progenitor cells with increased (SM10-Id2) or decreased (SM10-Id2-shRNA) Id2 expression and determined the effect on TGF-β-induced differentiation. Our Id2 overexpression and knockdown analyses indicate that Id2 mediates TGF-β-induced morphological differentiation of labyrinthine trophoblast cells, as Id2 overexpression prevents differentiation and Id2 knockdown results in differentiation. Thus, our data indicate that Id2 is an important molecular mediator of labyrinthine trophoblast differentiation. An understanding of the regulators of trophoblast progenitor differentiation toward the labyrinthine lineage may offer insights into events governing pregnancy-associated disorders, such as placental insufficiency, fetal growth restriction, and preeclampsia.
Collapse
Affiliation(s)
- Kaisa Selesniemi
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University Boonshoft School of Medicine , Dayton, Ohio
| | - Renee E Albers
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University Boonshoft School of Medicine , Dayton, Ohio
| | - Thomas L Brown
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University Boonshoft School of Medicine , Dayton, Ohio
| |
Collapse
|
57
|
Induction of specific neuron types by overexpression of single transcription factors. In Vitro Cell Dev Biol Anim 2016; 52:961-973. [PMID: 27251161 DOI: 10.1007/s11626-016-0056-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/04/2016] [Indexed: 12/12/2022]
Abstract
Specific neuronal types derived from embryonic stem cells (ESCs) can facilitate mechanistic studies and potentially aid in regenerative medicine. Existing induction methods, however, mostly rely on the effects of the combined action of multiple added growth factors, which generally tend to result in mixed populations of neurons. Here, we report that overexpression of specific transcription factors (TFs) in ESCs can rather guide the differentiation of ESCs towards specific neuron lineages. Analysis of data on gene expression changes 2 d after induction of each of 185 TFs implicated candidate TFs for further ESC differentiation studies. Induction of 23 TFs (out of 49 TFs tested) for 6 d facilitated neural differentiation of ESCs as inferred from increased proportion of cells with neural progenitor marker PSA-NCAM. We identified early activation of the Notch signaling pathway as a common feature of most potent inducers of neural differentiation. The majority of neuron-like cells generated by induction of Ascl1, Smad7, Nr2f1, Dlx2, Dlx4, Nr2f2, Barhl2, and Lhx1 were GABA-positive and expressed other markers of GABAergic neurons. In the same way, we identified Lmx1a and Nr4a2 as inducers for neurons bearing dopaminergic markers and Isl1, Fezf2, and St18 for cholinergic motor neurons. A time-course experiment with induction of Ascl1 showed early upregulation of most neural-specific messenger RNA (mRNA) and microRNAs (miRNAs). Sets of Ascl1-induced mRNAs and miRNAs were enriched in Ascl1 targets. In further studies, enrichment of cells obtained with the induction of Ascl1, Smad7, and Nr2f1 using microbeads resulted in essentially pure population of neuron-like cells with expression profiles similar to neural tissues and expressed markers of GABAergic neurons. In summary, this study indicates that induction of transcription factors is a promising approach to generate cultures that show the transcription profiles characteristic of specific neural cell types.
Collapse
|
58
|
Garg V, Morgani S, Hadjantonakis AK. Capturing Identity and Fate Ex Vivo: Stem Cells from the Mouse Blastocyst. Curr Top Dev Biol 2016; 120:361-400. [PMID: 27475857 DOI: 10.1016/bs.ctdb.2016.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
During mouse preimplantation development, three molecularly, morphologically, and spatially distinct lineages are formed, the embryonic epiblast, the extraembryonic primitive endoderm, and the trophectoderm. Stem cell lines representing each of these lineages have now been derived and can be indefinitely maintained and expanded in culture, providing an unlimited source of material to study the interplay of tissue-specific transcription factors and signaling pathways involved in these fundamental cell fate decisions. Here we outline our current understanding of the derivation, maintenance, and properties of these in vitro stem cell models representing the preimplantation embryonic lineages.
Collapse
Affiliation(s)
- V Garg
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, United States; Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, United States
| | - S Morgani
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - A-K Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, United States; Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, United States.
| |
Collapse
|
59
|
Sergent F, Hoffmann P, Brouillet S, Garnier V, Salomon A, Murthi P, Benharouga M, Feige JJ, Alfaidy N. Sustained Endocrine Gland-Derived Vascular Endothelial Growth Factor Levels Beyond the First Trimester of Pregnancy Display Phenotypic and Functional Changes Associated With the Pathogenesis of Pregnancy-Induced Hypertension. Hypertension 2016; 68:148-56. [PMID: 27141059 DOI: 10.1161/hypertensionaha.116.07442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/01/2016] [Indexed: 12/14/2022]
Abstract
Pregnancy-induced hypertension diseases are classified as gestational hypertension, preeclampsia, or eclampsia. The mechanisms of their development and prediction are still to be discovered. Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) is an angiogenic factor secreted by the placenta during the first trimester of human pregnancy that was shown to control trophoblast invasion, to be upregulated by hypoxia, and to be abnormally elevated in pathological pregnancies complicated with preeclampsia and intrauterine growth restriction. These findings suggested that sustaining EG-VEGF levels beyond the first trimester of pregnancy may contribute to pregnancy-induced hypertension. To test this hypothesis, osmotic minipumps delivering EG-VEGF were implanted subcutaneously into gravid OF1 (Oncins France 1) mice on day 11.5 post coitus, which is equivalent to the end of the first trimester of human pregnancy. Mice were euthanized at 15.5 and 18.5 days post coitus to assess (1) litter size, placental, and fetal weights; (2) placental histology and function; (3) maternal blood pressure; (4) renal histology and function; and (5) circulating soluble fms-like tyrosine kinase 1 and soluble endoglin. Increased EG-VEGF levels caused significant defects in placental organization and function. Both increased hypoxia and decreased trophoblast invasion were observed. Treated mice had elevated circulating soluble fms-like tyrosine kinase 1 and soluble endoglin and developed gestational hypertension with dysregulated maternal kidney function. EG-VEGF effect on the kidney function was secondary to its effects on the placenta as similarly treated male mice had normal kidney functions. Altogether, these data provide a strong evidence to confirm that sustained EG-VEGF beyond the first trimester of pregnancy contributes to the development of pregnancy-induced hypertension.
Collapse
Affiliation(s)
- Frédéric Sergent
- From the Institut National de la Santé et de la Recherche Médicale, Grenoble, France (F.S., P.H., S.B., V.G., A.S., J.-J.F., N.A.); University Grenoble-Alpes, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Commissariat à l'Energie Atomique (CEA), BIG (Biosciences Biotechnology Institute of Grenoble)-Biology of Cancer and Infection, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Department of Obstetrics and Gynaecology, University Hospital of Grenoble, La Tronche, France (P.H.); Laboratoire d'Aide à la Procréation-CECOS, University Hospital of Grenoble, La Tronche, France (S.B.); Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5249, Laboratoire de Chimie et Biologie des Métaux, Grenoble, France (M.B.); and Department of Medicine, School of Clinical Sciences, Monash university and the Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia (P.M.)
| | - Pascale Hoffmann
- From the Institut National de la Santé et de la Recherche Médicale, Grenoble, France (F.S., P.H., S.B., V.G., A.S., J.-J.F., N.A.); University Grenoble-Alpes, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Commissariat à l'Energie Atomique (CEA), BIG (Biosciences Biotechnology Institute of Grenoble)-Biology of Cancer and Infection, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Department of Obstetrics and Gynaecology, University Hospital of Grenoble, La Tronche, France (P.H.); Laboratoire d'Aide à la Procréation-CECOS, University Hospital of Grenoble, La Tronche, France (S.B.); Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5249, Laboratoire de Chimie et Biologie des Métaux, Grenoble, France (M.B.); and Department of Medicine, School of Clinical Sciences, Monash university and the Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia (P.M.)
| | - Sophie Brouillet
- From the Institut National de la Santé et de la Recherche Médicale, Grenoble, France (F.S., P.H., S.B., V.G., A.S., J.-J.F., N.A.); University Grenoble-Alpes, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Commissariat à l'Energie Atomique (CEA), BIG (Biosciences Biotechnology Institute of Grenoble)-Biology of Cancer and Infection, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Department of Obstetrics and Gynaecology, University Hospital of Grenoble, La Tronche, France (P.H.); Laboratoire d'Aide à la Procréation-CECOS, University Hospital of Grenoble, La Tronche, France (S.B.); Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5249, Laboratoire de Chimie et Biologie des Métaux, Grenoble, France (M.B.); and Department of Medicine, School of Clinical Sciences, Monash university and the Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia (P.M.)
| | - Vanessa Garnier
- From the Institut National de la Santé et de la Recherche Médicale, Grenoble, France (F.S., P.H., S.B., V.G., A.S., J.-J.F., N.A.); University Grenoble-Alpes, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Commissariat à l'Energie Atomique (CEA), BIG (Biosciences Biotechnology Institute of Grenoble)-Biology of Cancer and Infection, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Department of Obstetrics and Gynaecology, University Hospital of Grenoble, La Tronche, France (P.H.); Laboratoire d'Aide à la Procréation-CECOS, University Hospital of Grenoble, La Tronche, France (S.B.); Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5249, Laboratoire de Chimie et Biologie des Métaux, Grenoble, France (M.B.); and Department of Medicine, School of Clinical Sciences, Monash university and the Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia (P.M.)
| | - Aude Salomon
- From the Institut National de la Santé et de la Recherche Médicale, Grenoble, France (F.S., P.H., S.B., V.G., A.S., J.-J.F., N.A.); University Grenoble-Alpes, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Commissariat à l'Energie Atomique (CEA), BIG (Biosciences Biotechnology Institute of Grenoble)-Biology of Cancer and Infection, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Department of Obstetrics and Gynaecology, University Hospital of Grenoble, La Tronche, France (P.H.); Laboratoire d'Aide à la Procréation-CECOS, University Hospital of Grenoble, La Tronche, France (S.B.); Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5249, Laboratoire de Chimie et Biologie des Métaux, Grenoble, France (M.B.); and Department of Medicine, School of Clinical Sciences, Monash university and the Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia (P.M.)
| | - Padma Murthi
- From the Institut National de la Santé et de la Recherche Médicale, Grenoble, France (F.S., P.H., S.B., V.G., A.S., J.-J.F., N.A.); University Grenoble-Alpes, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Commissariat à l'Energie Atomique (CEA), BIG (Biosciences Biotechnology Institute of Grenoble)-Biology of Cancer and Infection, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Department of Obstetrics and Gynaecology, University Hospital of Grenoble, La Tronche, France (P.H.); Laboratoire d'Aide à la Procréation-CECOS, University Hospital of Grenoble, La Tronche, France (S.B.); Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5249, Laboratoire de Chimie et Biologie des Métaux, Grenoble, France (M.B.); and Department of Medicine, School of Clinical Sciences, Monash university and the Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia (P.M.)
| | - Mohamed Benharouga
- From the Institut National de la Santé et de la Recherche Médicale, Grenoble, France (F.S., P.H., S.B., V.G., A.S., J.-J.F., N.A.); University Grenoble-Alpes, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Commissariat à l'Energie Atomique (CEA), BIG (Biosciences Biotechnology Institute of Grenoble)-Biology of Cancer and Infection, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Department of Obstetrics and Gynaecology, University Hospital of Grenoble, La Tronche, France (P.H.); Laboratoire d'Aide à la Procréation-CECOS, University Hospital of Grenoble, La Tronche, France (S.B.); Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5249, Laboratoire de Chimie et Biologie des Métaux, Grenoble, France (M.B.); and Department of Medicine, School of Clinical Sciences, Monash university and the Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia (P.M.)
| | - Jean-Jacques Feige
- From the Institut National de la Santé et de la Recherche Médicale, Grenoble, France (F.S., P.H., S.B., V.G., A.S., J.-J.F., N.A.); University Grenoble-Alpes, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Commissariat à l'Energie Atomique (CEA), BIG (Biosciences Biotechnology Institute of Grenoble)-Biology of Cancer and Infection, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Department of Obstetrics and Gynaecology, University Hospital of Grenoble, La Tronche, France (P.H.); Laboratoire d'Aide à la Procréation-CECOS, University Hospital of Grenoble, La Tronche, France (S.B.); Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5249, Laboratoire de Chimie et Biologie des Métaux, Grenoble, France (M.B.); and Department of Medicine, School of Clinical Sciences, Monash university and the Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia (P.M.)
| | - Nadia Alfaidy
- From the Institut National de la Santé et de la Recherche Médicale, Grenoble, France (F.S., P.H., S.B., V.G., A.S., J.-J.F., N.A.); University Grenoble-Alpes, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Commissariat à l'Energie Atomique (CEA), BIG (Biosciences Biotechnology Institute of Grenoble)-Biology of Cancer and Infection, Grenoble, France (F.S., P.H., S.B., V.G., A.S., M.B., J.-J.F., N.A.); Department of Obstetrics and Gynaecology, University Hospital of Grenoble, La Tronche, France (P.H.); Laboratoire d'Aide à la Procréation-CECOS, University Hospital of Grenoble, La Tronche, France (S.B.); Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5249, Laboratoire de Chimie et Biologie des Métaux, Grenoble, France (M.B.); and Department of Medicine, School of Clinical Sciences, Monash university and the Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia (P.M.)
| |
Collapse
|
60
|
Sarkar AA, Sabatino JA, Sugrue KF, Zohn IE. Abnormal labyrinthine zone in the Hectd1-null placenta. Placenta 2016; 38:16-23. [PMID: 26907377 PMCID: PMC4768308 DOI: 10.1016/j.placenta.2015.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 10/21/2015] [Accepted: 12/06/2015] [Indexed: 11/30/2022]
Abstract
INTRODUCTION The labyrinthine zone of the placenta is where exchange of nutrients and waste occurs between maternal and fetal circulations. Proper development of the placental labyrinth is essential for successful growth of the developing fetus and abnormalities in placental development are associated with intrauterine growth restriction (IUGR), preeclampsia and fetal demise. Our previous studies demonstrate that Hectd1 is essential for development of the junctional and labyrinthine zones of the placenta. Here we further characterize labyrinthine zone defects in the Hectd1 mutant placenta. METHODS The structure of the mutant placenta was compared to wildtype littermates using histological methods. The expression of cell type specific markers was examined by immunohistochemistry and in situ hybridization. RESULTS Hectd1 is expressed in the labyrinthine zone throughout development and the protein is enriched in syncytiotrophoblast layer type I cells (SynT-I) and Sinusoidal Trophoblast Giant cells (S-TGCs) in the mature placenta. Mutation of Hectd1 results in pale placentas with frequent hemorrhages along with gross abnormalities in the structure of the labyrinthine zone including a smaller overall volume and a poorly elaborated fetal vasculature that contain fewer fetal blood cells. Examination of molecular markers of labyrinthine trophoblast cell types reveals increased Dlx3 positive cells and Syna positive SynT-I cells, along with decreased Hand1 and Ctsq positive sinusoidal trophoblast giant cells (S-TGCs). DISCUSSION Together these defects indicate that Hectd1 is required for development of the labyrinthine zonethe mouse placenta.
Collapse
Affiliation(s)
- Anjali A Sarkar
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA
| | - Julia A Sabatino
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA
| | - Kelsey F Sugrue
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA; Institute for Biomedical Sciences, The George Washington University, Washington, DC 20052, USA
| | - Irene E Zohn
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA; Institute for Biomedical Sciences, The George Washington University, Washington, DC 20052, USA.
| |
Collapse
|
61
|
Van Gronigen Caesar G, Dale JM, Osman EY, Garcia ML, Lorson CL, Schulz LC. Placental development in a mouse model of spinal muscular atrophy. Biochem Biophys Res Commun 2015; 470:82-87. [PMID: 26748185 DOI: 10.1016/j.bbrc.2015.12.120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 12/22/2015] [Indexed: 01/30/2023]
Abstract
Spinal Muscular Atrophy (SMA) is an autosomal recessive disorder, leading to fatal loss of motor neurons. It is caused by loss of function of the SMN gene, which is expressed throughout the body, and there is increasing evidence of dysfunction in non-neuronal tissues. Birthweight is one of most powerful prognostic factors for infants born with SMA, and intrauterine growth restriction is common. In the SMNΔ7 mouse model of SMA, pups with the disease lived 25% longer when their mothers were fed a higher fat, "breeder" diet. The placenta is responsible for transport of nutrients from mother to fetus, and is a major determinant of fetal growth. Thus, the present study tested the hypothesis that placental development is impaired in SMNΔ7 conceptuses. Detailed morphological characterization revealed no defects in SMNΔ7 placental development, and expression of key transcription factors regulating mouse placental development was unaffected. The intrauterine growth restriction observed in SMA infants likely does not result from impaired placental development.
Collapse
Affiliation(s)
- Gerialisa Van Gronigen Caesar
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA; Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Jeffrey M Dale
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Erkan Y Osman
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA; Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Michael L Garcia
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Christian L Lorson
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA; Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA; Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Laura C Schulz
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA; Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
62
|
Sex Specification and Heterogeneity of Primordial Germ Cells in Mice. PLoS One 2015; 10:e0144836. [PMID: 26700643 PMCID: PMC4689518 DOI: 10.1371/journal.pone.0144836] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/23/2015] [Indexed: 11/19/2022] Open
Abstract
In mice, primordial germ cells migrate into the genital ridges by embryonic day 13.5 (E13.5), where they are then subjected to a sex-specific fate with female and male primordial germ cells undergoing mitotic arrest and meiosis, respectively. However, the sex-specific basis of primordial germ cell differentiation is poorly understood. The aim of this study was to investigate the sex-specific features of mouse primordial germ cells. We performed RNA-sequencing (seq) of E13.5 female and male mouse primordial germ cells using next-generation sequencing. We identified 651 and 428 differentially expressed transcripts (>2-fold, P < 0.05) in female and male primordial germ cells, respectively. Of these, many transcription factors were identified. Gene ontology and network analysis revealed differing functions of the identified female- and male-specific genes that were associated with primordial germ cell acquisition of sex-specific properties required for differentiation into germ cells. Furthermore, DNA methylation and ChIP-seq analysis of histone modifications showed that hypomethylated gene promoter regions were bound with H3K4me3 and H3K27me3. Our global transcriptome data showed that in mice, primordial germ cells are decisively assigned to a sex-specific differentiation program by E13.5, which is necessary for the development of vital germ cells.
Collapse
|
63
|
Tuteja G, Chung T, Bejerano G. Changes in the enhancer landscape during early placental development uncover a trophoblast invasion gene-enhancer network. Placenta 2015; 37:45-55. [PMID: 26604129 DOI: 10.1016/j.placenta.2015.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/21/2015] [Accepted: 11/02/2015] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Trophoblast invasion establishes adequate blood flow between mother and fetus in early placental development. However, little is known about the cis-regulatory mechanisms underlying this important process. We aimed to identify enhancer elements that are active during trophoblast invasion, and build a trophoblast invasion gene-enhancer network. METHODS We carried out ChIP-Seq for an enhancer-associated mark (H3k27Ac) at two time points during early placental development in mouse. One time point when invasion is at its peak (e7.5) and another time point shortly afterwards (e9.5). We use computational analysis to identify putative enhancers, as well as the transcription factor binding sites within them, that are specific to the time point of trophoblast invasion. RESULTS We compared read profiles at e7.5 and e9.5 to identify 1,977 e7.5-specific enhancers. Within a subset of e7.5-specific enhancers, we discovered a cell migration associated regulatory code, consisting of three transcription factor motifs: AP1, Ets, and Tcfap2. To validate differential expression of the transcription factors that bind these motifs, we performed RNA-Seq in the same context. Finally, we integrated these data with publicly available protein-protein interaction data and constructed a trophoblast invasion gene-enhancer network. DISCUSSION The data we generated and analysis we carried out improves our understanding of the regulatory mechanisms of trophoblast invasion, by suggesting a transcriptional code exists in the enhancers of cell migration genes. Furthermore, the network we constructed highlights novel candidate genes that may be critical for trophoblast invasion.
Collapse
Affiliation(s)
- Geetu Tuteja
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Tisha Chung
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Gill Bejerano
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA; Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Division of Medical Genetics, Department of Pediatrics, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
64
|
Koppes E, Himes KP, Chaillet JR. Partial Loss of Genomic Imprinting Reveals Important Roles for Kcnq1 and Peg10 Imprinted Domains in Placental Development. PLoS One 2015; 10:e0135202. [PMID: 26241757 PMCID: PMC4524636 DOI: 10.1371/journal.pone.0135202] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/19/2015] [Indexed: 01/24/2023] Open
Abstract
Mutations in imprinted genes or their imprint control regions (ICRs) produce changes in imprinted gene expression and distinct abnormalities in placental structure, indicating the importance of genomic imprinting to placental development. We have recently shown that a very broad spectrum of placental abnormalities associated with altered imprinted gene expression occurs in the absence of the oocyte-derived DNMT1o cytosine methyltransferase, which normally maintains parent-specific imprinted methylation during preimplantation. The absence of DNMT1o partially reduces inherited imprinted methylation while retaining the genetic integrity of imprinted genes and their ICRs. Using this novel system, we undertook a broad and inclusive approach to identifying key ICRs involved in placental development by correlating loss of imprinted DNA methylation with abnormal placental phenotypes in a mid-gestation window (E12.5-E15.5). To these ends we measured DNA CpG methylation at 15 imprinted gametic differentially methylated domains (gDMDs) that overlap known ICRs using EpiTYPER-mass array technology, and linked these epigenetic measurements to histomorphological defects. Methylation of some imprinted gDMDs, most notably Dlk1, was nearly normal in mid-gestation DNMT1o-deficient placentas, consistent with the notion that cells having lost methylation on these DMDs do not contribute significantly to placental development. Most imprinted gDMDs however showed a wide range of methylation loss among DNMT1o-deficient placentas. Two striking associations were observed. First, loss of DNA methylation at the Peg10 imprinted gDMD associated with decreased embryonic viability and decreased labyrinthine volume. Second, loss of methylation at the Kcnq1 imprinted gDMD was strongly associated with trophoblast giant cell (TGC) expansion. We conclude that the Peg10 and Kcnq1 ICRs are key regulators of mid-gestation placental function.
Collapse
Affiliation(s)
- Erik Koppes
- Magee-Womens Research Institute, Program in Integrative Molecular Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Katherine P. Himes
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - J. Richard Chaillet
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
65
|
Li Q, Suasnavas E, Xiao L, Heywood S, Qi X, Zhou A, Isom SC. Label-free and non-invasive monitoring of porcine trophoblast derived cells: differentiation in serum and serum-free media. JOURNAL OF BIOPHOTONICS 2015; 8:638-645. [PMID: 25250877 DOI: 10.1002/jbio.201400062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 08/03/2014] [Accepted: 08/27/2014] [Indexed: 06/03/2023]
Abstract
Traditional approaches to characterize stem cell differentiation are time-consuming, lengthy and invasive. Here, Raman microspectroscopy (RM) and atomic force microscopy (AFM) - both considered as non-invasive techniques - are applied to detect the biochemical and biophysical properties of trophoblast derived stem-like cells incubated up to 10 days under conditions designed to induce differentiation. Significant biochemical and biophysical differences between control cells and differentiated cells were observed. Quantitative real time PCR was also applied to analyze gene expression. The relationship between cell differentiation and associated cellular biochemical and biomechanical changes were discussed. Monitoring trophoblast cells differentiation.
Collapse
Affiliation(s)
- Qifei Li
- Department of Biological Engineering, Utah State University, Logan, UT, 84322
| | - Edison Suasnavas
- Department of Animal Science, Dairy and Veterinary Sciences, Utah State University, Logan, UT, 84322
| | - Lifu Xiao
- Department of Biological Engineering, Utah State University, Logan, UT, 84322
| | - Sierra Heywood
- Department of Animal Science, Dairy and Veterinary Sciences, Utah State University, Logan, UT, 84322
| | - Xiaojun Qi
- Department of Computer Science, Utah State University, Logan, UT, 84322
| | - Anhong Zhou
- Department of Biological Engineering, Utah State University, Logan, UT, 84322
| | - S Clay Isom
- Department of Animal Science, Dairy and Veterinary Sciences, Utah State University, Logan, UT, 84322
| |
Collapse
|
66
|
Tabata T, Petitt M, Zydek M, Fang-Hoover J, Larocque N, Tsuge M, Gormley M, Kauvar LM, Pereira L. Human cytomegalovirus infection interferes with the maintenance and differentiation of trophoblast progenitor cells of the human placenta. J Virol 2015; 89:5134-47. [PMID: 25741001 PMCID: PMC4403461 DOI: 10.1128/jvi.03674-14] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 01/19/2015] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Human cytomegalovirus (HCMV) is a major cause of birth defects that include severe neurological deficits, hearing and vision loss, and intrauterine growth restriction. Viral infection of the placenta leads to development of avascular villi, edema, and hypoxia associated with symptomatic congenital infection. Studies of primary cytotrophoblasts (CTBs) revealed that HCMV infection impedes terminal stages of differentiation and invasion by various molecular mechanisms. We recently discovered that HCMV arrests earlier stages involving development of human trophoblast progenitor cells (TBPCs), which give rise to the mature cell types of chorionic villi-syncytiotrophoblasts on the surfaces of floating villi and invasive CTBs that remodel the uterine vasculature. Here, we show that viral proteins are present in TBPCs of the chorion in cases of symptomatic congenital infection. In vitro studies revealed that HCMV replicates in continuously self-renewing TBPC lines derived from the chorion and alters expression and subcellular localization of proteins required for cell cycle progression, pluripotency, and early differentiation. In addition, treatment with a human monoclonal antibody to HCMV glycoprotein B rescues differentiation capacity, and thus, TBPCs have potential utility for evaluation of the efficacies of novel antiviral antibodies in protecting and restoring placental development. Our results suggest that HCMV replicates in TBPCs in the chorion in vivo, interfering with the earliest steps in the growth of new villi, contributing to virus transmission and impairing compensatory development. In cases of congenital infection, reduced responsiveness of the placenta to hypoxia limits the transport of substances from maternal blood and contributes to fetal growth restriction. IMPORTANCE Human cytomegalovirus (HCMV) is a leading cause of birth defects in the United States. Congenital infection can result in permanent neurological defects, mental retardation, hearing loss, visual impairment, and pregnancy complications, including intrauterine growth restriction, preterm delivery, and stillbirth. Currently, there is neither a vaccine nor any approved treatment for congenital HCMV infection during gestation. The molecular mechanisms underlying structural deficiencies in the placenta that undermine fetal development are poorly understood. Here we report that HCMV replicates in trophoblast progenitor cells (TBPCs)-precursors of the mature placental cells, syncytiotrophoblasts and cytotrophoblasts, in chorionic villi-in clinical cases of congenital infection. Virus replication in TBPCs in vitro dysregulates key proteins required for self-renewal and differentiation and inhibits normal division and development into mature placental cells. Our findings provide insights into the underlying molecular mechanisms by which HCMV replication interferes with placental maturation and transport functions.
Collapse
Affiliation(s)
- Takako Tabata
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California, USA
| | - Matthew Petitt
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California, USA
| | - Martin Zydek
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California, USA
| | - June Fang-Hoover
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California, USA
| | - Nicholas Larocque
- Center for Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA The Eli & Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA
| | - Mitsuru Tsuge
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California, USA
| | - Matthew Gormley
- Center for Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA The Eli & Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA
| | | | - Lenore Pereira
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
67
|
Pappas A, Chaiworapongsa T, Romero R, Korzeniewski SJ, Cortez JC, Bhatti G, Gomez-Lopez N, Hassan SS, Shankaran S, Tarca AL. Transcriptomics of maternal and fetal membranes can discriminate between gestational-age matched preterm neonates with and without cognitive impairment diagnosed at 18-24 months. PLoS One 2015; 10:e0118573. [PMID: 25822971 PMCID: PMC4379164 DOI: 10.1371/journal.pone.0118573] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/20/2015] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Neurocognitive impairment among children born preterm may arise from complex interactions between genes and the intra-uterine environment. OBJECTIVES (1) To characterize the transcriptomic profiles of chorioamniotic membranes in preterm neonates with and without neurocognitive impairment via microarrays and (2) to determine if neonates with neurocognitive impairment can be identified at birth. MATERIALS/METHODS A retrospective case-control study was conducted to examine the chorioamniotic transcriptome of gestational-age matched very preterm neonates with and without neurocognitive impairment at 18-24 months' corrected-age defined by a Bayley-III Cognitive Composite Score <80 (n = 14 each). Pathway analysis with down-weighting of overlapping genes (PADOG) was performed to identify KEGG pathways relevant to the phenotype. Select differentially expressed genes were profiled using qRT-PCR and a multi-gene disease prediction model was developed using linear discriminant analysis. The model's predictive performance was tested on a new set of cases and controls (n = 19 each). RESULTS 1) 117 genes were differentially expressed among neonates with and without subsequent neurocognitive impairment (p<0.05 and fold change >1.5); 2) Gene ontology analysis indicated enrichment of 19 biological processes and 3 molecular functions; 3)PADOG identified 4 significantly perturbed KEGG pathways: oxidative phosphorylation, Parkinson's disease, Alzheimer's disease and Huntington's disease (q-value <0.1); 4) 48 of 90 selected differentially expressed genes were confirmed by qRT-PCR, including genes implicated in energy metabolism, neuronal signaling, vascular permeability and response to injury (e.g., up-regulation of SEPP1, APOE, DAB2, CD163, CXCL12, VWF; down-regulation of HAND1, OSR1)(p<0.05); and 5) a multi-gene model predicted 18-24 month neurocognitive impairment (using the ratios of OSR1/VWF and HAND1/VWF at birth) in a larger, independent set (sensitivity = 74%, at specificity = 83%). CONCLUSIONS Gene expression patterns in the chorioamniotic membranes link neurocognitive impairment in preterm infants to neurodegenerative disease pathways and might be used to predict neurocognitive impairment. Further prospective studies are needed.
Collapse
Affiliation(s)
- Athina Pappas
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Pediatrics, Division of Neonatal and Perinatal Medicine, Wayne State University, Detroit, MI, United States of America
- * E-mail: (AP); (AT)
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States of America
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States of America
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States of America
| | - Steven J. Korzeniewski
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States of America
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States of America
| | - Josef C. Cortez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Pediatrics, Division of Neonatal and Perinatal Medicine, Wayne State University, Detroit, MI, United States of America
| | - Gaurav Bhatti
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States of America
- Department of Immunology and Microbiology, Wayne State University, Detroit, MI, United States of America
| | - Sonia S. Hassan
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States of America
| | - Seetha Shankaran
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Pediatrics, Division of Neonatal and Perinatal Medicine, Wayne State University, Detroit, MI, United States of America
| | - Adi L. Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States of America
- * E-mail: (AP); (AT)
| |
Collapse
|
68
|
Watkins AJ, Lucas ES, Marfy-Smith S, Bates N, Kimber SJ, Fleming TP. Maternal nutrition modifies trophoblast giant cell phenotype and fetal growth in mice. Reproduction 2015; 149:563-75. [PMID: 25755287 PMCID: PMC4418750 DOI: 10.1530/rep-14-0667] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/09/2015] [Indexed: 01/15/2023]
Abstract
Mammalian placentation is dependent upon the action of trophoblast cells at the time of implantation. Appropriate fetal growth, regulated by maternal nutrition and nutrient transport across the placenta, is a critical factor for adult offspring long-term health. We have demonstrated that a mouse maternal low-protein diet (LPD) fed exclusively during preimplantation development (Emb-LPD) increases offspring growth but programmes adult cardiovascular and metabolic disease. In this study, we investigate the impact of maternal nutrition on post-implantation trophoblast phenotype and fetal growth. Ectoplacental cone explants were isolated at day 8 of gestation from female mice fed either normal protein diet (NPD: 18% casein), LPD (9% casein) or Emb-LPD and cultured in vitro. We observed enhanced spreading and cell division within proliferative and secondary trophoblast giant cells (TGCs) emerging from explants isolated from LPD-fed females when compared with NPD and Emb-LPD explants after 24 and 48 h. Moreover, both LPD and Emb-LPD explants showed substantial expansion of TGC area during 24–48 h, not observed in NPD. No difference in invasive capacity was observed between treatments using Matrigel transwell migration assays. At day 17 of gestation, LPD- and Emb-LPD-fed conceptuses displayed smaller placentas and larger fetuses respectively, resulting in increased fetal:placental ratios in both groups compared with NPD conceptuses. Analysis of placental and yolk sac nutrient signalling within the mammalian target of rapamycin complex 1 pathway revealed similar levels of total and phosphorylated downstream targets across groups. These data demonstrate that early post-implantation embryos modify trophoblast phenotype to regulate fetal growth under conditions of poor maternal nutrition.
Collapse
Affiliation(s)
- Adam J Watkins
- Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Emma S Lucas
- Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Stephanie Marfy-Smith
- Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Nicola Bates
- Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Susan J Kimber
- Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Tom P Fleming
- Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
69
|
Isolation and characterization of trophoblast-derived stem-like cells from peri-implantation porcine embryos. Anim Reprod Sci 2015; 154:128-41. [PMID: 25660622 DOI: 10.1016/j.anireprosci.2015.01.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 12/09/2014] [Accepted: 01/14/2015] [Indexed: 12/13/2022]
Abstract
In mammals, the trophoblast lineage of the embryo is specified before attachment/implantation to become the fetal portion of the placenta. Trophoblast-derived cells were isolated and cultured from day 10 and day 13 porcine embryos and were grown in vitro in a defined, serum-free culture medium for over 2 years without showing any signs of senescence. However, trophoblast-derived cells placed into serum-containing medium rapidly senesce and fail to proliferate. Semiquantitative and quantitative gene expression analyses of cells in culture from 0 to 30 days confirmed the presence (and relative abundance) of mRNA transcripts from genes involved in trophoblast function (CDX2, TEAD4, CYP17A1, HSD17B1, FGFR2, PLET, HAND1) as well as some genes known to mediate pluripotency (POU5F1, KLF4, CMYC). Protein immunolocalization demonstrated expression of both trophoblast and mesenchymal cell markers. DNA methylation patterns in promoters of three critical developmental genes (HAND1, KLF4, TEAD4) did not change appreciably over 4 months of culture in vitro. It was demonstrated that these trophoblast-derived cells are easily stably transfected with an exogenous transgene (eGFP) by a variety of methods, and show the ability to survive and to be passaged repeatedly after transfection. In summary, early embryonic porcine trophoblast-derived cells have demonstrated unique characteristics, which means they could be used as valuable tools for laboratory work. Anticipated applications include the study of trophoblast physiology as well as possible solutions for improving efficiency of transgenesis by somatic cell nuclear transfer and for pluripotency reprogramming of cells.
Collapse
|
70
|
Rai A, Cross JC. Three-dimensional cultures of trophoblast stem cells autonomously develop vascular-like spaces lined by trophoblast giant cells. Dev Biol 2014; 398:110-9. [PMID: 25499676 DOI: 10.1016/j.ydbio.2014.11.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 11/12/2014] [Accepted: 11/26/2014] [Indexed: 10/24/2022]
Abstract
The maternal blood space in the mouse placenta is lined not by endothelial cells but rather by various subtypes of trophoblast giant cells (TGCs), defined by their location and different patterns of gene expression. While TGCs invade the spiral arteries to displace the maternal endothelium, the rest of the vascular space is created de novo but the mechanisms are not well understood. We cultured mouse trophoblast stem (TS) cells in suspension and found that they readily form spheroids (trophospheres). Compared to cells grown in monolayer, differentiating trophospheres showed accelerated expression of TGC-specific genes. Morphological and gene expression studies showed that cavities form within the trophospheres that are primarily lined by Prl3d1/Pl1α-positive cells analogous to parietal-TGCs (P-TGCs) which line the maternal venous blood within the placenta. Lumen formation in trophospheres and in vivo was associated with cell polarization including CD34 sialomucin deposition on the apical side and cytoskeletal rearrangement. While P-TGCs preferentially formed in trophospheres at atmospheric oxygen levels (19%), decreasing oxygen to 3% shifted differentiation towards Ctsq-positive sinusoidal and/or channel TGCs. These studies show that trophoblast cells have the intrinsic ability to form vascular channels in ways analogous to endothelial cells. The trophosphere system will be valuable for assessing mechanisms that regulate specification of different TGC subtypes and their morphogenesis into vascular spaces.
Collapse
Affiliation(s)
- Anshita Rai
- Departments of Biochemistry and Molecular Biology, Comparative Biology and Experimental Medicine, Obstetrics and Gynecology, and Medical Genetics, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1
| | - James C Cross
- Departments of Biochemistry and Molecular Biology, Comparative Biology and Experimental Medicine, Obstetrics and Gynecology, and Medical Genetics, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1.
| |
Collapse
|
71
|
Sarkar AA, Nuwayhid SJ, Maynard T, Ghandchi F, Hill JT, Lamantia AS, Zohn IE. Hectd1 is required for development of the junctional zone of the placenta. Dev Biol 2014; 392:368-80. [PMID: 24855001 PMCID: PMC4578812 DOI: 10.1016/j.ydbio.2014.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 05/06/2014] [Accepted: 05/07/2014] [Indexed: 01/17/2023]
Abstract
The placenta plays a critical role in the growth and survival of the fetus. Here we demonstrate that the Homologous to the E6-AP Carboxyl Terminus (HECT) domain E3 ubiquitin ligase, Hectd1, is essential for development of the mouse placenta. Hectd1 is widely expressed during placentation with enrichment in trophoblast giant cells (TGCs) and other trophoblast-derived cell subtypes in the junctional and labyrinth zones of the placenta. Disruption of Hectd1 results in mid-gestation lethality and intrauterine growth restriction (IUGR). Variable defects in the gross structure of the mutant placenta are found including alterations in diameter, thickness and lamination. The number and nuclear size of TGCs is reduced. Examination of subtype specific markers reveals altered TGC development with decreased expression of Placental lactogen-1 and -2 (Pl1 and Pl2) and increased expression of Proliferin (Plf). Reduced numbers of spongiotrophoblasts and glycogen trophoblasts were also found at the junctional zone of the Hectd1 mutant placenta. Finally, there was an increase in immature uterine natural killer (uNK) cells in the maternal decidua of the Hectd1 mutant placenta. Proliferation and apoptosis are differentially altered in the layers of the placenta with an increase in both apoptosis and proliferation in the maternal decidua, a decrease in proliferation and increase in apoptosis in the labyrinth layer and both unchanged in the junctional zone. Together these data demonstrate that Hectd1 is required for development of multiple cell types within the junctional zone of the placenta.
Collapse
Affiliation(s)
- Anjali A Sarkar
- Center for Neuroscience Research, Children׳s Research Institute, and Children׳s National Medical Center, Washington, DC 20010, USA
| | - Samer J Nuwayhid
- Center for Neuroscience Research, Children׳s Research Institute, and Children׳s National Medical Center, Washington, DC 20010, USA
| | - Thomas Maynard
- Department of Pharmacology and Physiology, The George Washington Institute for Neuroscience, George Washington University, Washington, DC 20052, USA; The George Washington Institute for Neuroscience, George Washington University, Washington, DC 20052, USA
| | - Frederick Ghandchi
- Center for Neuroscience Research, Children׳s Research Institute, and Children׳s National Medical Center, Washington, DC 20010, USA
| | | | - Anthony S Lamantia
- Department of Pharmacology and Physiology, The George Washington Institute for Neuroscience, George Washington University, Washington, DC 20052, USA; The George Washington Institute for Neuroscience, George Washington University, Washington, DC 20052, USA
| | - Irene E Zohn
- Center for Neuroscience Research, Children׳s Research Institute, and Children׳s National Medical Center, Washington, DC 20010, USA; Department of Pharmacology and Physiology, The George Washington Institute for Neuroscience, George Washington University, Washington, DC 20052, USA; The George Washington Institute for Neuroscience, George Washington University, Washington, DC 20052, USA.
| |
Collapse
|
72
|
Koo BK, Clevers H. Stem cells marked by the R-spondin receptor LGR5. Gastroenterology 2014; 147:289-302. [PMID: 24859206 DOI: 10.1053/j.gastro.2014.05.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 05/12/2014] [Accepted: 05/16/2014] [Indexed: 12/14/2022]
Abstract
Since the discovery of LGR5 as a marker of intestinal stem cells, the field has developed explosively and led to many new avenues of research. The inner workings of the intestinal crypt stem cell niche are now well understood. The study of stem cell-enriched genes has uncovered some previously unknown aspects of the Wnt signaling pathway, the major driver of crypt dynamics. LGR5(+) stem cells can now be cultured over long periods in vitro as epithelial organoids or "mini-guts." This technology opens new possibilities of using cultured adult stem cells for drug development, disease modeling, gene therapy, and regenerative medicine. This review describes the rediscovery of crypt base columnar cells as LGR5(+) adult stem cells and summarizes subsequent progress, promises, unresolved issues, and challenges of the field.
Collapse
Affiliation(s)
- Bon-Kyoung Koo
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, England; Department of Genetics, University of Cambridge, Cambridge, England
| | - Hans Clevers
- Hubrecht Institute/KNAW, Utrecht, The Netherlands; University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
73
|
Integrative epigenome analysis identifies a Polycomb-targeted differentiation program as a tumor-suppressor event epigenetically inactivated in colorectal cancer. Cell Death Dis 2014; 5:e1324. [PMID: 25032847 PMCID: PMC4123077 DOI: 10.1038/cddis.2014.283] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 05/26/2014] [Accepted: 05/27/2014] [Indexed: 12/14/2022]
Abstract
Aberrant DNA hypermethylation in human cancer has been associated with Polycomb target genes in embryonic stem (ES) cells, but a functional link of the Polycomb-targeted differentiation program to tumorigenesis remains to be established. Here, through epigenome analysis correlating DNA hypermethylation in colon cancer with ES cell pluripotency and differentiation, we identified a set of DNA hypermethylated genes in cancer cells that are Polycomb targets strongly associated with ES cell differentiation, including HAND1, a developmental regulator. Intriguingly, HAND1 is silenced in over 90% of human primary colorectal tumors, and re-expression of HAND1 in colon cancer cells induces terminal differentiation, inhibits proliferation and prevents xenograft tumor formation. Moreover, hypermethylated HAND1 has a minimum enrichment of EZH2-H3K27me3 in cancer cells, but becomes EZH2 bound and bivalent upon the loss of DNA methylation, suggesting a sequential gene silencing event during oncogenesis. These findings established a functional role of Polycomb-targeted differentiation program as a tumor-suppressor event epigenetically inactivated in human cancer.
Collapse
|
74
|
Asuthkar S, Gogineni VR, Rao JS, Velpula KK. Nuclear Translocation of Hand-1 Acts as a Molecular Switch to Regulate Vascular Radiosensitivity in Medulloblastoma Tumors: The Protein uPAR Is a Cytoplasmic Sequestration Factor for Hand-1. Mol Cancer Ther 2014; 13:1309-22. [DOI: 10.1158/1535-7163.mct-13-0892] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
75
|
Holloway AC, Salomon A, Soares MJ, Garnier V, Raha S, Sergent F, Nicholson CJ, Feige JJ, Benharouga M, Alfaidy N. Characterization of the adverse effects of nicotine on placental development: in vivo and in vitro studies. Am J Physiol Endocrinol Metab 2014; 306:E443-56. [PMID: 24368670 PMCID: PMC4865199 DOI: 10.1152/ajpendo.00478.2013] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In utero exposure to nicotine is associated with increased risk of numerous adverse fetal and neonatal outcomes, which suggests that it acts directly to affect placental development and the establishment of the fetomaternal circulation (FC). This study used both in vivo [Wistar rats treated with 1 mg/kg nicotine from 2 wk prior to mating until gestational day (GD) 15] and in vitro (RCHO-1 cell line; treated with 10(-9) to 10(-3)M nicotine) models to examine the effects of nicotine on these pathways. At GD 15, control and treated placentas were examined for the impact of nicotine on 1) trophoblast invasion, proliferation, and degree of hypoxia, 2) labyrinth vascularization, 3) expression of key genes of placental development, and 4) expression of placental angiogenic factors. The RCHO-1 cell line was used to determine the direct effects of nicotine on trophoblast differentiation. Our in vivo experiments show that nicotine inhibits trophoblast interstitial invasion, increases placental hypoxia, downregulates labyrinth vascularization as well as key transcription factors Hand1 and GCM1, and decreases local and circulating EG-VEGF, a key placental angiogenic factor. The in vitro experiments confirmed the inhibitory effects of nicotine on the trophoblast migration, invasion, and differentiation processes and demonstrated that those effects are most likely due to a dysregulation in the expression of nicotine receptors and a decrease in MMP9 activity. Taken together, these data suggest that adverse effects of maternal smoking on pregnancy outcome are due in part to direct and endocrine effects of nicotine on the main processes of placental development and establishment of FC.
Collapse
Affiliation(s)
- A. C. Holloway
- 5Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada;
| | - A. Salomon
- 2Commissariat à l'Energie Atomique, Grenoble, France;
- 3Université Joseph Fourrier, Grenoble, France;
- 4Institut National de la Santé et de la Recherche Médicale, Grenoble, France;
| | - M. J. Soares
- 7Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - V. Garnier
- 2Commissariat à l'Energie Atomique, Grenoble, France;
- 3Université Joseph Fourrier, Grenoble, France;
- 4Institut National de la Santé et de la Recherche Médicale, Grenoble, France;
| | - S. Raha
- 6Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada; and
| | - F. Sergent
- 2Commissariat à l'Energie Atomique, Grenoble, France;
- 3Université Joseph Fourrier, Grenoble, France;
- 4Institut National de la Santé et de la Recherche Médicale, Grenoble, France;
| | - C. J. Nicholson
- 5Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada;
| | - J. J. Feige
- 2Commissariat à l'Energie Atomique, Grenoble, France;
- 3Université Joseph Fourrier, Grenoble, France;
- 4Institut National de la Santé et de la Recherche Médicale, Grenoble, France;
| | - M. Benharouga
- 1Centre National de la Recherche Scientifique, Grenoble, France;
- 2Commissariat à l'Energie Atomique, Grenoble, France;
- 3Université Joseph Fourrier, Grenoble, France;
| | - N. Alfaidy
- 2Commissariat à l'Energie Atomique, Grenoble, France;
- 3Université Joseph Fourrier, Grenoble, France;
- 4Institut National de la Santé et de la Recherche Médicale, Grenoble, France;
| |
Collapse
|
76
|
Rai A, Cross JC. Development of the hemochorial maternal vascular spaces in the placenta through endothelial and vasculogenic mimicry. Dev Biol 2014; 387:131-41. [PMID: 24485853 DOI: 10.1016/j.ydbio.2014.01.015] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 01/14/2014] [Accepted: 01/19/2014] [Indexed: 11/27/2022]
Abstract
The maternal vasculature within the placenta in primates and rodents is unique because it is lined by fetal cells of the trophoblast lineage and not by maternal endothelial cells. In addition to trophoblast cells that invade the uterine spiral arteries that bring blood into the placenta, other trophoblast subtypes sit at different levels of the vascular space. In mice, at least five distinct subtypes of trophoblast cells have been identified which engage maternal endothelial cells on the arterial and venous frontiers of the placenta, but which also form the channel-like spaces within it through a process analogous to formation of blood vessels (vasculogenic mimicry). These cells are all large, post-mitotic trophoblast giant cells. In addition to assuming endothelial cell-like characteristics (endothelial mimicry), they produce dozens of different hormones that are thought to regulate local and systemic maternal adaptations to pregnancy. Recent work has identified distinct molecular pathways in mice that regulate the morphogenesis of trophoblast cells on the arterial and venous sides of the vascular circuit that may be analogous to specification of arterial and venous endothelial cells.
Collapse
Affiliation(s)
- Anshita Rai
- Department of Biochemistry and Molecular Biology, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, Alta., Canada; Department of Comparative Biology and Experimental Medicine, Obstetrics and Gynecology, and Medical Genetics, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1
| | - James C Cross
- Department of Biochemistry and Molecular Biology, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, Alta., Canada; Department of Comparative Biology and Experimental Medicine, Obstetrics and Gynecology, and Medical Genetics, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1.
| |
Collapse
|
77
|
Hirasawa R, Matoba S, Inoue K, Ogura A. Somatic donor cell type correlates with embryonic, but not extra-embryonic, gene expression in postimplantation cloned embryos. PLoS One 2013; 8:e76422. [PMID: 24146866 PMCID: PMC3797840 DOI: 10.1371/journal.pone.0076422] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 08/23/2013] [Indexed: 11/19/2022] Open
Abstract
The great majority of embryos generated by somatic cell nuclear transfer (SCNT) display defined abnormal phenotypes after implantation, such as an increased likelihood of death and abnormal placentation. To gain better insight into the underlying mechanisms, we analyzed genome-wide gene expression profiles of day 6.5 postimplantation mouse embryos cloned from three different cell types (cumulus cells, neonatal Sertoli cells and fibroblasts). The embryos retrieved from the uteri were separated into embryonic (epiblast) and extraembryonic (extraembryonic ectoderm and ectoplacental cone) tissues and were subjected to gene microarray analysis. Genotype- and sex-matched embryos produced by in vitro fertilization were used as controls. Principal component analysis revealed that whereas the gene expression patterns in the embryonic tissues varied according to the donor cell type, those in extraembryonic tissues were relatively consistent across all groups. Within each group, the embryonic tissues had more differentially expressed genes (DEGs) (>2-fold vs. controls) than did the extraembryonic tissues (P<1.0×10–26). In the embryonic tissues, one of the common abnormalities was upregulation of Dlk1, a paternally imprinted gene. This might be a potential cause of the occasional placenta-only conceptuses seen in SCNT-generated mouse embryos (1–5% per embryos transferred in our laboratory), because dysregulation of the same gene is known to cause developmental failure of embryos derived from induced pluripotent stem cells. There were also some DEGs in the extraembryonic tissues, which might explain the poor development of SCNT-derived placentas at early stages. These findings suggest that SCNT affects the embryonic and extraembryonic development differentially and might cause further deterioration in the embryonic lineage in a donor cell-specific manner. This could explain donor cell-dependent variations in cloning efficiency using SCNT.
Collapse
Affiliation(s)
| | - Shogo Matoba
- RIKEN BioResouce Center, Tsukuba, Ibaraki, Japan
| | - Kimiko Inoue
- RIKEN BioResouce Center, Tsukuba, Ibaraki, Japan
- Graduate School of Life and Environmental Science, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Atsuo Ogura
- RIKEN BioResouce Center, Tsukuba, Ibaraki, Japan
- Graduate School of Life and Environmental Science, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Center for Disease Biology and Integrative Medicine, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
78
|
Schiffmacher AT, Keefer CL. CDX2 regulates multiple trophoblast genes in bovine trophectoderm CT-1 cells. Mol Reprod Dev 2013; 80:826-39. [PMID: 23836438 DOI: 10.1002/mrd.22212] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 06/27/2013] [Indexed: 11/06/2022]
Abstract
The bovine trophectoderm (TE) undergoes a dramatic morphogenetic transition prior to uterine endometrial attachment. Many studies have documented trophoblast-specific gene expression profiles at various pre-attachment stages, yet genetic interactions within the transitioning TE gene regulatory network are not well characterized. During bovine embryogenesis, transcription factors OCT4 and CDX2 are co-expressed during early trophoblast elongation. In this study, the bovine trophectoderm-derived CT-1 cell line was utilized as a genetic model to examine the roles of CDX2 and OCT4 within the bovine trophoblast gene regulatory network. An RT-PCR screen for TE-lineage transcription factors identified expression of CDX2, ERRB, ID2, SOX15, ELF5, HAND1, and ASCL2. CT-1 cells also express a nuclear-localized, 360 amino acid OCT4 ortholog of the pluripotency-specific human OCT4A. To delineate the roles of CDX2 and OCT4 within the CT-1 gene network, CDX2 and OCT4 levels were manipulated via overexpression and siRNA-mediated knockdown. An increase in CDX2 negatively regulated OCT4 expression, but increased expression of IFNT, HAND1, ASCL2, SOX15, and ELF5. A reduction of CDX2 levels exhibited a reciprocal effect, resulting in decreased expression of IFNT, HAND1, ASCL2, and SOX15. Both overexpression and knockdown of CDX2 increased ETS2 transcription. In contrast to CDX2, manipulation of OCT4 levels only revealed a positive autoregulatory mechanism and upregulation of ASCL2. Together, these results suggest that CDX2 is a core regulator of multiple trophoblast genes within CT-1 cells.
Collapse
Affiliation(s)
- Andrew T Schiffmacher
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | | |
Collapse
|
79
|
Gasperowicz M, Surmann-Schmitt C, Hamada Y, Otto F, Cross JC. The transcriptional co-repressor TLE3 regulates development of trophoblast giant cells lining maternal blood spaces in the mouse placenta. Dev Biol 2013; 382:1-14. [PMID: 23954203 DOI: 10.1016/j.ydbio.2013.08.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 08/07/2013] [Accepted: 08/08/2013] [Indexed: 11/17/2022]
Abstract
TLE3 is a transcriptional co-repressor that interacts with several DNA-binding repressors, including downstream effectors of the Notch signaling pathway. We generated Tle3-deficient mice and found that they die in utero and their death is associated with abnormal development of the placenta with major defects in the maternal vasculature. In the normal placenta, maternal blood spaces are lined, not as usual in the mammalian circulation by endothelial cells, but rather by specialized embryo-derived cells of the trophoblast cell lineage named trophoblast giant cells (TGC). Tle3 mRNA is expressed in those specialized TGC and Tle3 mutants show severe defects in differentiation of TGC-lined channels and lacunar spaces that take blood out of the labyrinth zone of the placenta and into the uterine veins. The mutants also show somewhat milder defects on the arterial-side of the maternal vascular circuit in spiral arteries and canals that take blood into the labyrinth. Notch2 and Tle3 expression patterns overlap in several TGC subtypes and we found that Tle3 and Notch2 mutants have some overlapping features. However, they also show differences implying that TLE3 may mediate some but not all of the effects of Notch2 signaling during placenta development. Therefore, formation of the different types of maternal blood spaces by different TGC subtypes is regulated by distinct molecular mechanisms.
Collapse
Affiliation(s)
- Malgorzata Gasperowicz
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Room HSC 2279, 3330 Hospital Drive N.W., Calgary, Alberta, Canada T2N 4N1
| | | | | | | | | |
Collapse
|
80
|
Hu D, Scott IC, Snider F, Geary-Joo C, Zhao X, Simmons DG, Cross JC. The basic helix-loop-helix transcription factor Hand1 regulates mouse development as a homodimer. Dev Biol 2013; 382:470-81. [PMID: 23911935 DOI: 10.1016/j.ydbio.2013.07.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 07/04/2013] [Accepted: 07/28/2013] [Indexed: 10/26/2022]
Abstract
Hand1 is a basic helix-loop-helix transcription factor that is essential for development of the placenta, yolk sac and heart during mouse development. While Hand1 is essential for trophoblast giant cell (TGC) differentiation, its potential heterodimer partners are not co-expressed in TGCs. To test the hypothesis that Hand1 functions as homodimer, we generated knock-in mice in which the Hand1 gene was altered to encode a tethered homodimer (TH). Some Hand1(TH/-) conceptuses in which the only form of Hand1 is Hand1(TH) are viable and fertile, indicating that homodimer Hand1 is sufficient for mouse survival. ~2/3 of Hand1(TH/-) and all Hand1(TH/TH) mice died in utero and displayed severe placental defects and variable cardial and cranial-facial abnormalities, indicating a dosage-dependent effect of Hand1(TH). Meanwhile, expression of the Hand1(TH) protein did not have negative effects on viability or fertility in all Hand1(TH/+) mice. These data imply that Hand1 homodimer plays a dominant role during development and its expression dosage is critical for survival, whereas Hand1 heterodimers can be either dispensable or play a regulatory role to modulate the activity of Hand1 homodimer in vivo.
Collapse
Affiliation(s)
- Dong Hu
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, AB, Canada; Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada; Center for Stem Cell Application and Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | | | | | |
Collapse
|
81
|
Henke C, Ruebner M, Faschingbauer F, Stolt CC, Schaefer N, Lang N, Beckmann MW, Strissel PL, Strick R. Regulation of murine placentogenesis by the retroviral genes Syncytin-A, Syncytin-B and Peg10. Differentiation 2013; 85:150-60. [PMID: 23807393 DOI: 10.1016/j.diff.2013.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 02/05/2013] [Accepted: 02/06/2013] [Indexed: 01/27/2023]
Abstract
The murine placenta has a trichorial structure with two multinucleated syncytiotrophoblast (SCT) layers representing a barrier between the maternal and fetal blood system. Genes of endogenous retroviruses and retrotransposon-derived paternally expressed genes (Peg), remnants of past infections and integrations in the genome, have essential functions in placentogenesis. Previous studies showed that the envelope genes Syncytin-A and Syncytin-B were essential for cell-cell fusion of the SCT. The goal of this study was to analyze the temporal localization and expression of nine genes throughout placental development from embryonic day (E)8.5 to E18.5 using in situ-hybridization and absolute RNA-quantification. These included a comparison of previously characterized genes from the labyrinth Syncytin-A, Syncytin-B, Gcm1, the junctional zone PL-1, PL-2, Plf, Tpbpa with two further characterized genes Peg10 and Tpbpb. Syncytin-A and Syncytin-B RNA localized to SCT-I and SCT-II, respectively. Peg10 RNA localized to all extraembryonic tissues, specifically to the parietal and sinusoidal TGC of the labyrinth layer, which is in contact with SCT-I and the maternal blood. All three retroviral/retrotransposon-derived genes showed the highest expression at E16.5, but Peg10 with 188,917.1 molecules/ng cDNA was 208-fold and 106.8-fold higher expressed than Syncytin-A and Syncytin-B, respectively. Tpbpb localized to the junctional zone and showed the highest expression at E16.5 along with PL-2, Plf, Tpbpa, but not PL-1, which decreased in expression at E10.5. To investigate a role of Syncytin-A, Syncytin-B and Peg10 in cell-cell fusion, we established a cell culture system with fractionated primary trophoblasts from murine placentae. Culturing trophoblasts for up to 72h partly resembled trophoblast development in vivo according to the nine marker genes. Knockdown of Syncytin-A demonstrated a functional regulation of cell-cell fusion, where knockdown of Peg10 showed no involvement in cell fusion. Due to the expression of Peg10 in TGCs, we propose an essential functional role in the fetal-maternal blood system.
Collapse
Affiliation(s)
- Christine Henke
- University-Clinic Erlangen, Department of Gynaecology and Obstetrics, Laboratory for Molecular Medicine, Universitaetsstr. 21-23, D-91054 Erlangen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Zhou W, Suntharalingam K, Brand NJ, Barton PJR, Vilar R, Ying L. Possible regulatory roles of promoter g-quadruplexes in cardiac function-related genes - human TnIc as a model. PLoS One 2013; 8:e53137. [PMID: 23326389 PMCID: PMC3541360 DOI: 10.1371/journal.pone.0053137] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 11/23/2012] [Indexed: 12/15/2022] Open
Abstract
G-quadruplexes (G4s) are four-stranded DNA secondary structures, which are involved in a diverse range of biological processes. Although the anti-cancer potential of G4s in oncogene promoters has been thoroughly investigated, the functions of promoter G4s in non-cancer-related genes are not well understood. We have explored the possible regulatory roles of promoter G4s in cardiac function-related genes using both computational and a wide range of experimental approaches. According to our bioinformatics results, it was found that potential G4-forming sequences are particularly enriched in the transcription regulatory regions (TRRs) of cardiac function-related genes. Subsequently, the promoter of human cardiac troponin I (TnIc) was chosen as a model, and G4s found in this region were subjected to biophysical characterisations. The chromosome 19 specific minisatellite G4 sequence (MNSG4) and near transcription start site (TSS) G4 sequence (−80 G4) adopt anti-parallel and parallel structures respectively in 100 mM KCl, with stabilities comparable to those of oncogene G4s. It was also found that TnIc G4s act cooperatively as enhancers in gene expression regulation in HEK293 cells, when stabilised by a synthetic G4-binding ligand. This study provides the first evidence of the biological significance of promoter G4s in cardiac function-related genes. The feasibility of using a single ligand to target multiple G4s in a particular gene has also been discussed.
Collapse
Affiliation(s)
- Wenhua Zhou
- Molecular Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Nigel J. Brand
- Harefield Heart Science Centre, National Heart and Lung Institute, Imperial College London, Middlesex, United Kingdom
| | - Paul J. R. Barton
- Harefield Heart Science Centre, National Heart and Lung Institute, Imperial College London, Middlesex, United Kingdom
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, United Kingdom
| | - Ramon Vilar
- Department of Chemistry, Imperial College London, London, United Kingdom
| | - Liming Ying
- Molecular Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
83
|
Mould A, Morgan MAJ, Li L, Bikoff EK, Robertson EJ. Blimp1/Prdm1 governs terminal differentiation of endovascular trophoblast giant cells and defines multipotent progenitors in the developing placenta. Genes Dev 2012; 26:2063-74. [PMID: 22987638 DOI: 10.1101/gad.199828.112] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Developmental arrest of Blimp1/Prdm1 mutant embryos at around embryonic day 10.5 (E10.5) has been attributed to placental disturbances. Here we investigate Blimp1/Prdm1 requirements in the trophoblast cell lineage. Loss of function disrupts specification of the invasive spiral artery-associated trophoblast giant cells (SpA-TGCs) surrounding maternal blood vessels and severely compromises the ability of the spongiotrophoblast layer to expand appropriately, secondarily causing collapse of the underlying labyrinth layer. Additionally, we identify a population of proliferating Blimp1(+) diploid cells present within the spongiotrophoblast layer. Lineage tracing experiments exploiting a novel Prdm1.Cre-LacZ allele demonstrate that these Blimp1(+) cells give rise to the mature SpA-TGCs, canal TGCs, and glycogen trophoblasts. In sum, the transcriptional repressor Blimp1/Prdm1 is required for terminal differentiation of SpA-TGCs and defines a lineage-restricted progenitor cell population contributing to placental growth and morphogenesis.
Collapse
Affiliation(s)
- Arne Mould
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | | | | | | | | |
Collapse
|
84
|
Takao T, Asanoma K, Tsunematsu R, Kato K, Wake N. The maternally expressed gene Tssc3 regulates the expression of MASH2 transcription factor in mouse trophoblast stem cells through the AKT-Sp1 signaling pathway. J Biol Chem 2012; 287:42685-94. [PMID: 23071113 PMCID: PMC3522269 DOI: 10.1074/jbc.m112.388777] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Tssc3 is a maternally expressed/paternally silenced imprinted gene. Recent evidence suggests that the loss of TSSC3 results in placental overgrowth in mice. These findings showed that the TSSC3 gene functions as a negative regulator of placental growth. In this study, we describe the function of TSSC3 and its signaling pathway in mouse trophoblast stem (TS) cell differentiation. First of all, we tested Tssc3 expression levels in TS cells. TS cells expressed Tssc3, and its expression level was the highest from day 1 to 4 but was down-regulated at day 5 after the induction of differentiation. Overexpression of TSSC3 in TS cells up-regulated Gcm1 and Mash2, which are marker genes of mouse trophoblast differentiation. Down-regulation of TSSC3 by siRNA enhanced Pl1 and Tpbpa expression in TS cells cultured under stem cell conditions, suggesting the contribution of TSSC3 to the differentiation from TS to trophoblast progenitors and/or labyrinth trophoblasts. TSSC3 activated the PI3K/AKT pathway through binding with phosphatidylinositol phosphate lipids and enhanced the activity of a promoter containing an E-box structure, which is the binding sequence of the Mash2 downstream target gene promoter. PI3K inhibitor suppressed the promoter activity induced by TSSC3. TSSC3 induced Sp1 translocation from cytoplasm to nucleus through the PI3K/AKT pathway. Nuclear Sp1 activated the Mash2 transcription by Sp1 binding with a consensus Sp1-binding motif. This is the first report describing that TSSC3 plays an important role in the differentiation from TS to trophoblast progenitors and/or labyrinth trophoblasts through the TSSC3/PI3K/AKT/MASH2 signaling pathway.
Collapse
Affiliation(s)
- Tomoka Takao
- Research Center for Environment and Developmental Medical Sciences, Graduate School of Medical Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | | | | | | | | |
Collapse
|
85
|
Human placentation from nidation to 5 weeks of gestation. Part I: What do we know about formative placental development following implantation? Placenta 2012; 33:327-34. [DOI: 10.1016/j.placenta.2012.01.020] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 01/19/2012] [Accepted: 01/30/2012] [Indexed: 11/19/2022]
|
86
|
Abstract
This review summarises current knowledge about the specification, commitment and maintenance of the trophoblast lineage in mice and cattle. Results from gene expression studies, in vivo loss-of-function models and in vitro systems using trophoblast and embryonic stem cells have been assimilated into a model seeking to explain trophoblast ontogeny via gene regulatory networks. While trophoblast differentiation is quite distinct between cattle and mice, as would be expected from their different modes of implantation, recent studies have demonstrated that differences arise much earlier during trophoblast development.
Collapse
|
87
|
Hu D, Cross JC. Ablation of Tpbpa-positive trophoblast precursors leads to defects in maternal spiral artery remodeling in the mouse placenta. Dev Biol 2011; 358:231-9. [DOI: 10.1016/j.ydbio.2011.07.036] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 07/24/2011] [Accepted: 07/27/2011] [Indexed: 11/16/2022]
|
88
|
Forkhead factor FoxO1 is essential for placental morphogenesis in the developing embryo. Proc Natl Acad Sci U S A 2011; 108:16307-12. [PMID: 21930913 DOI: 10.1073/pnas.1107341108] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Forkhead box O1 (FoxO1), a member of the Forkhead box-containing O family of transcription factors, is a key regulator of numerous genes that govern a wide array of cellular functions, including differentiation, homeostasis, and survival. However, the role of FoxO1 in development remains elusive. Here, we describe an essential and previously undefined role for FoxO1 in placental development. We demonstrate that FoxO1-null embryos up to embryonic day 9.0 (E9.0) are indistinguishable, including their morphology, cardiovascular structure, and vascular gene expression, from wild-type (WT) littermates. However, FoxO1-nulls manifested a profoundly swollen/hydropic allantois, which failed to fuse with the chorion, a phenotype that leads to subsequent cardiovascular malformation, progressive apoptotic cell death, and embryonic lethality at E10.5. Quantitative RT-PCR analysis of genes involved in placental development revealed significant attenuation of VCAM1 expression in FoxO1-null embryos. Using immunohistochemical, transcriptional, and chromatin immunoprecipitation assays, we further discovered that FoxO1 is an essential upstream regulator of the VCAM1 gene. Collectively, our findings provide critical molecular insight into a unique FoxO1-VCAM1 axis that governs placental morphogenesis, a process that is essential for subsequent normal cardiovascular development and fetal life.
Collapse
|
89
|
Garred MM, Wang MM, Guo X, Harrington CA, Lein PJ. Transcriptional responses of cultured rat sympathetic neurons during BMP-7-induced dendritic growth. PLoS One 2011; 6:e21754. [PMID: 21765909 PMCID: PMC3135585 DOI: 10.1371/journal.pone.0021754] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 06/06/2011] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Dendrites are the primary site of synapse formation in the vertebrate nervous system; however, relatively little is known about the molecular mechanisms that regulate the initial formation of primary dendrites. Embryonic rat sympathetic neurons cultured under defined conditions extend a single functional axon, but fail to form dendrites. Addition of bone morphogenetic proteins (BMPs) triggers these neurons to extend multiple dendrites without altering axonal growth or cell survival. We used this culture system to examine differential gene expression patterns in naïve vs. BMP-treated sympathetic neurons in order to identify candidate genes involved in regulation of primary dendritogenesis. METHODOLOGY/PRINCIPAL FINDINGS To determine the critical transcriptional window during BMP-induced dendritic growth, morphometric analysis of microtubule-associated protein (MAP-2)-immunopositive processes was used to quantify dendritic growth in cultures exposed to the transcription inhibitor actinomycin-D added at varying times after addition of BMP-7. BMP-7-induced dendritic growth was blocked when transcription was inhibited within the first 24 hr after adding exogenous BMP-7. Thus, total RNA was isolated from sympathetic neurons exposed to three different experimental conditions: (1) no BMP-7 treatment; (2) treatment with BMP-7 for 6 hr; and (3) treatment with BMP-7 for 24 hr. Affymetrix oligonucleotide microarrays were used to identify differential gene expression under these three culture conditions. BMP-7 significantly regulated 56 unique genes at 6 hr and 185 unique genes at 24 hr. Bioinformatic analyses implicate both established and novel genes and signaling pathways in primary dendritogenesis. CONCLUSIONS/SIGNIFICANCE This study provides a unique dataset that will be useful in generating testable hypotheses regarding transcriptional control of the initial stages of dendritic growth. Since BMPs selectively promote dendritic growth in central neurons as well, these findings may be generally applicable to dendritic growth in other neuronal cell types.
Collapse
Affiliation(s)
- Michelle M. Garred
- Gene Microarray Shared Resource, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Michael M. Wang
- Departments of Neurology and Molecular & Integrative Physiology, University of Michigan, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, United States of America
| | - Xin Guo
- Department of Environmental Health Sciences, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Christina A. Harrington
- Gene Microarray Shared Resource, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| |
Collapse
|
90
|
Marchand M, Horcajadas JA, Esteban FJ, McElroy SL, Fisher SJ, Giudice LC. Transcriptomic Signature of Trophoblast Differentiation in a Human Embryonic Stem Cell Model1. Biol Reprod 2011; 84:1258-71. [DOI: 10.1095/biolreprod.110.086413] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
91
|
Hoya-Arias R, Tomishima M, Perna F, Voza F, Nimer SD. L3MBTL1 deficiency directs the differentiation of human embryonic stem cells toward trophectoderm. Stem Cells Dev 2011; 20:1889-900. [PMID: 21341991 DOI: 10.1089/scd.2010.0437] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human embryonic stem cells (hESCs) can be used to study the early events in human development and, hopefully, to understand how to differentiate human pluripotent cells for clinical use. To define how L3MBTL1, a chromatin-associated polycomb group protein with transcriptional repressive activities, regulates early events in embryonic cell differentiation, we created hESC lines that constitutively express shRNAs directed against L3MBTL1. The L3MBTL1 knockdown (KD) hESCs maintained normal morphology, proliferation, cell cycle kinetics, cell surface markers, and karyotype after 40 passages. However, under conditions that promote spontaneous differentiation, the L3MBTL1 KD cells differentiated into a relatively homogeneous population of large, flat trophoblast-like cells, unlike the multilineage differentiation seen with the control cells. The differentiated L3MBTL1 KD cells expressed numerous trophoblast markers and secreted placental hormones. Although the L3MBTL1 KD cells could be induced to differentiate into various embryonic lineages, they adopted an exclusive trophoblast fate during spontaneous differentiation. Our data demonstrate that depletion of L3MBTL1 does not affect hESC self-renewal, rather it enhances differentiation toward extra-embryonic trophoblast tissues.
Collapse
Affiliation(s)
- Ruben Hoya-Arias
- Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | | | | | | | | |
Collapse
|
92
|
Zhou W, Brand NJ, Ying L. G-quadruplexes-novel mediators of gene function. J Cardiovasc Transl Res 2011; 4:256-70. [PMID: 21302011 DOI: 10.1007/s12265-011-9258-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Accepted: 01/24/2011] [Indexed: 11/30/2022]
Abstract
Since the famous double-helix model was proposed, chromosomal DNA has been regarded as a rigid molecule containing the genetic information of an organism. It is clear now that DNA can adopt many transient, complex structures that can perform different biological functions. The G4 DNA (also called DNA G-quadruplex or G-tetraplex), a four-stranded DNA structure composed of stacked G-tetrads (guanine tetrads), has attracted much attention during the past two decades due to its ability to adopt a variety of structures and its possible biological functions. This review gives a glimpse on the structural diversity and biophysical properties of these fascinating DNA structures. Common methods that are widely used in investigating biophysical properties and biological functions of G4 DNA are described briefly. Next, bioinformatics studies that indicate evidence of evolutionary selection and potential functions of G4 DNA are discussed. Finally, examples of various biological functions of different G4 DNA are given, and potential roles of G4 DNA in respect of cardiovascular science are discussed.
Collapse
Affiliation(s)
- Wenhua Zhou
- Molecular Medicine, National Heart and Lung Institute, Imperial College London, UK.
| | | | | |
Collapse
|
93
|
Shibata M, García-García MJ. The mouse KRAB zinc-finger protein CHATO is required in embryonic-derived tissues to control yolk sac and placenta morphogenesis. Dev Biol 2010; 349:331-41. [PMID: 21094155 DOI: 10.1016/j.ydbio.2010.11.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 11/07/2010] [Accepted: 11/10/2010] [Indexed: 11/18/2022]
Abstract
Yolk sac and placenta are required to sustain embryonic development in mammals, yet our understanding of the genes and processes that control morphogenesis of these extraembryonic tissues is still limited. The chato mutation disrupts ZFP568, a Krüppel-Associated-Box (KRAB) domain Zinc finger protein, and causes a unique set of extraembryonic malformations, including ruffling of the yolk sac membrane, defective extraembryonic mesoderm morphogenesis and vasculogenesis, failure to close the ectoplacental cavity, and incomplete placental development. Phenotypic analysis of chato embryos indicated that ZFP568 does not control proliferation or differentiation of extraembryonic lineages but rather regulates the morphogenetic events that shape extraembryonic tissues. Analysis of chimeric embryos showed that Zfp568 function is required in embryonic-derived lineages, including the extraembryonic mesoderm. Depleting Zfp568 affects the ability of extraembryonic mesoderm cells to migrate. However, explanted Zfp568 mutant cells could migrate properly when plated on appropriate extracellular matrix conditions. We show that expression of Fibronectin and Indian Hedgehog are reduced in chato mutant yolk sacs. These data suggest that ZFP568 controls the production of secreted factors required to promote morphogenesis of extraembryonic tissues. Our results support previously undescribed roles of the extraembryonic mesoderm in yolk sac morphogenesis and in the closure of the ectoplacental cavity and identify a novel role of ZFP568 in the development of extraembryonic tissues.
Collapse
Affiliation(s)
- Maho Shibata
- Department of Molecular Biology and Genetics, Cornell University, 259 Biotechnology Building, Ithaca, NY 14853, USA
| | | |
Collapse
|
94
|
Endocannabinoid signaling directs differentiation of trophoblast cell lineages and placentation. Proc Natl Acad Sci U S A 2010; 107:16887-92. [PMID: 20837524 DOI: 10.1073/pnas.1010892107] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In most mammals, placentation is critical for fetal development and pregnancy success. Exposure to marijuana during pregnancy has adverse effects, but whether the placenta is a target of cannabinoid/endocannabinoid signaling is not known. Using mice as a model system, we found that the endocannabinoid system is present in the ectoplacental cone and spongiotrophoblast cells. We also observed that aberrant endocannabinoid signaling confers premature trophoblast stem cell differentiation, and defective trophoblast development and invasion. These defects are reflected in retarded fetal development and compromised pregnancy outcome. Because the endocannabinoid system is conserved in mice and humans, our study suggests that endocannabinoid signaling is critical to placentation and pregnancy success in humans and implicates its potential significance in stem cell biology.
Collapse
|
95
|
Abstract
Mammalian androgenones have two paternally or sperm-derived genomes. In mice (Mus musculus) they die at peri-implantation due to the misexpression of imprinted genes-the genes that are expressed monoallelically according to the parent of origin. The misexpressions involved are poorly defined. To gain further insight, we examined the causes of midgestation death of embryos with paternal duplication (PatDp) of distal chromosome 7 (dist7), a region replete with imprinted genes. PatDp(dist7) embryos have a similar phenotype to mice with a knockout of a maternally expressed imprinted gene, Ascl2 [achaete-scute complex homolog-like 2 (Drosophila)], and their death at midgestation could result from two inactive paternal copies of this gene. However, other dist7 misexpressions could duplicate this phenotype, and the potential epistatic load is undefined. We show that an Ascl2 transgene is able to promote the development of PatDp(dist7) embryos to term, providing strong evidence that Ascl2 is the only imprinted gene in the genome for which PatDp results in early embryonic death. While some of the defects in perinatal transgenic PatDp(dist7) fetuses were consistent with known misexpressions of dist7 imprinted genes, the overall phenotype indicates a role for additional undefined misexpressions of imprinted genes. This study provides implications for the human imprinting-related fetal overgrowth disorder, Beckwith-Wiedemann syndrome.
Collapse
|
96
|
Pereira CF, Piccolo FM, Tsubouchi T, Sauer S, Ryan NK, Bruno L, Landeira D, Santos J, Banito A, Gil J, Koseki H, Merkenschlager M, Fisher AG. ESCs Require PRC2 to Direct the Successful Reprogramming of Differentiated Cells toward Pluripotency. Cell Stem Cell 2010; 6:547-56. [DOI: 10.1016/j.stem.2010.04.013] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Revised: 03/05/2010] [Accepted: 04/26/2010] [Indexed: 11/28/2022]
|
97
|
Maltepe E, Bakardjiev AI, Fisher SJ. The placenta: transcriptional, epigenetic, and physiological integration during development. J Clin Invest 2010; 120:1016-25. [PMID: 20364099 DOI: 10.1172/jci41211] [Citation(s) in RCA: 196] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The placenta provides critical transport functions between the maternal and fetal circulations during intrauterine development. Formation of this interface relies on coordinated interactions among transcriptional, epigenetic, and environmental factors. Here we describe these mechanisms in the context of the differentiation of placental cells (trophoblasts) and synthesize current knowledge about how they interact to generate a functional placenta. Developing an understanding of these pathways contributes to an improvement of our models for studying trophoblast biology and sheds light on the etiology of pregnancy complications and the in utero programming of adult diseases.
Collapse
Affiliation(s)
- Emin Maltepe
- Department of Pediatrics, Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, 513 Parnassus Ave. HSE-1401, Box 1346, San Francisco, CA 94143-1346, USA.
| | | | | |
Collapse
|
98
|
Stk40 links the pluripotency factor Oct4 to the Erk/MAPK pathway and controls extraembryonic endoderm differentiation. Proc Natl Acad Sci U S A 2010; 107:1402-7. [PMID: 20080709 DOI: 10.1073/pnas.0905657107] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Self-renewal and differentiation of embryonic stem cells (ESCs) are controlled by intracellular transcriptional factors and extracellular factor-activated signaling pathways. Transcription factor Oct4 is a key player maintaining ESCs in an undifferentiated state, whereas the Erk/MAPK pathway is known to be important for ESC differentiation. However, the manner in which intracellular pluripotency factors modulate extracellular factor-activated signaling pathways in ESCs is not well understood. Here, we report identification of a target gene of Oct4, serine/threonine kinase 40 (Stk40), which is able to activate the Erk/MAPK pathway and induce extraembryonic-endoderm (ExEn) differentiation in mouse ESCs. Interestingly, cells overexpressing Stk40 exclusively contribute to the ExEn layer of chimeric embryos when injected into host blastocysts. In contrast, deletion of Stk40 in ESCs markedly reduces ExEn differentiation in vitro. Mechanistically, Stk40 interacts with Rcn2, which also activates Erk1/2 to induce ExEn specification in mouse ESCs. Moreover, Rcn2 proteins are specifically located in the cytoplasm of the ExEn layer of early mouse embryos. Importantly, knockdown of Rcn2 blocks Stk40-activated Erk1/2 and ESC differentiation. Therefore, our study establishes a link between the pluripotency factor Oct4 and the Erk/MAPK signaling pathway, and it uncovers cooperating signals in the Erk/MAPK activation that control ExEn differentiation.
Collapse
|
99
|
|
100
|
El-Hashash AHK, Warburton D, Kimber SJ. Genes and signals regulating murine trophoblast cell development. Mech Dev 2009; 127:1-20. [PMID: 19755154 DOI: 10.1016/j.mod.2009.09.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2009] [Revised: 09/05/2009] [Accepted: 09/08/2009] [Indexed: 11/25/2022]
Abstract
A fundamental step in embryonic development is cell differentiation whereby highly specialised cell types are developed from a single undifferentiated, fertilised egg. One of the earliest lineages to form in the mammalian conceptus is the trophoblast, which contributes exclusively to the extraembryonic structures that form the placenta. Trophoblast giant cells (TGCs) in the rodent placenta form the outermost layer of the extraembryonic compartment, establish direct contact with maternal cells, and produce a number of pregnancy-specific cytokine hormones. Giant cells differentiate from proliferative trophoblasts as they exit the cell cycle and enter a genome-amplifying endocycle. Normal differentiation of secondary TGCs is a critical step toward the formation of the placenta and normal embryonic development. Trophoblast development is also of particular interest to the developmental biologist and immunobiologist, as these cells constitute the immediate cellular boundary between the embryonic and maternal tissues. Abnormalities in the development of secondary TGCs results in severe malfunction of the placenta. Herein we review new information that has been accumulated recently regarding the molecular and cellular regulation of trophoblast and placenta development. In particular, we discuss the molecular aspects of murine TGC differentiation. We also focus on the role of growth and transcription factors in TGC development.
Collapse
Affiliation(s)
- Ahmed H K El-Hashash
- Developmental Biology, Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine of University of Southern California, Los Angeles, CA 90027, USA
| | | | | |
Collapse
|