651
|
Crawford NG, Kelly DE, Hansen MEB, Beltrame MH, Fan S, Bowman SL, Jewett E, Ranciaro A, Thompson S, Lo Y, Pfeifer SP, Jensen JD, Campbell MC, Beggs W, Hormozdiari F, Mpoloka SW, Mokone GG, Nyambo T, Meskel DW, Belay G, Haut J, Rothschild H, Zon L, Zhou Y, Kovacs MA, Xu M, Zhang T, Bishop K, Sinclair J, Rivas C, Elliot E, Choi J, Li SA, Hicks B, Burgess S, Abnet C, Watkins-Chow DE, Oceana E, Song YS, Eskin E, Brown KM, Marks MS, Loftus SK, Pavan WJ, Yeager M, Chanock S, Tishkoff SA. Loci associated with skin pigmentation identified in African populations. Science 2017; 358:eaan8433. [PMID: 29025994 PMCID: PMC5759959 DOI: 10.1126/science.aan8433] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 10/03/2017] [Indexed: 12/13/2022]
Abstract
Despite the wide range of skin pigmentation in humans, little is known about its genetic basis in global populations. Examining ethnically diverse African genomes, we identify variants in or near SLC24A5, MFSD12, DDB1, TMEM138, OCA2, and HERC2 that are significantly associated with skin pigmentation. Genetic evidence indicates that the light pigmentation variant at SLC24A5 was introduced into East Africa by gene flow from non-Africans. At all other loci, variants associated with dark pigmentation in Africans are identical by descent in South Asian and Australo-Melanesian populations. Functional analyses indicate that MFSD12 encodes a lysosomal protein that affects melanogenesis in zebrafish and mice, and that mutations in melanocyte-specific regulatory regions near DDB1/TMEM138 correlate with expression of ultraviolet response genes under selection in Eurasians.
Collapse
Affiliation(s)
- Nicholas G Crawford
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Derek E Kelly
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Genomics and Computational Biology Graduate Program, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew E B Hansen
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marcia H Beltrame
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shaohua Fan
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shanna L Bowman
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ethan Jewett
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94704, USA
- Department of Statistics, University of California, Berkeley, Berkeley, CA 94704, USA
| | - Alessia Ranciaro
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Simon Thompson
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yancy Lo
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Susanne P Pfeifer
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Jeffrey D Jensen
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Michael C Campbell
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Biology, Howard University, Washington, DC 20059, USA
| | - William Beggs
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Farhad Hormozdiari
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA
| | | | - Gaonyadiwe George Mokone
- Department of Biomedical Sciences, University of Botswana School of Medicine, Gaborone, Botswana
| | - Thomas Nyambo
- Department of Biochemistry, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | | | - Gurja Belay
- Department of Biology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Jake Haut
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Harriet Rothschild
- Stem Cell Program, Division of Hematology and Oncology, Pediatric Hematology Program, Boston Children's Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Leonard Zon
- Stem Cell Program, Division of Hematology and Oncology, Pediatric Hematology Program, Boston Children's Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Yi Zhou
- Stem Cell Program, Division of Hematology and Oncology, Pediatric Hematology Program, Boston Children's Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Michael A Kovacs
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mai Xu
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tongwu Zhang
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kevin Bishop
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jason Sinclair
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cecilia Rivas
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eugene Elliot
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jiyeon Choi
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shengchao A Li
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD 20892, USA
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21701, USA
| | - Belynda Hicks
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD 20892, USA
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21701, USA
| | - Shawn Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christian Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD 20892, USA
| | - Dawn E Watkins-Chow
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elena Oceana
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, RI 02912, USA
| | - Yun S Song
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94704, USA
- Department of Statistics, University of California, Berkeley, Berkeley, CA 94704, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Mathematics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eleazar Eskin
- Department of Computer Science and Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kevin M Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael S Marks
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stacie K Loftus
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD 20892, USA
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21701, USA
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD 20892, USA
| | - Sarah A Tishkoff
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
652
|
Imam MU, Ismail M. The Impact of Traditional Food and Lifestyle Behavior on Epigenetic Burden of Chronic Disease. GLOBAL CHALLENGES (HOBOKEN, NJ) 2017; 1:1700043. [PMID: 31565292 PMCID: PMC6607231 DOI: 10.1002/gch2.201700043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/12/2017] [Indexed: 05/11/2023]
Abstract
Noncommunicable chronic diseases (NCCDs) are the leading causes of morbidity and mortality globally. The mismatch between present day diets and ancestral genome is suggested to contribute to the NCCDs burden, which is promoted by traditional risk factors like unhealthy diets, physical inactivity, alcohol and tobacco. However, epigenetic evidence now suggests that cumulatively inherited epigenetic modifications may have made humans more prone to the effects of present day lifestyle factors. Perinatal starvation was widespread in the 19th century. This together with more recent events like increasing consumption of western and low fiber diets, smoking, harmful use of alcohol, physical inactivity, and environmental pollutants may have programed the human epigenome for higher NCCDs risk. In this review, on the basis of available epigenetic data it is hypothesized that transgenerational effects of lifestyle factors may be contributing to the current global burden of NCCDs. Thus, there is a need to reconsider prevention strategies so that the subsequent generations will not have to pay for our sins and those of our ancestors.
Collapse
Affiliation(s)
- Mustapha U. Imam
- Precision Nutrition Innovation InstituteCollege of Public HealthZhengzhou UniversityZhengzhou450001China
| | - Maznah Ismail
- Laboratory of Molecular BiomedicineInstitute of BioscienceUniversiti Putra MalaysiaSerdangSelangor43400Malaysia
| |
Collapse
|
653
|
Greenfield A, Braude P, Flinter F, Lovell-Badge R, Ogilvie C, Perry ACF. Assisted reproductive technologies to prevent human mitochondrial disease transmission. Nat Biotechnol 2017; 35:1059-1068. [PMID: 29121011 DOI: 10.1038/nbt.3997] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/02/2017] [Indexed: 12/31/2022]
Abstract
Mitochondria are essential cytoplasmic organelles that generate energy (ATP) by oxidative phosphorylation and mediate key cellular processes such as apoptosis. They are maternally inherited and in humans contain a 16,569-base-pair circular genome (mtDNA) encoding 37 genes required for oxidative phosphorylation. Mutations in mtDNA cause a range of pathologies, commonly affecting energy-demanding tissues such as muscle and brain. Because mitochondrial diseases are incurable, attention has focused on limiting the inheritance of pathogenic mtDNA by mitochondrial replacement therapy (MRT). MRT aims to avoid pathogenic mtDNA transmission between generations by maternal spindle transfer, pronuclear transfer or polar body transfer: all involve the transfer of nuclear DNA from an egg or zygote containing defective mitochondria to a corresponding egg or zygote with normal mitochondria. Here we review recent developments in animal and human models of MRT and the underlying biology. These have led to potential clinical applications; we identify challenges to their technical refinement.
Collapse
Affiliation(s)
- Andy Greenfield
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Harwell, Oxfordshire, UK
| | - Peter Braude
- Division of Women's Health, King's College, London, UK
| | - Frances Flinter
- Clinical Genetics Department, Guy's Hospital, Great Maze Pond, London, UK
| | | | - Caroline Ogilvie
- Genetics Department, Guy's & St Thomas' NHS Foundation Trust and Division of Women's Health, King's College, London, UK
| | - Anthony C F Perry
- Laboratory of Mammalian Molecular Embryology, Department of Biology and Biochemistry, University of Bath, Bath, UK
| |
Collapse
|
654
|
Radzvilavicius AL, Lane N, Pomiankowski A. Sexual conflict explains the extraordinary diversity of mechanisms regulating mitochondrial inheritance. BMC Biol 2017; 15:94. [PMID: 29073898 PMCID: PMC5658935 DOI: 10.1186/s12915-017-0437-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/10/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Mitochondria are predominantly inherited from the maternal gamete, even in unicellular organisms. Yet an extraordinary array of mechanisms enforce uniparental inheritance, which implies shifting selection pressures and multiple origins. RESULTS We consider how this high turnover in mechanisms controlling uniparental inheritance arises using a novel evolutionary model in which control of mitochondrial transmission occurs either during spermatogenesis (by paternal nuclear genes) or at/after fertilization (by maternal nuclear genes). The model treats paternal leakage as an evolvable trait. Our evolutionary analysis shows that maternal control consistently favours strict uniparental inheritance with complete exclusion of sperm mitochondria, whereas some degree of paternal leakage of mitochondria is an expected outcome under paternal control. This difference arises because mito-nuclear linkage builds up with maternal control, allowing the greater variance created by asymmetric inheritance to boost the efficiency of purifying selection and bring benefits in the long term. In contrast, under paternal control, mito-nuclear linkage tends to be much weaker, giving greater advantage to the mixing of cytotypes, which improves mean fitness in the short term, even though it imposes a fitness cost to both mating types in the long term. CONCLUSIONS Sexual conflict is an inevitable outcome when there is competition between maternal and paternal control of mitochondrial inheritance. If evolution has led to complete uniparental inheritance through maternal control, it creates selective pressure on the paternal nucleus in favour of subversion through paternal leakage, and vice versa. This selective divergence provides a reason for the repeated evolution of novel mechanisms that regulate the transmission of paternal mitochondria, both in the fertilized egg and spermatogenesis. Our analysis suggests that the widespread occurrence of paternal leakage and prevalence of heteroplasmy are natural outcomes of this sexual conflict.
Collapse
Affiliation(s)
- Arunas L Radzvilavicius
- Centre for Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, Gower Street, London, WC1E 6BT, UK
- Department of Genetics, Evolution and Environment, University College London, Gower Street, London, WC1E 6BT, UK
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nick Lane
- Centre for Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, Gower Street, London, WC1E 6BT, UK
- Department of Genetics, Evolution and Environment, University College London, Gower Street, London, WC1E 6BT, UK
| | - Andrew Pomiankowski
- Centre for Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
- Department of Genetics, Evolution and Environment, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
655
|
Magnusson Å, Nilsson L, Oleröd G, Thurin-Kjellberg A, Bergh C. The addition of anti-Müllerian hormone in an algorithm for individualized hormone dosage did not improve the prediction of ovarian response-a randomized, controlled trial. Hum Reprod 2017; 32:811-819. [PMID: 28175316 DOI: 10.1093/humrep/dex012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/11/2017] [Indexed: 11/13/2022] Open
Abstract
Study question Does the addition of anti-Müllerian hormone (AMH) to a conventional dosage regimen, including age, antral follicle count (AFC) and BMI, improve the rate of targeted ovarian response, defined as 5-12 oocytes after IVF? Summary answer The addition of AMH did not alter the rate of targeted ovarian response, 5-12 oocytes, or decreased the rate of ovarian hyperstimulation syndrome (OHSS) or cancelled cycles due to poor ovarian response. What is known already Controlled ovarian hyperstimulation (COH) in connection with IVF is sometimes associated with poor ovarian response resulting in low pregnancy and live birth rates or leading to cycle cancellations, but also associated with excessive ovarian response, causing an increased risk of OHSS. Even though it is well-established that both AMH and AFC are strong predictors of ovarian response in IVF, few randomized trials have investigated their impact on achieving an optimal number of oocytes. Study design, size and duration Between January 2013 and May 2016, 308 patients starting their first IVF treatment were randomly assigned, using a computerized randomization program with concealed allocation of patients and in the proportions of 1:1, to one of two dosage algorithms for decisions on hormone starting dose, an algorithm, including AMH, AFC, age and BMI (intervention group), or an algorithm, including only AFC, age and BMI (control group). The study was blinded to patients and treating physicians. Participants/materials, setting, methods Women aged >18 and <40 years, with a BMI above 18.0 and below 35.0 kg/m2 starting their first IVF cycle where standard IVF was planned, were eligible. All patients were treated with a GnRH agonist protocol and recombinant FSH was used for stimulation. The study was performed as a single-centre study at a large IVF unit at a university hospital. Main results and the role of chance The rate of patients having the targeted number of oocytes retrieved was 81/152 (53.3%) in the intervention group versus 96/155 (61.9%) in the control group (P = 0.16, difference: -8.6, 95% CI: -20.3; 3.0). Cycles with poor response (<5 oocytes) were more frequent in the AMH group, 39/152 (25.7%) versus the non-AMH group, 17/155 (11.0%) (P < 0.01), while the number of cancelled cycles due to poor ovarian response did not differ 7/152 (4.6%) and 4/155 (2.6%) (P = 0.52). An excessive response (>12 oocytes) was seen in 32/152 (21.1%) and 42/155 (27.1%) patients, respectively (P = 0.27). Moderate or severe OHSS was observed among 5/152 (3.3%) and 6/155 (3.9%) patients, respectively (P = 1.0). Live birth rates were 48/152 (31.6%) and 42/155 (27.1%) per started cycle. Limitations, reasons for caution The categorization of AMH values in predicted low, normal and high responders was originally established using the Diagnostic Systems Laboratories assay and was translated to more recently released assays, lacking international standards and well-established reference intervals. The interpretation of AMH values between different assays should therefore be made with some caution. Wider implications of the findings An individualised dosage regimen including AMH compared with a non-AMH dosage regimen in an unselected patient population did not alter the number of women achieving the targeted number of oocytes, or the cancellation rate due to poor response or the occurrence of moderate/severe OHSS. However, this study cannot answer the question if using an algorithm for dose decision of FSH is superior to a standard dose and neither which ovarian reserve test is the most effective. Study funding/competing interest Financial support was received through Sahlgrenska University Hospital (ALFGBG-70 940) and unrestricted grants from Ferring Pharmaceuticals and the Hjalmar Svensson Research Foundation. None of the authors declares any conflict of interest. Trial registration The study was registered at www.clinicaltrials.gov NCT02013973. Trial registration date 6 December 2013. DATE OF FIRST PATIENT RANDOMIZED 14 January 2013.
Collapse
Affiliation(s)
- Å Magnusson
- Department of Obstetrics and Gynaecology, Institute of Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Reproductive Medicine, Sahlgrenska University Hospital, SE Gothenburg, Sweden
| | - L Nilsson
- Department of Obstetrics and Gynaecology, Institute of Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Reproductive Medicine, Sahlgrenska University Hospital, SE Gothenburg, Sweden
| | - G Oleröd
- Department of Clinical Chemistry, Sahlgrenska University Hospital, SE Gothenburg, Sweden
| | - A Thurin-Kjellberg
- Department of Obstetrics and Gynaecology, Institute of Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Reproductive Medicine, Sahlgrenska University Hospital, SE Gothenburg, Sweden
| | - C Bergh
- Department of Obstetrics and Gynaecology, Institute of Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Reproductive Medicine, Sahlgrenska University Hospital, SE Gothenburg, Sweden
| |
Collapse
|
656
|
Sampino S, Stankiewicz AM, Zacchini F, Goscik J, Szostak A, Swiergiel AH, Drago G, Modlinski JA, Ptak GE. Pregnancy at Advanced Maternal Age Affects Behavior and Hippocampal Gene Expression in Mouse Offspring. J Gerontol A Biol Sci Med Sci 2017; 72:1465-1473. [PMID: 28329103 PMCID: PMC5861961 DOI: 10.1093/gerona/glx016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 01/20/2017] [Indexed: 12/28/2022] Open
Abstract
There is growing evidence that advanced maternal age is a risk factor for neurological and neuropsychiatric disorders in offspring. However, it remains unclear whether the altered brain programming induced by advanced maternal age is mediated by pre- or postnatal factors. Here, a mouse model was used to investigate whether pregnancy at advanced age may provoke behavioral and brain gene expression changes in offspring. Swiss Albino mice conceived by 3-month-old males and either 15-18-month-old (n = 11) or 3-month-old control females (n = 5), were delivered by cesarean section, fostered after birth by 3-month-old dams and subjected to a battery of behavioral tests. Furthermore, genome-wide mRNA expression was analyzed in the hippocampi of 4-month-old males offspring using microarrays. Offspring conceived by old mothers exhibited increased ultrasound vocalization activity during separation from the foster mother, increased anxiety-like behaviors in adult life, and altered patterns of hippocampal gene expression, compared to controls. These effects were not reversed by the postnatal maternal care provided by the young foster mothers, suggesting that the altered brain programming is already established at birth, consistent with prenatal effects related to maternal aging.
Collapse
Affiliation(s)
- Silvestre Sampino
- Department of Experimental Embryology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, Poland
- Faculty of Veterinary Medicine, University of Teramo, Italy
| | - Adrian Mateusz Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of The Polish Academy of Sciences, Jastrzebiec, Poland
| | - Federica Zacchini
- Department of Experimental Embryology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, Poland
| | - Joanna Goscik
- Faculty of Computer Science, Bialystok University of Technology, Poland
| | - Agnieszka Szostak
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Breeding of The Polish Academy of Sciences, Jastrzebiec, Poland
| | - Artur Hugo Swiergiel
- Faculty of Biology, University of Gdansk, Poland
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center, Shreveport
| | - Gaspare Drago
- Laboratory of Clinical Epidemiology, Institute of Biomedicine and Molecular Immunology, National Research Center of Italy, Palermo
| | - Jacek Andrzej Modlinski
- Department of Experimental Embryology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, Poland
| | - Grazyna Ewa Ptak
- Department of Experimental Embryology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, Poland
- Faculty of Veterinary Medicine, University of Teramo, Italy
- Department of Animal Reproduction Biotechnology, National Research Institute of Animal Production, Balice, Poland
| |
Collapse
|
657
|
Sato K, Sato M. Multiple ways to prevent transmission of paternal mitochondrial DNA for maternal inheritance in animals. J Biochem 2017; 162:247-253. [PMID: 28981751 DOI: 10.1093/jb/mvx052] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 06/10/2017] [Indexed: 11/14/2022] Open
Abstract
Mitochondria contain their own DNA (mtDNA). In most sexually reproducing organisms, mtDNA is inherited maternally (uniparentally); this type of inheritance is thus referred to as 'maternal (uniparental) inheritance'. Recent studies have revealed various mechanisms to prevent the transmission of sperm-derived paternal mtDNA to the offspring, thereby ensuring maternal inheritance of mtDNA. In the nematode Caenorhabditis elegans, paternal mitochondria and their mtDNA degenerate almost immediately after fertilization and are selectively degraded by autophagy, which is referred to as 'allophagy' (allogeneic [non-self] organelle autophagy). In the fruit fly Drosophila melanogaster, paternal mtDNA is largely eliminated by an endonuclease G-mediated mechanism. Paternal mitochondria are subsequently removed by endocytic and autophagic pathways after fertilization. In many mammals, including humans, paternal mitochondria enter fertilized eggs. However, the fate of paternal mitochondria and their mtDNA in mammals is still a matter of debate. In this review, we will summarize recent knowledge on the molecular mechanisms underlying the prevention of paternal mtDNA transmission, which ensures maternal mtDNA inheritance in animals.
Collapse
Affiliation(s)
| | - Miyuki Sato
- Laboratory of Molecular Membrane Biology, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15, Showa-machi, Maebashi, Gunma 371-8512, Japan
| |
Collapse
|
658
|
Wang H, Airola MV, Reue K. How lipid droplets "TAG" along: Glycerolipid synthetic enzymes and lipid storage. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1131-1145. [PMID: 28642195 PMCID: PMC5688854 DOI: 10.1016/j.bbalip.2017.06.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/15/2017] [Accepted: 06/15/2017] [Indexed: 02/06/2023]
Abstract
Triacylglycerols (TAG) serve as the predominant form of energy storage in mammalian cells, and TAG synthesis influences conditions such as obesity, fatty liver, and insulin resistance. In most tissues, the glycerol 3-phosphate pathway enzymes are responsible for TAG synthesis, and the regulation and function of these enzymes is therefore important for metabolic homeostasis. Here we review the sites and regulation of glycerol-3-phosphate acyltransferase (GPAT), acylglycerol-3-phosphate acyltransferase (AGPAT), lipin phosphatidic acid phosphatase (PAP), and diacylglycerol acyltransferase (DGAT) enzyme action. We highlight the critical roles that these enzymes play in human health by reviewing Mendelian disorders that result from mutation in the corresponding genes. We also summarize the valuable insights that genetically engineered mouse models have provided into the cellular and physiological roles of GPATs, AGPATs, lipins and DGATs. Finally, we comment on the status and feasibility of therapeutic approaches to metabolic disease that target enzymes of the glycerol 3-phosphate pathway. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
Affiliation(s)
- Huan Wang
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Michael V Airola
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, United States
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; Molecular Biology Institute, University of California, Los Angeles, CA, United States.
| |
Collapse
|
659
|
Siu MT, Weksberg R. Epigenetics of Autism Spectrum Disorder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 978:63-90. [PMID: 28523541 DOI: 10.1007/978-3-319-53889-1_4] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorder (ASD), one of the most common childhood neurodevelopmental disorders (NDDs), is diagnosed in 1 of every 68 children. ASD is incredibly heterogeneous both clinically and aetiologically. The etiopathogenesis of ASD is known to be complex, including genetic, environmental and epigenetic factors. Normal epigenetic marks modifiable by both genetics and environmental exposures can result in epigenetic alterations that disrupt the regulation of gene expression, negatively impacting biological pathways important for brain development. In this chapter we aim to summarize some of the important literature that supports a role for epigenetics in the underlying molecular mechanism of ASD. We provide evidence from work in genetics, from environmental exposures and finally from more recent studies aimed at directly determining ASD-specific epigenetic patterns, focusing mainly on DNA methylation (DNAm). Finally, we briefly discuss some of the implications of current research on potential epigenetic targets for therapeutics and novel avenues for future work.
Collapse
Affiliation(s)
- Michelle T Siu
- Program in Genetics and Genome Biology, The Hospital for Sick Children, 555 University Ave, Toronto, ON, M5G 1X8, Canada
| | - Rosanna Weksberg
- Program in Genetics and Genome Biology, The Hospital for Sick Children, 555 University Ave, Toronto, ON, M5G 1X8, Canada. .,Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, 555 University Ave, Toronto, ON, M5G 1X8, Canada. .,Department of Paediatrics, University of Toronto, Toronto, ON, M5S 1A1, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
660
|
El Yakoubi W, Buffin E, Cladière D, Gryaznova Y, Berenguer I, Touati SA, Gómez R, Suja JA, van Deursen JM, Wassmann K. Mps1 kinase-dependent Sgo2 centromere localisation mediates cohesin protection in mouse oocyte meiosis I. Nat Commun 2017; 8:694. [PMID: 28947820 PMCID: PMC5612927 DOI: 10.1038/s41467-017-00774-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 07/27/2017] [Indexed: 01/10/2023] Open
Abstract
A key feature of meiosis is the step-wise removal of cohesin, the protein complex holding sister chromatids together, first from arms in meiosis I and then from the centromere region in meiosis II. Centromeric cohesin is protected by Sgo2 from Separase-mediated cleavage, in order to maintain sister chromatids together until their separation in meiosis II. Failures in step-wise cohesin removal result in aneuploid gametes, preventing the generation of healthy embryos. Here, we report that kinase activities of Bub1 and Mps1 are required for Sgo2 localisation to the centromere region. Mps1 inhibitor-treated oocytes are defective in centromeric cohesin protection, whereas oocytes devoid of Bub1 kinase activity, which cannot phosphorylate H2A at T121, are not perturbed in cohesin protection as long as Mps1 is functional. Mps1 and Bub1 kinase activities localise Sgo2 in meiosis I preferentially to the centromere and pericentromere respectively, indicating that Sgo2 at the centromere is required for protection.In meiosis I centromeric cohesin is protected by Sgo2 from Separase-mediated cleavage ensuring that sister chromatids are kept together until their separation in meiosis II. Here the authors demonstrate that Bub1 and Mps1 kinase activities are required for Sgo2 localisation to the centromere region.
Collapse
Affiliation(s)
- Warif El Yakoubi
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France
| | - Eulalie Buffin
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France
| | - Damien Cladière
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France
| | - Yulia Gryaznova
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France
| | - Inés Berenguer
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France
- Unidad de Biología Celular, Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Sandra A Touati
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France
- Chromosome Segregation Laboratory, Lincoln's Inn Fields Laboratory, The Francis Crick Institute, London, WC2A 3LY, UK
| | - Rocío Gómez
- Unidad de Biología Celular, Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - José A Suja
- Unidad de Biología Celular, Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Jan M van Deursen
- Department of Pediatric and Adolescent Medicine and Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Katja Wassmann
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France.
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France.
| |
Collapse
|
661
|
Li BT, Yu C, Xu Y, Liu SB, Fan HY, Pan WW. TET1 inhibits cell proliferation by inducing RASSF5 expression. Oncotarget 2017; 8:86395-86409. [PMID: 29156803 PMCID: PMC5689693 DOI: 10.18632/oncotarget.21189] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 08/23/2017] [Indexed: 02/01/2023] Open
Abstract
Tet methylcytosine dioxygenases (TETs) catalyze the oxidative reactions of 5-methylcytosine to 5-hydroxymethylcytosine (5hmC). However, TET1 roles in ovarian cancer cell growth are unknown. Here, we show that ectopic expression of TET1 increased 5hmC levels, and inhibited proliferation and colony formation in ovarian cancer cell lines. Furthermore, in vitro and in vivo functional studies demonstrated that TET1 overexpression is necessary for the suppression of ovarian cancer growth, whereas depletion of TET1 expression had the opposite effect. Furthermore, the results of RNA-seq and qRT-PCR analyses identified a tumor suppressor, Ras association domain family member 5 (RASSF5), as the key downstream target of TET1. TET1 promotes RASSF5 expression by demethylating a CpG site within RASSF5 promoter. Up-regulated RASSF5 expression leads to the suppression of ovarian cancer cells growth. Additionally, we demonstrated that inhibition of CUL4-DDB1 ubiquitin ligase complex decrease 5hmC levels in ovarian cancer cells. These results provide new insights into the understanding of how ovarian cancers develop and grow, and identify TET1 as a key player in this process.
Collapse
Affiliation(s)
- Bo-Tai Li
- Life Sciences Institute, Zhejiang University, Hangzhou 301158, China
| | - Chao Yu
- Life Sciences Institute, Zhejiang University, Hangzhou 301158, China
| | - Ying Xu
- Department of Cell Biology, College of Medicine, Jiaxing University, Jiaxing 314001, China
| | - Sheng-Bing Liu
- Department of Cell Biology, College of Medicine, Jiaxing University, Jiaxing 314001, China
| | - Heng-Yu Fan
- Life Sciences Institute, Zhejiang University, Hangzhou 301158, China
| | - Wei-Wei Pan
- Department of Cell Biology, College of Medicine, Jiaxing University, Jiaxing 314001, China
| |
Collapse
|
662
|
Frozen embryo transfer can be performed in the cycle immediately following the freeze-all cycle. J Assist Reprod Genet 2017; 35:135-142. [PMID: 28939988 DOI: 10.1007/s10815-017-1048-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 09/12/2017] [Indexed: 10/18/2022] Open
Abstract
PURPOSE In this study, we investigated whether the time interval between oocyte retrieval and frozen embryo transfer (FET) affected the live birth (LB) rates of human segmented-IVF cycles. METHOD A total of 1338 ICSI freeze-all cycles were performed between February 2015 and January 2016, with 1121 FET cycles being retrospectively analyzed. All vitrified-warmed blastocyst transfers were performed in artificial FET cycles, using gonadotropin-releasing hormone (GnRH) agonist downregulation and oral estrogen endometrial preparation. The primary outcome measure was LB. Cycles were investigated in oocyte retrieval-to-FET interval groups of 32-46, 47-61, 62-76, 77-91, and ≥ 92 days, with the 47-61-day group used as the reference group. RESULTS There were no significant differences in LB rates between the groups in the overall analysis, as well as, in sub-analyses investigating LB in terms of single blastocyst transfer (SBT), trigger type (GnRH agonist, triggers including hCG), oocyte number (≤ 5 and ≥ 15), and maternal age (> 35 years). CONCLUSION The present study showed that it is feasible to perform transfers 36 days after oocyte retrieval and that delaying FET in freeze-all beyond the cycle immediately following oocyte retrieval does not increase LB rates.
Collapse
|
663
|
Angelica sinensis Suppresses Body Weight Gain and Alters Expression of the FTO Gene in High-Fat-Diet Induced Obese Mice. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6280972. [PMID: 29098158 PMCID: PMC5632476 DOI: 10.1155/2017/6280972] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/12/2017] [Accepted: 07/24/2017] [Indexed: 01/15/2023]
Abstract
The root of Angelica sinensis (RAS) is a traditional Chinese medicine used for preventing and treating various diseases. In this study, we assessed RAS supplementation effects on body weight and the FTO gene expression and methylation status in a high-fat-diet (HFD) induced obese mouse model. Female obese mice were divided into groups according to RAS dosage in diet as follows: normal diet, HFD diet (HC), HFD with low-dosage RAS (DL), HFD with medium-dosage RAS (DM), and HFD with high-dosage RAS (DH). After RAS supplementation for 4 weeks, body weight suppression and FTO expression in DH mice were significantly higher than in HC mice, whereas no significant change in FTO expression was detected between DM and DL mice or in their offspring. Bisulfite sequencing PCR (BSP) revealed that the CpG island in the FTO promoter was hypermethylated up to 95.44% in the HC group, 91.67% in the DH group, and 90.00% in the normal diet group. Histological examination showed that adipocytes in the DH group were smaller than those in the HC group, indicating a potential role of RAS in obesity. This study indicated that RAS could ameliorate obesity induced by HFD and that the molecular mechanism might be associated with the expression of the FTO gene.
Collapse
|
664
|
Kšiňanová M, Čikoš Š, Babel’ová J, Šefčíková Z, Špirková A, Koppel J, Fabian D. The Responses of Mouse Preimplantation Embryos to Leptin In Vitro in a Transgenerational Model for Obesity. Front Endocrinol (Lausanne) 2017; 8:233. [PMID: 28959235 PMCID: PMC5604062 DOI: 10.3389/fendo.2017.00233] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/28/2017] [Indexed: 11/14/2022] Open
Abstract
The aim of the present study was to test the hypothesis that leptin can directly mediate the negative effect of maternal obesity on preimplantation embryos. As previously shown, maternal obesity retards early embryonic development in vivo and increases the incidence of apoptosis in blastocysts. When two-cell embryos isolated from control and obese mice were transferred to identical (leptin free) conditions in vitro, no differences in any growth or quality parameters were recorded, including apoptosis incidence in blastocysts. Embryos isolated from control mice responded to transfer to environments with a high concentration of leptin (10 ng/mL) with a significant increase in arrest at the first or subsequent cell cycle. However, the majority of non-arrested embryos developed into blastocysts, showing morphology comparable to those cultured in the leptin-free group. On the other hand, the exposure of embryos isolated from obese mice to high leptin concentration in vitro did not retard their development. Furthermore, these embryos developed into blastocysts, showing a lower incidence of apoptosis. In vivo-developed blastocysts recovered from obese mice showed elevated expression levels of the proapoptotic gene BAX and the insulin-responsive glucose transporter gene SLC2A4. In conclusion, elevated leptin levels have both positive and negative effects on preimplantation embryo development in vitro, a response that likely depends on the body condition of the embryo donor. Moreover, these results suggest that leptin acts as a survival factor rather than an apoptotic inductor in embryonic cells. Since no elevations in the expression of the leptin receptor gene (LEPR) or fat metabolism-associated genes (PLIN2, SLC27A4) were recorded in blastocysts recovered from obese mice, the role of leptin in mediating the effects of obesity on embryos at the peripheral level is likely lower than expected.
Collapse
Affiliation(s)
- Martina Kšiňanová
- Institute of Animal Physiology, Slovak Academy of Sciences, Košice, Slovakia
| | - Štefan Čikoš
- Institute of Animal Physiology, Slovak Academy of Sciences, Košice, Slovakia
| | - Janka Babel’ová
- Institute of Animal Physiology, Slovak Academy of Sciences, Košice, Slovakia
| | - Zuzana Šefčíková
- Institute of Animal Physiology, Slovak Academy of Sciences, Košice, Slovakia
| | - Alexandra Špirková
- Institute of Animal Physiology, Slovak Academy of Sciences, Košice, Slovakia
| | - Juraj Koppel
- Institute of Animal Physiology, Slovak Academy of Sciences, Košice, Slovakia
| | - Dušan Fabian
- Institute of Animal Physiology, Slovak Academy of Sciences, Košice, Slovakia
| |
Collapse
|
665
|
Chu N, Gui Y, Qiu X, Zhang N, Li L, Li D, Tang W, Gober HJ, Zhang B, Wang L. The effect of DHEA on apoptosis and cohesin levels in oocytes in aged mice. Biosci Trends 2017; 11:427-438. [PMID: 28717062 DOI: 10.5582/bst.2017.01108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Female fertility declines with age as the number of ovarian follicles decreases and aneuploidy increases. Degradation of the cohesin complex might be responsible for age-related aneuploidy. Dehydroepiandrosterone (DHEA) can improve the ovarian reserve and reduce the rate of aneuploidy, but the relationship between DHEA and cohesin levels in oocytes is still unknown. The aim of the current study was to evaluate the effect of the supplement DHEA on ovarian function, including the number of follicles and cohesin levels in oocytes. C57BL/6J mice at 3 weeks, 6 weeks, 12 weeks, 6 months, and 10 months of age were used to obtain a systematic view into follicle apoptosis and cohesin levels in oocytes. Nine-month-old C57BL/6J mice were administered saline (n = 5), 17β-estradiol (100 µg/kg per day, n = 5), or DHEA (5mg/Kg per day, n = 5). After 4 weeks, aged mice were weighed and sacrificed, and ovarian tissue samples were prepared. Anti-VASA staining and HE staining were used to count the number of follicles. Anti-γH2AX staining and TUNEL were used to measure follicle apoptosis and immunofluorescent staining was used to detect the levels of three oocyte cohesin subunits: REC8, SMC1β, and SMC3. Administration of the supplements 17β-estradiol and DHEA to aged mice increased the number of primordial and primary follicles and decreased the age-related apoptosis of follicles. Levels of the cohesin subunits REC8 and SMC1β declined with age, but DHEA and 17β-estradiol tended to delay that decline. The supplement DHEA increased the number of primordial and primary follicles in aged mice by inhibiting follicle apoptosis and tended to delay the decrease in cohesin levels in oocytes.
Collapse
Affiliation(s)
- Nan Chu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
| | - Yuyan Gui
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Xuemin Qiu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Na Zhang
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Lisha Li
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Dajin Li
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Wei Tang
- Department of Surgery, Graduate School of Medicine, The University of Tokyo
| | | | - Bin Zhang
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
| | - Ling Wang
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Fudan University Shanghai Medical College
- The Academy of Integrative Medicine of Fudan University
| |
Collapse
|
666
|
Zhou J, Wang B, Hu Y, Sun H. Association between the number of oocytes retrieved and cumulative live birth rate in women aged 35-40 years undergoing long GnRH agonist IVF/ICSI cycles. Arch Gynecol Obstet 2017; 296:1005-1012. [PMID: 28879481 DOI: 10.1007/s00404-017-4503-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/23/2017] [Indexed: 01/22/2023]
Abstract
PURPOSE To study the association between the numbers of oocytes retrieved and the cumulative live birth rates (LBR) in women aged 35-40 years undergoing long GnRH agonist IVF/ICSI cycles. METHODS A total of 931 women aged 35-40 years who underwent their first cycle of IVF/ICSI treatment between January 2010 and December 2013 at Nanjing Drum Tower Hospital were identified and reviewed. The main endpoint of this study was the cumulative LBR after one complete oocyte retrieval, which included fresh and all subsequent frozen-thaw embryo transfer cycles. Odds ratios (OR) and 95% confidence interval (CI) for live birth were estimated by multivariate logistic regression analysis. Furthermore, all the women were divided into four groups based on the number of oocytes retrieved: 0-4, 5-9, 10-14 or ≥15 oocytes group. Variables were then compared among groups. RESULTS We found that 634 out of the 931 patients (68.1%) achieved at least one live birth. The number of oocytes retrieved was an independent predictive factor for live birth, with OR 1.20 (95% CI 1.15-1.26) when adjusted for age (years), duration of infertility and Gn (gonadotrophin) doses. The cumulative LBR in the four different oocyte groups was 35.6, 68.8, 83.4 and 89.2%, respectively. When the 1-4 oocytes group was issued as a reference, the ORs for cumulative LBR gradually increased to 3.66, 6.74 and 11.77 in other three oocytes groups, respectively. The moderate-severe ovarian hyperstimulation syndrome (OHSS) rate was dramatically increased in the ≥15 oocytes group (6.9%) when compared to that in the 10-14 oocytes group (0.8%), while the cumulative LBR only increased 5.8% (from 83.4 to 89.2%). CONCLUSIONS The ideal number of oocytes retrieved in women aged 35-40 years is 10-14 oocytes, which achieves a high cumulative LBR while maintaining an acceptable low OHSS rate.
Collapse
Affiliation(s)
- Jianjun Zhou
- Centre for Reproductive Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan North Road 53, Nanjing, 210008, China
| | - Bin Wang
- Centre for Reproductive Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan North Road 53, Nanjing, 210008, China
| | - Yali Hu
- Centre for Reproductive Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan North Road 53, Nanjing, 210008, China
| | - Haixiang Sun
- Centre for Reproductive Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan North Road 53, Nanjing, 210008, China.
| |
Collapse
|
667
|
Varghese AC, Tan G, Chan P, Tan SL. Clinical usefulness of sperm DNA fragmentation testing. Transl Androl Urol 2017; 6:S484-S487. [PMID: 29082166 PMCID: PMC5643608 DOI: 10.21037/tau.2017.06.12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
| | - Grace Tan
- OriginElle Fertility Clinic and Women's Health Centre, Montreal, QC, Canada.,Department of Epidemiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Peter Chan
- OriginElle Fertility Clinic and Women's Health Centre, Montreal, QC, Canada.,Division of Urology, Department of Surgery, McGill University, Montreal, QC, Canada
| | - Seang Lin Tan
- OriginElle Fertility Clinic and Women's Health Centre, Montreal, QC, Canada.,Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
| |
Collapse
|
668
|
Ruggieri M, Praticò AD, Serra A, Maiolino L, Cocuzza S, Di Mauro P, Licciardello L, Milone P, Privitera G, Belfiore G, Di Pietro M, Di Raimondo F, Romano A, Chiarenza A, Muglia M, Polizzi A, Evans DG. Childhood neurofibromatosis type 2 (NF2) and related disorders: from bench to bedside and biologically targeted therapies. ACTA OTORHINOLARYNGOLOGICA ITALICA 2017; 36:345-367. [PMID: 27958595 PMCID: PMC5225790 DOI: 10.14639/0392-100x-1093] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 04/29/2016] [Indexed: 12/22/2022]
Abstract
Neurofibromatosis type 2 [NF2; MIM # 101000] is an autosomal dominant disorder characterised by the occurrence of vestibular schwannomas (VSs), schwannomas of other cranial, spinal and cutaneous nerves, cranial and spinal meningiomas and/or other central nervous system (CNS) tumours (e.g., ependymomas, astrocytomas). Additional features include early onset cataracts, optic nerve sheath meningiomas, retinal hamartomas, dermal schwannomas (i.e., NF2-plaques), and (few) café-au-lait spots. Clinically, NF2 children fall into two main groups: (1) congenital NF2 - with bilateral VSs detected as early as the first days to months of life, which can be stable/asymptomatic for one-two decades and suddenly progress; and (2) severe pre-pubertal (Wishart type) NF2- with multiple (and rapidly progressive) CNS tumours other-than-VS, which usually present first, years before VSs [vs. the classical adult (Gardner type) NF2, with bilateral VSs presenting in young adulthood, sometimes as the only disease feature]. Some individuals can develop unilateral VS associated with ipsilateral meningiomas or multiple schwannomas localised to one part of the peripheral nervous system [i.e., mosaic NF2] or multiple non-VS, non-intradermal cranial, spinal and peripheral schwannomas (histologically proven) [schwannomatosis]. NF2 is caused by mutations in the NF2 gene at chromosome 22q12.1, which encodes for a protein called merlin or schwannomin, most similar to the exrin-readixin-moesin (ERM) proteins; mosaicNF2 is due to mosaic phenomena for the NF2 gene, whilst schwannomatosis is caused by coupled germ-line and mosaic mutations either in the SMARCB1 gene [SWNTS1; MIM # 162091] or the LZTR1 gene [SWNTS2; MIM # 615670] both falling within the 22q region and the NF2 gene. Data driven from in vitro and animal studies on the merlin pathway [e.g., post-translational and upstream/downstream regulation] allowed biologically targeted treatment strategies [e.g., Lapatinib, Erlotinib, Bevacizumab] aimed to multiple tumour shrinkage and/or regression and tumour arrest of progression with functional improvement.
Collapse
Affiliation(s)
- M Ruggieri
- Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Italy
| | - A D Praticò
- Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Italy.,Department of Biomedical and Biotechnological Sciences, University of Catania, Italy
| | - A Serra
- Department of Medical and Surgical Sciences and Advanced Technologies "G. Ingrassia", Institute of Otorhinolaryngology, University of Catania, Italy
| | - L Maiolino
- Department of Medical and Surgical Sciences and Advanced Technologies "G. Ingrassia", Institute of Otorhinolaryngology, University of Catania, Italy
| | - S Cocuzza
- Department of Medical and Surgical Sciences and Advanced Technologies "G. Ingrassia", Institute of Otorhinolaryngology, University of Catania, Italy
| | - P Di Mauro
- Department of Medical and Surgical Sciences and Advanced Technologies "G. Ingrassia", Institute of Otorhinolaryngology, University of Catania, Italy
| | - L Licciardello
- Department of Medical and Surgical Sciences and Advanced Technologies "G. Ingrassia", Institute of Otorhinolaryngology, University of Catania, Italy
| | - P Milone
- Department of Medical and Surgical Sciences and Advanced Technologies "G. Ingrassia", Institute of Radiology, University of Catania, Italy
| | - G Privitera
- Department of Medical and Surgical Sciences and Advanced Technologies "G. Ingrassia", Institute of Radiology, University of Catania, Italy
| | - G Belfiore
- Unit of Paediatric Radiology, AOU "Policlinico-Vittorio Emanuele", Catania, Italy
| | - M Di Pietro
- Department of Medical and Surgical Sciences and Advanced Technologies "G. Ingrassia", Institute of Ophthalmology, University of Catania, Italy
| | - F Di Raimondo
- Division of Hematology, AOU "Policlinico-Vittorio Emanuele", University of Catania, Italy
| | - A Romano
- Division of Hematology, AOU "Policlinico-Vittorio Emanuele", University of Catania, Italy
| | - A Chiarenza
- Division of Hematology, AOU "Policlinico-Vittorio Emanuele", University of Catania, Italy
| | - M Muglia
- Unit of Genetics, Institute of Neurological Sciences, National Research Council, Piano Lago di Mangone, Italy
| | - A Polizzi
- National Centre for Rare Disease, Istituto Superiore di Sanità, Rome, Italy.,Institute of Neurological Sciences, National Research Council, Catania, Italy
| | - D G Evans
- Genomic Medicine, University of Manchester, Manchester Academic Health Science Centre, Institute of Human Development, Central Manchester NHS Foundation Trust, Manchester Royal Infirmary, Manchester, UK
| |
Collapse
|
669
|
Haas J, Bassil R, Meriano J, Samara N, Barzilay E, Gonen N, Casper RF. Does daily co-administration of letrozole and gonadotropins during ovarian stimulation improve IVF outcome? Reprod Biol Endocrinol 2017; 15:70. [PMID: 28854933 PMCID: PMC5577762 DOI: 10.1186/s12958-017-0288-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 08/24/2017] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND For the last year we have been treating normal responders with gonadotropins and letrozole during the whole stimulation in order to improve response to FSH by increasing the intrafollicular androgen concentration, and to reduce circulating estrogen concentrations. The aim of this study was to compare the IVF outcome of normal responders treated with letrozole and gonadotropins during ovarian stimulation with patients treated with gonadotropins only. METHODS A single centre retrospective cohort study of 174 patients (87 in each group). RESULTS The age of the patients was comparable between the groups. Estradiol levels were significantly higher in the control group (6760 pmol/L vs. 2420 pmol/L respectively, p < 0.01), and the number of follicles ≥15 mm at the trigger day was significantly lower in the control group (7.9 vs. 10, p = 0.02). The number of retrieved oocytes (10 vs. 14.5, p < 0.01), MII oocytes (7.9 vs. 11.2, p < 0.01) and blastocysts (2.7 vs. 4.0, p = 0.02) was significantly higher in the study group. We found no significant differences in the cumulative pregnancy outcome between the two groups (65.2% vs 58.3% p = NS). CONCLUSIONS We conclude that co-treatment with letrozole improves the IVF outcome in normal responders in terms of increased number of blastocysts obtained without increasing the pregnancy rate or the risk of OHSS.
Collapse
Affiliation(s)
- Jigal Haas
- 0000 0001 2157 2938grid.17063.33Division of Reproductive Sciences, University of Toronto, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada
- TRIO fertility partners, 655 Bay St, Toronto, ON M5G 2K4 Canada
- 0000 0004 1937 0546grid.12136.37Chaim Sheba medical Center, Tel Hashomer, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rawad Bassil
- 0000 0001 2157 2938grid.17063.33Division of Reproductive Sciences, University of Toronto, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada
- TRIO fertility partners, 655 Bay St, Toronto, ON M5G 2K4 Canada
| | - Jim Meriano
- 0000 0001 2157 2938grid.17063.33Division of Reproductive Sciences, University of Toronto, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada
- TRIO fertility partners, 655 Bay St, Toronto, ON M5G 2K4 Canada
| | - Nivin Samara
- 0000 0001 2157 2938grid.17063.33Division of Reproductive Sciences, University of Toronto, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada
- TRIO fertility partners, 655 Bay St, Toronto, ON M5G 2K4 Canada
| | - Eran Barzilay
- 0000 0001 2157 2938grid.17063.33Division of Reproductive Sciences, University of Toronto, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada
- TRIO fertility partners, 655 Bay St, Toronto, ON M5G 2K4 Canada
- 0000 0004 1937 0546grid.12136.37Chaim Sheba medical Center, Tel Hashomer, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Noa Gonen
- 0000 0001 2157 2938grid.17063.33Division of Reproductive Sciences, University of Toronto, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada
- TRIO fertility partners, 655 Bay St, Toronto, ON M5G 2K4 Canada
| | - Robert F. Casper
- 0000 0001 2157 2938grid.17063.33Division of Reproductive Sciences, University of Toronto, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada
- TRIO fertility partners, 655 Bay St, Toronto, ON M5G 2K4 Canada
| |
Collapse
|
670
|
Miyamoto T, Iijima M, Shin T, Minase G, Ueda H, Okada H, Sengoku K. CUL4B mutations are uncommon in Japanese patients with Sertoli-cell-only syndrome and azoospermia. J OBSTET GYNAECOL 2017; 38:293-294. [PMID: 28816568 DOI: 10.1080/01443615.2017.1336755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- T Miyamoto
- a Department of Obstetrics and Gynecology , Asahikawa Medical University , Asahikawa , Japan
| | - M Iijima
- b Department of Urology , Kanazawa University Graduate School of Medical Science , Kanazawa , Japan
| | - T Shin
- c Department of Urology , Dokkyo Medical University Koshigaya Hospital , Koshigaya , Japan
| | - G Minase
- a Department of Obstetrics and Gynecology , Asahikawa Medical University , Asahikawa , Japan
| | - H Ueda
- a Department of Obstetrics and Gynecology , Asahikawa Medical University , Asahikawa , Japan
| | - H Okada
- c Department of Urology , Dokkyo Medical University Koshigaya Hospital , Koshigaya , Japan
| | - K Sengoku
- a Department of Obstetrics and Gynecology , Asahikawa Medical University , Asahikawa , Japan
| |
Collapse
|
671
|
Krishnan S, Smits AH, Vermeulen M, Reinberg D. Phospho-H1 Decorates the Inter-chromatid Axis and Is Evicted along with Shugoshin by SET during Mitosis. Mol Cell 2017; 67:579-593.e6. [PMID: 28781233 PMCID: PMC5562512 DOI: 10.1016/j.molcel.2017.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/26/2017] [Accepted: 07/06/2017] [Indexed: 12/22/2022]
Abstract
Precise control of sister chromatid separation during mitosis is pivotal to maintaining genomic integrity. Yet, the regulatory mechanisms involved are not well understood. Remarkably, we discovered that linker histone H1 phosphorylated at S/T18 decorated the inter-chromatid axial DNA on mitotic chromosomes. Sister chromatid resolution during mitosis required the eviction of such H1S/T18ph by the chaperone SET, with this process being independent of and most likely downstream of arm-cohesin dissociation. SET also directed the disassembly of Shugoshins in a polo-like kinase 1-augmented manner, aiding centromere resolution. SET ablation compromised mitotic fidelity as evidenced by unresolved sister chromatids with marked accumulation of H1S/T18ph and centromeric Shugoshin. Thus, chaperone-assisted eviction of linker histones and Shugoshins is a fundamental step in mammalian mitotic progression. Our findings also elucidate the functional implications of the decades-old observation of mitotic linker histone phosphorylation, serving as a paradigm to explore the role of linker histones in bio-signaling processes.
Collapse
Affiliation(s)
- Swathi Krishnan
- Howard Hughes Medical Institute, New York University Langone School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Langone School of Medicine, New York, NY 10016, USA
| | - Arne H Smits
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, 6525 GA Nijmegen, the Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, 6525 GA Nijmegen, the Netherlands
| | - Danny Reinberg
- Howard Hughes Medical Institute, New York University Langone School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Langone School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
672
|
Fornes R, Maliqueo M, Hu M, Hadi L, Jimenez-Andrade JM, Ebefors K, Nyström J, Labrie F, Jansson T, Benrick A, Stener-Victorin E. The effect of androgen excess on maternal metabolism, placental function and fetal growth in obese dams. Sci Rep 2017; 7:8066. [PMID: 28808352 PMCID: PMC5556034 DOI: 10.1038/s41598-017-08559-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/11/2017] [Indexed: 12/11/2022] Open
Abstract
Pregnant women with polycystic ovary syndrome (PCOS) are often overweight or obese. To study the effects of maternal androgen excess in obese dams on metabolism, placental function and fetal growth, female C57Bl6J mice were fed a control (CD) or a high fat/high sucrose (HF/HS) diet for 4-10 weeks, and then mated. On gestational day (GD) 15.5-17.5, dams were injected with dihydrotestosterone (CD-DHT, HF/HS-DHT) or a vehicle (CD-Veh, HF/HS-Veh). HF/HS dams had higher fat content, both before mating and on GD18.5, with no difference in glucose homeostasis, whereas the insulin sensitivity was higher in DHT-exposed dams. Compared to the CD groups, the livers from HF/HS dams weighed more on GD18.5, the triglyceride content was higher, and there was a dysregulation of liver enzymes related to lipogenesis and higher mRNA expression of Fitm1. Fetuses from HF/HS-Veh dams had lower liver triglyceride content and mRNA expression of Srebf1c. Maternal DHT exposure, regardless of diet, decreased fetal liver Pparg mRNA expression and increased placental androgen receptor protein expression. Maternal diet-induced obesity, together with androgen excess, affects maternal and fetal liver function as demonstrated by increased triglyceride content and dysfunctional expression of enzymes and transcription factors involved in de novo lipogenesis and fat storage.
Collapse
Affiliation(s)
- Romina Fornes
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Manuel Maliqueo
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77, Stockholm, Sweden
- Endocrinology and Metabolism Laboratory, Department of Medicine, West Division, University of Chile, Santiago, Chile
| | - Min Hu
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Laila Hadi
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Juan M Jimenez-Andrade
- Unidad Académica Multidisciplinaria Reynosa Aztlán, Universidad Autónoma de Tamaulipas, Reynosa, Tamaulipas, Mexico
| | - Kerstin Ebefors
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jenny Nyström
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fernand Labrie
- Laval University Research Center in Molecular Endocrinology, Oncology and Human Genomics, CHUL Research Center, Quebec, G1V 4G2, Canada
| | - Thomas Jansson
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| | - Anna Benrick
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- School of Health and Education, University of Skövde, 54128, Skovde, Sweden
| | | |
Collapse
|
673
|
Dalgaard JZ. What is the underlying cause of type II diabetes? – Are cells protecting themselves against the reactivity of glucose? Med Hypotheses 2017; 105:22-24. [DOI: 10.1016/j.mehy.2017.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/23/2017] [Accepted: 06/22/2017] [Indexed: 12/27/2022]
|
674
|
Bowman RL, Levine RL. TET2 in Normal and Malignant Hematopoiesis. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a026518. [PMID: 28242787 DOI: 10.1101/cshperspect.a026518] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The ten-eleven translocation (TET) family of enzymes were originally cloned from the translocation breakpoint of t(10;11) in infant acute myeloid leukemia (AML) with subsequent genomic analyses revealing somatic mutations and suppressed expression of TET family members across a range of malignancies, particularly enriched in hematological neoplasms. The TET family of enzymes is responsible for the hydroxylation of 5-methylcytosines (5-mC) to 5-hydroxymethylcytosine (5-hmC), followed by active and passive mechanisms leading to DNA demethylation. Given the complexity and importance of DNA methylation events in cellular proliferation and differentiation, it comes as no surprise that the TET family of enzymes is intricately regulated by both small molecules and regulatory cooperating proteins. Here, we review the structure and function of TET2, its interactions with cooperating mutations and small molecules, and its role in aberrant hematopoiesis.
Collapse
Affiliation(s)
- Robert L Bowman
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10021
| | - Ross L Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10021.,Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021
| |
Collapse
|
675
|
Contreras ZA, Hansen J, Ritz B, Olsen J, Yu F, Heck JE. Parental age and childhood cancer risk: A Danish population-based registry study. Cancer Epidemiol 2017; 49:202-215. [PMID: 28715709 DOI: 10.1016/j.canep.2017.06.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 06/27/2017] [Accepted: 06/30/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Though the association between parental age at child's birth and the risk of childhood cancer has been previously investigated, the evidence to date is inconclusive and scarce for rarer cancer types. METHODS Cancer cases (N=5,856) were selected from all children born from 1968 to 2014 and diagnosed from 1968 to 2015 in Denmark at less than 16 years of age listed in the nationwide Danish Cancer Registry. Cases were individually matched to controls (1:100) on sex and year of birth with a total of 585,594 controls randomly sampled from all live births in Denmark from the Danish Central Population Registry. Parental age at child's birth was extracted from the Central Population Registry. Conditional logistic regression models were used to estimate odds ratios for the association between parental age at child's birth and childhood cancer risk. Parental age was modeled as both categorical (referent group, parents aged 25-29) and continuous per 5-year increase in age. RESULTS Offspring of older mothers were at an increased risk of acute lymphoblastic leukemia [OR=1.10, 95% CI: (1.02, 1.19) per 5-year increase in age]. Older maternal age (40+) increased the risk of non-Hodgkin lymphoma [OR=1.96, 95%CI: (1.12, 3.43)]. The risk of Wilms' tumor also appeared elevated with older paternal age [OR=1.11, 95% CI: (0.97, 1.28) per 5-year increment in age]. CONCLUSION Older parental age was a risk factor for various childhood cancers in Danish children. Further investigation of the biological and social factors that may be contributing to these associations is warranted.
Collapse
Affiliation(s)
- Zuelma A Contreras
- Department of Epidemiology, School of Public Health, University of California, Los Angeles, CA, USA
| | - Johnni Hansen
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Beate Ritz
- Department of Epidemiology, School of Public Health, University of California, Los Angeles, CA, USA
| | - Jorn Olsen
- Department of Clinical Epidemiology, Aarhus University, Denmark
| | - Fei Yu
- Department of Biostatistics, School of Public Health, University of California, Los Angeles, CA, USA
| | - Julia E Heck
- Department of Epidemiology, School of Public Health, University of California, Los Angeles, CA, USA.
| |
Collapse
|
676
|
Morphogenesis-related gene-expression profile in porcine oocytes before and after in vitro maturation. ZYGOTE 2017; 25:331-340. [PMID: 28669375 DOI: 10.1017/s096719941700020x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mammalian oocyte maturation is achieved when oocytes reach metaphase II (MII) stage, and accumulate mRNA and proteins in the cytoplasm following fertilization. It has been shown that oocytes investigated before and after in vitro maturation (IVM) differ significantly in transcriptomic and proteomic profiles. Additionally, folliculogenesis and oogenesis is accompanied by morphogenetic changes, which significantly influence further zygote formation and embryo growth. This study aimed to determine new transcriptomic markers of porcine oocyte morphogenesis that are associated with cell maturation competence. An Affymetrix microarray assay was performed on an RNA template isolated from porcine oocytes before (n = 150) and after (n = 150) IVM. The brilliant cresyl blue (BCB) staining test was used for identification of cells with the highest developmental capacity. DAVID (Database for Annotation, Visualization, and Integrated Discovery) software was used for the extraction of the genes belonging to a cell morphogenesis Gene Ontology group. The control group consisted of freshly isolated oocytes. In total, 12,000 different transcripts were analysed, from which 379 genes were downregulated and 40 were upregulated in oocytes following IVM. We found five genes, SOX9, MAP1B, DAB2, FN1, and CXCL12, that were significantly upregulated in oocytes after IVM (in vitro group) compared with oocytes analysed before IVM (in vivo group). In conclusion, we found new transcriptomic markers of oocyte morphogenesis, which may be also recognized as significant mediators of cellular maturation capacity in pigs. Genes SOX9, MAP1B, DAB2, FN1, and CXCL12 may be involved in the regulation of the MII stage oocyte formation and several other processes that are crucial for porcine reproductive competence.
Collapse
|
677
|
Wu G, Lei L, Schöler HR. Totipotency in the mouse. J Mol Med (Berl) 2017; 95:687-694. [PMID: 28102431 PMCID: PMC5487595 DOI: 10.1007/s00109-017-1509-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/20/2016] [Accepted: 01/12/2017] [Indexed: 12/31/2022]
Abstract
In mammals, the unicellular zygote starts the process of embryogenesis and differentiates into all types of somatic cells, including both fetal and extraembryonic lineages-in a highly organized manner to eventually give rise to an entire multicellular organism comprising more than 200 different tissue types. This feature is referred to as totipotency. Upon fertilization, oocyte maternal factors epigenetically reprogram the genomes of the terminally differentiated oocyte and spermatozoon and turn the zygote into a totipotent cell. Today, we still do not fully understand the molecular properties of totipotency. In this review, we discuss recent findings on the molecular signature and mechanism of transcriptional regulation networks in the totipotent mouse embryo.
Collapse
Affiliation(s)
- Guangming Wu
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
| | - Lei Lei
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Department of Histology and Embryology, Harbin Medical University, 194 Xuefu Road, Nangang District, Harbin, 150081, China
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany.
- Medical Faculty, University of Münster, Domagkstr. 3, 48149, Münster, Germany.
| |
Collapse
|
678
|
Zhai J, Liu J, Cheng X, Li S, Hong Y, Sun K, Chen ZJ, Du Y, Li W. Zinc finger gene 217 (ZNF217) Promoted Ovarian Hyperstimulation Syndrome (OHSS) through Regulating E 2 Synthesis and Inhibiting Thrombospondin-1 (TSP-1). Sci Rep 2017; 7:3245. [PMID: 28607476 PMCID: PMC5468349 DOI: 10.1038/s41598-017-03555-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/02/2017] [Indexed: 01/24/2023] Open
Abstract
Zinc finger gene 217 (ZNF217) is a candidate gene of polycystic ovary syndrome (PCOS) which is vulnerable to ovarian hyperstimulation syndrome (OHSS). However, the relationship between ZNF217 and OHSS is largely unknown. Our study demonstrated that ZNF217 was mainly distributed in the granulosa cells of rat ovary. Significantly higher expression of ovarian ZNF217 was detected in OHSS rats, being consistent with serum 17β-estradiol concentration and ovarian aromatase. Moreover, OHSS rats also showed decreased ovarian TSP-1 mRNA, an acknowledged VEGF signaling suppressor. The same changes were detected in human granulosa cells and follicular fluid. Thus, the increased ZNF217 and decreased TSP-1 may participate in OHSS onset. In vitro experiment revealed that ZNF217 positively regulated E2 synthesis through promoting cAMP response element binding protein (CREB) and thereby CYP19A1 in KGN cells. Furthermore, ZNF217 negatively regulated TSP-1 in KGN cells while TSP-1 promoted claudin1 and inhibited nitric oxide (NO) in HUVECs and HAECs. Both of claudin1 and NO are responsible for the regulation of vascular permeability (VP). Therefore, we demonstrated that ZNF217 contributed to OHSS onset through promoting E2 synthesis and the increase of VP. Moreover, the increased ZNF217 and decreased TSP-1 provided new targets for the prevention or treatment of OHSS in the future.
Collapse
Affiliation(s)
- Junyu Zhai
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Jiansheng Liu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Xiaoyue Cheng
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Shang Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Yan Hong
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory for Reproductive Endocrinology of Ministry of Education, Shandong Provincial Key Laboratory of Reproductive Medicine, Center for Reproductive Medicine, Shandong Provincial Hospital, Shandong University, Jinan, 250021, China
| | - Yanzhi Du
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China. .,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China.
| | - Weiping Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China. .,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China.
| |
Collapse
|
679
|
Li Q, Cui M, Yang F, Li N, Jiang B, Yu Z, Zhang D, Wang Y, Zhu X, Hu H, Li PS, Ning SL, Wang S, Qi H, Song H, He D, Lin A, Zhang J, Liu F, Zhao J, Gao L, Yi F, Xue T, Sun JP, Gong Y, Yu X. A cullin 4B-RING E3 ligase complex fine-tunes pancreatic δ cell paracrine interactions. J Clin Invest 2017; 127:2631-2646. [PMID: 28604389 DOI: 10.1172/jci91348] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 04/20/2017] [Indexed: 12/24/2022] Open
Abstract
Somatostatin secreted by pancreatic δ cells mediates important paracrine interactions in Langerhans islets, including maintenance of glucose metabolism through the control of reciprocal insulin and glucagon secretion. Disruption of this circuit contributes to the development of diabetes. However, the precise mechanisms that control somatostatin secretion from islets remain elusive. Here, we found that a super-complex comprising the cullin 4B-RING E3 ligase (CRL4B) and polycomb repressive complex 2 (PRC2) epigenetically regulates somatostatin secretion in islets. Constitutive ablation of CUL4B, the core component of the CRL4B-PRC2 complex, in δ cells impaired glucose tolerance and decreased insulin secretion through enhanced somatostatin release. Moreover, mechanistic studies showed that the CRL4B-PRC2 complex, under the control of the δ cell-specific transcription factor hematopoietically expressed homeobox (HHEX), determines the levels of intracellular calcium and cAMP through histone posttranslational modifications, thereby altering expression of the Cav1.2 calcium channel and adenylyl cyclase 6 (AC6) and modulating somatostatin secretion. In response to high glucose levels or urocortin 3 (UCN3) stimulation, increased expression of cullin 4B (CUL4B) and the PRC2 subunit histone-lysine N-methyltransferase EZH2 and reciprocal decreases in Cav1.2 and AC6 expression were found to regulate somatostatin secretion. Our results reveal an epigenetic regulatory mechanism of δ cell paracrine interactions in which CRL4B-PRC2 complexes, Cav1.2, and AC6 expression fine-tune somatostatin secretion and facilitate glucose homeostasis in pancreatic islets.
Collapse
Affiliation(s)
- Qing Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Min Cui
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Fan Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Na Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Baichun Jiang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Genetics, and
| | - Zhen Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Daolai Zhang
- Department of Biochemistry, Shandong University School of Medicine, Jinan, Shandong, China
| | - Yijing Wang
- Department of Biochemistry, Shandong University School of Medicine, Jinan, Shandong, China
| | - Xibin Zhu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Huili Hu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Genetics, and
| | - Pei-Shan Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Genetics, and
| | - Shang-Lei Ning
- Department of Biochemistry, Shandong University School of Medicine, Jinan, Shandong, China
| | - Si Wang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Haibo Qi
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Hechen Song
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Dongfang He
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology.,Department of Biochemistry, Shandong University School of Medicine, Jinan, Shandong, China
| | - Amy Lin
- Duke University, School of Medicine, Durham, North Carolina, USA
| | - Jingjing Zhang
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Liu
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Fan Yi
- Department of Pharmacology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Tian Xue
- Hefei National Laboratory for Physical Science at Microscale, School of Life Science, University of Science and Technology of China, Hefei, Anhui, China
| | - Jin-Peng Sun
- Department of Biochemistry, Shandong University School of Medicine, Jinan, Shandong, China.,Duke University, School of Medicine, Durham, North Carolina, USA
| | - Yaoqin Gong
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Genetics, and
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| |
Collapse
|
680
|
Gadella BM. Reproductive tract modifications of the boar sperm surface. Mol Reprod Dev 2017; 84:822-831. [PMID: 28452082 DOI: 10.1002/mrd.22821] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/10/2017] [Accepted: 04/19/2017] [Indexed: 01/25/2023]
Abstract
The sperm cell has a unique, polarized, and segregated surface that is modified extensively by the changing environments in both the male and the female reproductive tracts. The sperm cannot refresh its surface, as protein translation and membrane recycling by intracellular vesicular transport have ceased upon its maturation. So, how is the sperm surface modified in the reproductive tracts and how do these processes affect fertilization? This review traces these modifications as boar sperm travels from their liberation from the Sertoli cell into the lumen of seminiferous tubules of the testis to the site of fertilization in the ampulla of the oviduct in the sow, via an artificial insemination route. The effect of sperm dilution for artificial insemination, as well as more extensive sperm processing for in vitro fertilization, cryopreservation, or sex sorting, are also discussed with respect to how these procedures affect sperm surface organization and fertilization capacity.
Collapse
Affiliation(s)
- Bart M Gadella
- Faculty of Veterinary Medicine, Department of Farm Animal Health and Biochemistry and Cell Biology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
681
|
CENP-A regulates chromosome segregation during the first meiosis of mouse oocytes. ACTA ACUST UNITED AC 2017; 37:313-318. [PMID: 28585134 DOI: 10.1007/s11596-017-1733-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 09/09/2016] [Indexed: 02/07/2023]
Abstract
Proper chromosome separation in both mitosis and meiosis depends on the correct connection between kinetochores of chromosomes and spindle microtubules. Kinetochore dysfunction can lead to unequal distribution of chromosomes during cell division and result in aneuploidy, thus kinetochores are critical for faithful segregation of chromosomes. Centromere protein A (CENP-A) is an important component of the inner kinetochore plate. Multiple studies in mitosis have found that deficiencies in CENP-A could result in structural and functional changes of kinetochores, leading to abnormal chromosome segregation, aneuploidy and apoptosis in cells. Here we report the expression and function of CENP-A during mouse oocyte meiosis. Our study found that microinjection of CENP-A blocking antibody resulted in errors of homologous chromosome segregation and caused aneuploidy in eggs. Thus, our findings provide evidence that CENP-A is critical for the faithful chromosome segregation during mammalian oocyte meiosis.
Collapse
|
682
|
Ma WW, Xiao J, Song YF, Ding JH, Tan XJ, Song KK, Zhang MM. Effect and underlying mechanism of Bu-Shen-An-Tai recipe on ovarian apoptosis in mice with controlled ovarian hyperstimulation implantation dysfunction. ACTA ACUST UNITED AC 2017; 37:401-406. [PMID: 28585136 DOI: 10.1007/s11596-017-1747-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/04/2017] [Indexed: 12/19/2022]
Abstract
The effect and underlying mechanism of Bu-Shen-An-Tai recipe on ovarian apoptosis in mice with controlled ovarian hyperstimulation (COH) implantation dysfunction were studied. The COH implantation dysfunction model in mice was established by intraperitoneal injection of 7.5 IU pregnant mare's serum gonadotrophin (PMSG), followed by 7.5 IU human chorionic gonadotrophin (HCG) 48 h later. Then the female mice were mated with male at a ratio of 2:1 in the same cage at 6:00 p.m. The female mice from normal group were injected intraperitoneally with normal saline and mated at the corresponding time. Day 1 of pregnancy was recorded by examining its vaginal smears at 8:00 a.m. of the next day. Fifty successfully pregnant mice were equally randomly divided into 5 groups: normal control pregnant group (NC), COH implantation dysfunction model group (COH), low dosage of Bu-Shen-An-Tai recipe group (LOW), middle dosage of Bu-Shen-An-Tai recipe group (MID) and high dosage of Bu-Shen-An-Tai recipe group (HIGH). Then from day 1, the mice in different groups were respectively intragastrically given corresponding treatments at 9:00 a.m. for 5 consecutive days. The concentrations of 17β-estradiol (E2) and progesterone (P4) were determined by radioimmunoassay (RIA). The ultrastructural changes of ovarian tissues were observed by transmission electron microscope (TEM). The histopathological changes of ovarian tissues were observed by HE staining. The number of atretic follicles and pregnant corpus luteum were also recorded. TUNEL was applied to measure apoptotic cells of ovarian tissues. Western blotting was used to detect the protein expression of apoptosis- related factors like Bax, Bcl-2 and cleaved-caspase-3 in ovarian tissue of mice. The results showed that ovarian weight, the concentrations of E2 and P4, the number of atretic follicles and pregnant corpus luteum, as well as the apoptosis of granulosa cells were significantly increased in the COH group. The ultrastructures of ovarian tissues in the COH group showed that chromatin in granulosa cells was increased, agglutinated, aggregated or crescent-shaped. The focal cavitation and the typical apoptotic bodies could be seen in granulosa cells in the late stage of apoptosis. After the treatment with different doses of Bu-Shen-An-Tai recipe, the ultrastructural changes of ovarian granulosa cells apoptosis were dramatically improved and even disappeared under TEM. Visible mitochondria and mitochondrial cristae were increased and vacuoles were significantly reduced. The lipid dropltes were shown in a circluar or oval shape. The protein expression levels of Bax and cleaved-caspase-3 were decreased, and the expression of Bcl-2 protein was increased after treatment. It was concluded that Bu-Shen-An-Tai recipe can inhibit the apoptosis of ovarian granulosa cells, probably by up-regulating the protein expression of Bcl-2 and down-regulating Bax and cleaved-caspase-3, which contributes to the formation and maintenance of ovarian corpus luteum. It's helpful to promote the embryonic implantation, to reduce embryo loss and ultimately to improve the success rate of pregnancy.
Collapse
Affiliation(s)
- Wen-Wen Ma
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing Xiao
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu-Fan Song
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia-Hui Ding
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiu-Juan Tan
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kun-Kun Song
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ming-Min Zhang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
683
|
Chen YH, Wang Q, Zhang YN, Han X, Li DH, Zhang CL. Cumulative live birth and surplus embryo incidence after frozen-thaw cycles in PCOS: how many oocytes do we need? J Assist Reprod Genet 2017; 34:1153-1159. [PMID: 28580513 DOI: 10.1007/s10815-017-0959-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/17/2017] [Indexed: 12/18/2022] Open
Abstract
PURPOSE This study aimed to evaluate the cumulative live birth rate (CLBR) and surplus embryo rate of polycystic ovarian syndrome (PCOS) patients during in vitro fertilization and embryo transfer (IVF-ET) treatment. METHODS In this retrospective cohort study, we analyzed 1142 PCOS patients who underwent first IVF in our institution between January 2011 and December 2014. All patients were categorized into five groups according to the number of oocytes retrieved. Main outcomes include CLBR and surplus embryo rate. RESULTS A strong correlation was observed between number of oocytes retrieved and CLBR as well as surplus embryo rate in PCOS patients. CLBR was elevated with the increasing number of oocytes and plateaued when oocyte number was up to ten, whereas the surplus embryo rate steadily increased in line with the increase of oocyte number. Patients transferred with frozen embryos showed higher CLBR and LBR during first ET than patients transferred with fresh embryos. CONCLUSIONS For PCOS patients, retrieving more than ten oocytes leads to no significant benefit to CLBR but generates surplus embryos. Thus, moderate ovarian stimulation should be reconsidered during IVF treatment.
Collapse
Affiliation(s)
- Yuan-Hui Chen
- Reproductive Medical Center, Henan Provincial People's Hospital, Weiwu Road, Zhengzhou, 450003, Henan Province, People's Republic of China.,Reproductive Medical Center, People's Hospital of Zhengzhou University, Weiwu Road, Zhengzhou, 450003, Henan Province, People's Republic of China
| | - Qian Wang
- Reproductive Medical Center, Henan Provincial People's Hospital, Weiwu Road, Zhengzhou, 450003, Henan Province, People's Republic of China.,Reproductive Medical Center, People's Hospital of Zhengzhou University, Weiwu Road, Zhengzhou, 450003, Henan Province, People's Republic of China
| | - Ya-Nan Zhang
- Reproductive Medical Center, Henan Provincial People's Hospital, Weiwu Road, Zhengzhou, 450003, Henan Province, People's Republic of China.,Reproductive Medical Center, People's Hospital of Zhengzhou University, Weiwu Road, Zhengzhou, 450003, Henan Province, People's Republic of China
| | - Xiao Han
- Reproductive Medical Center, Henan Provincial People's Hospital, Weiwu Road, Zhengzhou, 450003, Henan Province, People's Republic of China.,Reproductive Medical Center, People's Hospital of Zhengzhou University, Weiwu Road, Zhengzhou, 450003, Henan Province, People's Republic of China
| | - Dong-Han Li
- Reproductive Medical Center, Henan Provincial People's Hospital, Weiwu Road, Zhengzhou, 450003, Henan Province, People's Republic of China.,Reproductive Medical Center, People's Hospital of Zhengzhou University, Weiwu Road, Zhengzhou, 450003, Henan Province, People's Republic of China
| | - Cui-Lian Zhang
- Reproductive Medical Center, Henan Provincial People's Hospital, Weiwu Road, Zhengzhou, 450003, Henan Province, People's Republic of China. .,Reproductive Medical Center, People's Hospital of Zhengzhou University, Weiwu Road, Zhengzhou, 450003, Henan Province, People's Republic of China.
| |
Collapse
|
684
|
Li R, Wen B, Zhao H, Ouyang N, Ou S, Wang W, Han J, Yang D. Embryo development after mitochondrial supplementation from induced pluripotent stem cells. J Assist Reprod Genet 2017; 34:1027-1033. [PMID: 28573522 DOI: 10.1007/s10815-017-0948-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 05/09/2017] [Indexed: 10/19/2022] Open
Abstract
PURPOSE The purpose of this study was to evaluate the effects of mitochondrial supplementation (MS) on early embryonic development and to assess the safety of MS treatments using induced pluripotent stem cells (iPSCs) as the mitochondrial donor. METHODS In this study, we evaluated the effect of MS on early embryonic development using induced pluripotent stem cells (iPSCs) as the donor. Mouse zygotes were injected with either mitochondria from iPSCs or a vehicle solution. Several parameters were evaluated, including the rates of blastocyst formation and implantation, the weight of E13.5 embryos and placentas, the distribution of the donor mitochondrial DNA (mtDNA), and the pattern of methylation in the differentially methylated regions (DMRs) of the H19 and Snrpn genes. RESULTS We found that neither the rates of blastocyst formation and implantation nor the weights of E13.5 embryos and placentas were significantly different between the MS and control groups. Additionally, the mtDNA from the iPSC donors could be detected in the muscle tissue of four fetuses and all placentas in the MS group. Finally, the methylation patterns of H19 and Snrpn DMRs remained unchanged by MS. CONCLUSIONS iPSC-derived mtDNA was directly involved in the process of embryonic development after MS. No adverse effects were seen when using iPSCs as a mitochondrial donor, but it remains to be seen whether this method can improve embryonic development, especially in older mice.
Collapse
Affiliation(s)
- Ruiqi Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Bingqiang Wen
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Haijing Zhao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Nengyong Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Songbang Ou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Wenjun Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Jianyong Han
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People's Republic of China.
| | - Dongzi Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China. .,Reproductive Medicine Centre, Department of Obstetrics and Gynaecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
| |
Collapse
|
685
|
Wu C, Xu B, Li X, Ma W, Zhang P, Chen X, Wu J. Tracing and Characterizing the Development of Transplanted Female Germline Stem Cells In Vivo. Mol Ther 2017; 25:1408-1419. [PMID: 28528817 DOI: 10.1016/j.ymthe.2017.04.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 04/16/2017] [Accepted: 04/23/2017] [Indexed: 12/25/2022] Open
Abstract
It has long been believed that most female mammalian species lose the ability to generate oocytes in postnatal ovaries. Recent evidence has demonstrated the isolation and culture of female germline stem cells (FGSCs) from adult mice and humans. However, the process and mechanisms of FGSC differentiation in vivo following transplantation have not yet been studied. Here, we isolated and characterized FGSCs from a single EGFP-transgenic mouse, and traced the development and behavior of transplanted FGSCs (F-TFs) in vivo. Comparisons of folliculogenesis between recipients with FGSC transplantation and wild-type (WT) mice were performed by single follicle RNA-sequencing (RNA-seq). Results showed that FGSCs exhibited a homing ability and began to differentiate into early-stage oocytes only when they reached the edge of the ovarian cortex. The F-TFs restored function of premature ovarian failure (gdf9iCre; PtenloxP/loxP genotype) and generated offspring. Furthermore, results demonstrated that the developmental mechanisms of follicles derived from F-TFs were similar to that of WT follicles. Weighted gene co-expression network analysis identified two potential sub-networks and core genes that played a critical role in follicular development. These findings provide a theoretical basis and lay a technology platform for specific or personalized medical treatment of ovarian failure or other ovarian diseases.
Collapse
Affiliation(s)
- Changqing Wu
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Bo Xu
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaoyong Li
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wenzhi Ma
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Ping Zhang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Xuejin Chen
- Department of Laboratory Animal Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ji Wu
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, China; Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China; Shanghai Key Laboratory of Reproductive Medicine, Shanghai 200025, China.
| |
Collapse
|
686
|
Jeon HJ, Cui XS, Guo J, Lee JM, Kim JS, Oh JS. TCTP regulates spindle assembly during postovulatory aging and prevents deterioration in mouse oocyte quality. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1328-1334. [PMID: 28476647 DOI: 10.1016/j.bbamcr.2017.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 04/27/2017] [Accepted: 05/01/2017] [Indexed: 12/11/2022]
Abstract
If no fertilization occurs for a prolonged time following ovulation, oocytes experience a time-dependent deterioration in quality both in vivo and in vitro due to processes called postovulatory aging. Because the postovulatory aging of oocytes has marked detrimental effects on embryo development and offspring, many efforts have been made to unveil the underlying mechanisms. Here we showed that translationally controlled tumor protein (TCTP) regulates spindle assembly during postovulatory aging and prevents deterioration in mouse oocyte quality. Spindle dynamics decreased with reduced TCTP level during aging of mouse oocytes. Knockdown of TCTP accelerated the reduction of spindle dynamics, accompanying with aging-related deterioration of oocyte quality. Conversely, overexpression of TCTP prevented aging-associated decline of spindle dynamics. Moreover, the aging-related abnormalities in oocytes were rescued after TCTP overexpression, thereby improving fertilization competency and subsequent embryo development. Therefore, our results demonstrate that TCTP-mediated spindle dynamics play a key role in maintaining oocyte quality during postovulatory aging and overexpression of TCTP is sufficient to prevent aging-associated abnormalities in mouse oocytes.
Collapse
Affiliation(s)
- Hyuk-Joon Jeon
- Department of Genetic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Xiang-Shun Cui
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Jing Guo
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Jae Man Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae-Sung Kim
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Jeong Su Oh
- Department of Genetic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
687
|
The relationship between mitochondrial DNA copy number and stallion sperm function. Theriogenology 2017; 94:94-99. [DOI: 10.1016/j.theriogenology.2017.02.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/30/2017] [Accepted: 02/19/2017] [Indexed: 11/18/2022]
|
688
|
Zhao YH, Ji TF, Luo Q, Yu JL. Long non-coding RNA H19 induces hippocampal neuronal apoptosis via Wnt signaling in a streptozotocin-induced rat model of diabetes mellitus. Oncotarget 2017; 8:64827-64839. [PMID: 29029394 PMCID: PMC5630294 DOI: 10.18632/oncotarget.17472] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 03/16/2017] [Indexed: 12/30/2022] Open
Abstract
Defects in hippocampal synaptic plasticity and disorders of memory and learning are the central nervous system complications of diabetes mellitus (DM). Here, we used a streptozotocin-induced rat DM model to investigate the effects of long non-coding RNA H19 (lncRNA H19) on learning and memory and apoptosis of hippocampal neurons, and the involvement of the Wnt signaling. Our data demonstrate that lncRNA H19 is highly expressed in rats with DM. Over-expression of lncRNA H19 increased positioning navigation latency in DM rats and decreased duration of space exploration. lncRNA H19 over-expression also increased hippocampal neuronal apoptosis and expression of Wnt3, β-catenin, TCF-1, Bax, caspase-8 and caspase-3. By contrast, expression of GSK-3β and Bcl-2 was suppressed in DM rats over-expressing lncRNA H19. These results suggest that lncRNA H19 induces hippocampal neuronal apoptosis via Wnt signaling, and that inhibition of lncRNA H19 may serve as a promising novel target for the treatment of cognitive decline in patients with DM.
Collapse
Affiliation(s)
- Yu-Hao Zhao
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, P.R. China
| | - Tie-Feng Ji
- Department of Radiology, The First Hospital of Jilin University, Changchun, P.R. China
| | - Qi Luo
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, P.R. China
| | - Jin-Lu Yu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
689
|
2i Maintains a Naive Ground State in ESCs through Two Distinct Epigenetic Mechanisms. Stem Cell Reports 2017; 8:1312-1328. [PMID: 28457889 PMCID: PMC5425728 DOI: 10.1016/j.stemcr.2017.04.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 01/08/2023] Open
Abstract
Mouse embryonic stem cells (ESCs) are maintained in serum with leukemia inhibitory factor (LIF) to maintain self-renewal and pluripotency. Recently, a 2i culture method was reported using a combination of MEK inhibition (MEKi) and GSK3 inhibition (GSK3i) with LIF to maintain ESCs in a naive ground state. How 2i maintains a ground state of ESCs remains elusive. Here we show that MEKi and GSK3i maintain the ESC ground state by downregulating global DNA methylation through two distinct mechanisms. MEK1 phosphorylates JMJD2C for ubiquitin-mediated protein degradation. Therefore, MEKi increased JMJD2C protein levels but decreased DNMT3 expression. JMJD2C promotes TET1 activity to increase 5-hydroxymethylcytosine (5hmC) levels. GSK3i suppressed DNMT3 expression, thereby decreasing DNA methylation without affecting 5hmC levels. Furthermore, 2i increased PRDM14 expression to inhibit DNMT3A/B protein expression by promoting G9a-mediated DNMT3A/B protein degradation. Collectively, 2i allows ESCs to maintain a naive ground state through JMJD2C-dependent TET1 activation and PRDM14/G9a-mediated DNMT3A/B protein degradation. MEKi increases JMJD2C protein levels and decreases DNMT3 expression in ESCs JMJD2C promotes TET1 hydroxylase activity to increase global 5hmC levels GSK3i decreases global DNA methylation without affecting 5hmC levels 2i-induced PRDM14 expression promotes G9a-mediated DNMT3A/B protein degradation
Collapse
|
690
|
The health outcomes of human offspring conceived by assisted reproductive technologies (ART). J Dev Orig Health Dis 2017; 8:388-402. [DOI: 10.1017/s2040174417000228] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Concerns have been raised about the health and development of children conceived by assisted reproductive technologies (ART) since 1978. Controversially, ART has been linked with adverse obstetric and perinatal outcomes, an increased risk of birth defects, cancers, and growth and development disorders. Emerging evidence suggests that ART treatment may also predispose individuals to an increased risk of chronic ageing related diseases such as obesity, type 2 diabetes and cardiovascular disease. This review will summarize the available evidence on the short-term and long-term health outcomes of ART singletons, as multiple pregnancies after multiple embryos transfer, are associated with low birth weight and preterm delivery, which can separately increase risk of adverse postnatal outcomes, and impact long-term health. We will also examine the potential factors that may contribute to these health risks, and discuss underlying mechanisms, including epigenetic changes that may occur during the preimplantation period and reprogram development in utero, and adult health, later in life. Lastly, this review will consider the future directions with the view to optimize the long-term health of ART children.
Collapse
|
691
|
Elahi F, Lee H, Lee J, Lee ST, Park CK, Hyun SH, Lee E. Effect of rapamycin treatment during post-activation and/or in vitro culture on embryonic development after parthenogenesis and in vitro fertilization in pigs. Reprod Domest Anim 2017; 52:741-748. [PMID: 28397300 DOI: 10.1111/rda.12974] [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: 10/17/2016] [Accepted: 02/26/2017] [Indexed: 11/30/2022]
Abstract
This study investigated the effects of early induction of autophagy on embryonic development in pigs. For this, oocytes or embryos were treated with an autophagy inducer, rapamycin (RP), during post-activation (Pa), in vitro fertilization (IVF) and/or in vitro culture (IVC). When parthenogenesis (PA) embryos were untreated (control) or treated with various concentrations of RP for 4 hr during Pa, 100 nm RP showed a higher blastocyst formation (48.8 ± 2.7%) than the control (34.6 ± 3.0%). When PA embryos were treated during the first 24 hr of IVC, blastocyst formation was increased (p < .05) by 1 and 10 nm RP (61.9 ± 3.0 and 59.6 ± 3.0%, respectively) compared to the control (43.2 ± 1.8%) and 100 nm RP (47.8 ± 3.2%), with a higher embryo cleavage in response to 10 nm RP (87.3 ± 2.4%) than the control (74.1 ± 3.2%). RP treatment during IVC and Pa + IVC showed increased blastocyst formation (44.7 ± 2.5 and 44.1 ± 2.0%, respectively) compared to the control (33.2 ± 2.0%). In addition, RP treatment during Pa and/or IVC increased glutathione content and inversely reduced reactive oxygen species. In IVF, RP treatment for 6 hr during IVF significantly increased embryonic development (34.0 ± 2.6%) compared to the control (24.8 ± 1.6%), but treatment during IVC for 24 hr with RP did not (23.0 ± 3.8%). Autophagy was significantly increased in PA oocytes by the RP treatment during Pa but not altered by the treatment during the first 24 hr of IVC. Overall, RP treatment positively regulated the pre-implantation development of pig embryos, probably by regulating cellular redox state and stimulating autophagy.
Collapse
Affiliation(s)
- F Elahi
- Laboratory of Theriogenology, College of Veterinary Medicine, Kangwon National University, Chuncheon, Korea
| | - H Lee
- Laboratory of Theriogenology, College of Veterinary Medicine, Kangwon National University, Chuncheon, Korea
| | - J Lee
- Laboratory of Theriogenology, College of Veterinary Medicine, Kangwon National University, Chuncheon, Korea.,Institute of Veterinary Science, Kangwon National University, Chuncheon, Korea
| | - S T Lee
- Division of Applied Animal Science, College of Animal Life Science, Kangwon National University, Chuncheon, Korea
| | - C K Park
- Division of Applied Animal Science, College of Animal Life Science, Kangwon National University, Chuncheon, Korea
| | - S-H Hyun
- Laboratory of Veterinary Embryology and Biotechnology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
| | - E Lee
- Laboratory of Theriogenology, College of Veterinary Medicine, Kangwon National University, Chuncheon, Korea.,Institute of Veterinary Science, Kangwon National University, Chuncheon, Korea
| |
Collapse
|
692
|
Gorshinova VK, Tsvirkun DV, Sukhanova IA, Tarasova NV, Volodina MA, Marey MV, Smolnikova VU, Vysokikh MY, Sukhikh GT. Cumulus cell mitochondrial activity in relation to body mass index in women undergoing assisted reproductive therapy. BBA CLINICAL 2017; 7:141-146. [PMID: 28660134 PMCID: PMC5481670 DOI: 10.1016/j.bbacli.2017.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/31/2017] [Accepted: 03/31/2017] [Indexed: 12/27/2022]
Abstract
Most studies have considered the negative influence of obesity on fertility in both genders. In the present study, we assessed mitochondrial activity expressed as the mitochondrial potential index (MPI) in cumulus cells from obese women and women with a normal body mass index (BMI) during assisted reproductive therapy. The results revealed a significant reduction of MPI with increased body mass. The lower MPI levels in cumulus cells from obese women may reflect mitochondrial dysfunction caused by oxidative stress, which can affect the cumulus-oocyte complex and have an impact on oocyte development.
Collapse
Affiliation(s)
- Victoria K Gorshinova
- Federal State Budget Institution "Research Center for Obstetrics, Gynecology and Perinatology", Ministry of Healthcare of the Russian Federation, 4 Oparina street, Moscow 117997, Russian Federation
| | - Daria V Tsvirkun
- Federal State Budget Institution "Research Center for Obstetrics, Gynecology and Perinatology", Ministry of Healthcare of the Russian Federation, 4 Oparina street, Moscow 117997, Russian Federation
| | - Iuliia A Sukhanova
- Federal State Budget Institution "Research Center for Obstetrics, Gynecology and Perinatology", Ministry of Healthcare of the Russian Federation, 4 Oparina street, Moscow 117997, Russian Federation.,Lomonosov Moscow State University, Biology Faculty, Moscow, 1/12 Leninskie Gory, 119234, Russian Federation
| | - Nadezhda V Tarasova
- Federal State Budget Institution "Research Center for Obstetrics, Gynecology and Perinatology", Ministry of Healthcare of the Russian Federation, 4 Oparina street, Moscow 117997, Russian Federation
| | - Maria A Volodina
- Federal State Budget Institution "Research Center for Obstetrics, Gynecology and Perinatology", Ministry of Healthcare of the Russian Federation, 4 Oparina street, Moscow 117997, Russian Federation
| | - Maria V Marey
- Federal State Budget Institution "Research Center for Obstetrics, Gynecology and Perinatology", Ministry of Healthcare of the Russian Federation, 4 Oparina street, Moscow 117997, Russian Federation
| | - Veronika U Smolnikova
- Federal State Budget Institution "Research Center for Obstetrics, Gynecology and Perinatology", Ministry of Healthcare of the Russian Federation, 4 Oparina street, Moscow 117997, Russian Federation
| | - Mikhail Yu Vysokikh
- Federal State Budget Institution "Research Center for Obstetrics, Gynecology and Perinatology", Ministry of Healthcare of the Russian Federation, 4 Oparina street, Moscow 117997, Russian Federation.,Belozerskii Institute of Physico-chemical Biology, Lomonosov Moscow State University, Moscow, 1 Leninskie gory, 119992, Russian Federation
| | - Gennady T Sukhikh
- Federal State Budget Institution "Research Center for Obstetrics, Gynecology and Perinatology", Ministry of Healthcare of the Russian Federation, 4 Oparina street, Moscow 117997, Russian Federation
| |
Collapse
|
693
|
Abstract
Autophagy is a cellular process that allows degradation by the lysosome of cytoplasmic components such as proteins or organelles. Many studies that used model organisms, showed that autophagy plays an important role in multiple developmental processes like degradation of mitochondria of spermatozoids after fertilization, fetal growth or resistance to nutrient starvation. It is also essential to programmed cell death. The involvement of autophagy in these processes may be related to the production of energy resources in conditions of stress or autophagy can selectively degrade specific proteins during development.
Collapse
Affiliation(s)
- Céline Jenzer
- Institut de biologie intégrative de la cellule (I2BC), CEA, CNRS, Université Paris-Sud, Université Paris-Saclay, avenue de La Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Renaud Legouis
- Institut de biologie intégrative de la cellule (I2BC), CEA, CNRS, Université Paris-Sud, Université Paris-Saclay, avenue de La Terrasse, 91198 Gif-sur-Yvette Cedex, France
| |
Collapse
|
694
|
Abstract
Mitochondrial DNA (mtDNA) is actively eliminated from the developing sperm in Drosophila. New work shows that the mitochondrial DNA polymerase, which normally replicates mtDNA, plays a surprising role in mtDNA elimination.
Collapse
Affiliation(s)
- Maulik R Patel
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville TN 37232, USA.
| |
Collapse
|
695
|
Metabolism and chromatin dynamics in health and disease. Mol Aspects Med 2017; 54:1-15. [DOI: 10.1016/j.mam.2016.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 09/22/2016] [Accepted: 09/27/2016] [Indexed: 01/04/2023]
|
696
|
Haertle L, El Hajj N, Dittrich M, Müller T, Nanda I, Lehnen H, Haaf T. Epigenetic signatures of gestational diabetes mellitus on cord blood methylation. Clin Epigenetics 2017; 9:28. [PMID: 28360945 PMCID: PMC5368916 DOI: 10.1186/s13148-017-0329-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 03/20/2017] [Indexed: 02/07/2023] Open
Abstract
Background Intrauterine exposure to gestational diabetes mellitus (GDM) confers a lifelong increased risk for metabolic and other complex disorders to the offspring. GDM-induced epigenetic modifications modulating gene regulation and persisting into later life are generally assumed to mediate these elevated disease susceptibilities. To identify candidate genes for fetal programming, we compared genome-wide methylation patterns of fetal cord bloods (FCBs) from GDM and control pregnancies. Methods and results Using Illumina’s 450K methylation arrays and following correction for multiple testing, 65 CpG sites (52 associated with genes) displayed significant methylation differences between GDM and control samples. Four candidate genes, ATP5A1, MFAP4, PRKCH, and SLC17A4, from our methylation screen and one, HIF3A, from the literature were validated by bisulfite pyrosequencing. The effects remained significant after adjustment for the confounding factors maternal BMI, gestational week, and fetal sex in a multivariate regression model. In general, GDM effects on FCB methylation were more pronounced in women with insulin-dependent GDM who had a more severe metabolic phenotype than women with dietetically treated GDM. Conclusions Our study supports an association between maternal GDM and the epigenetic status of the exposed offspring. Consistent with a multifactorial disease model, the observed FCB methylation changes are of small effect size but affect multiple genes/loci. The identified genes are primary candidates for transmitting GDM effects to the next generation. They also may provide useful biomarkers for the diagnosis, prognosis, and treatment of adverse prenatal exposures. Electronic supplementary material The online version of this article (doi:10.1186/s13148-017-0329-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Larissa Haertle
- Institute of Human Genetics, Julius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, Germany
| | - Nady El Hajj
- Institute of Human Genetics, Julius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, Germany
| | - Marcus Dittrich
- Institute of Human Genetics, Julius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, Germany.,Department of Bioinformatics, Julius Maximilians University, 97074 Würzburg, Germany
| | - Tobias Müller
- Department of Bioinformatics, Julius Maximilians University, 97074 Würzburg, Germany
| | - Indrajit Nanda
- Institute of Human Genetics, Julius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, Germany
| | - Harald Lehnen
- Department of Gynecology and Obstetrics, Municipal Clinics, 41239 Moenchengladbach, Germany
| | - Thomas Haaf
- Institute of Human Genetics, Julius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
697
|
Mitochondrial matters: Mitochondrial bottlenecks, self-assembling structures, and entrapment in the female germline. Stem Cell Res 2017; 21:178-186. [PMID: 28336253 DOI: 10.1016/j.scr.2017.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/22/2016] [Accepted: 03/10/2017] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial replacement therapy, a procedure to generate embryos with the nuclear genome of a donor mother and the healthy mitochondria of a recipient egg, has recently emerged as a promising strategy to prevent transmission of devastating mitochondrial DNA diseases and infertility. The procedure may produce an embryo that is free of diseased mitochondria. A recent study addresses important fundamental questions about the mechanisms underlying maternal inheritance and translational questions regarding the transgenerational effectiveness of this promising therapeutic strategy. This review considers recent advances in our understanding of maternal inheritance of mitochondria, implications for fertility and mitochondrial disease, and potential roles for the Balbiani body, an ancient oocyte structure, in mitochondrial selection in oocytes, with emphasis on therapies to remedy mitochondrial disorders.
Collapse
|
698
|
Yoon A, Tammen SA, Park S, Han SN, Choi SW. Genome-wide hepatic DNA methylation changes in high-fat diet-induced obese mice. Nutr Res Pract 2017; 11:105-113. [PMID: 28386383 PMCID: PMC5376528 DOI: 10.4162/nrp.2017.11.2.105] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 08/04/2016] [Accepted: 12/06/2016] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND/OBJECTIVES A high-fat diet (HFD) induces obesity, which is a major risk factor for cardiovascular disease and cancer, while a calorie-restricted diet can extend life span by reducing the risk of these diseases. It is known that health effects of diet are partially conveyed through epigenetic mechanism including DNA methylation. In this study, we investigated the genome-wide hepatic DNA methylation to identify the epigenetic effects of HFD-induced obesity. MATERIALS AND METHODS Seven-week-old male C57BL/6 mice were fed control diet (CD), calorie-restricted control diet (CRCD), or HFD for 16 weeks (after one week of acclimation to the control diet). Food intake, body weight, and liver weight were measured. Hepatic triacylglycerol and cholesterol levels were determined using enzymatic colorimetric methods. Changes in genome-wide DNA methylation were determined by a DNA methylation microarray method combined with methylated DNA immunoprecipitation. The level of transcription of individual genes was measured by real-time PCR. RESULTS The DNA methylation statuses of genes in biological networks related to lipid metabolism and hepatic steatosis were influenced by HFD-induced obesity. In HFD group, a proinflammatory Casp1 (Caspase 1) gene had hypomethylated CpG sites at the 1.5-kb upstream region of its transcription start site (TSS), and its mRNA level was higher compared with that in CD group. Additionally, an energy metabolism-associated gene Ndufb9 (NADH dehydrogenase 1 beta subcomplex 9) in HFD group had hypermethylated CpG sites at the 2.6-kb downstream region of its TSS, and its mRNA level was lower compared with that in CRCD group. CONCLUSIONS HFD alters DNA methylation profiles in genes associated with liver lipid metabolism and hepatic steatosis. The methylation statuses of Casp1 and Ndufb9 were particularly influenced by the HFD. The expression of these genes in HFD differed significantly compared with CD and CRCD, respectively, suggesting that the expressions of Casp1 and Ndufb9 in liver were regulated by their methylation statuses.
Collapse
Affiliation(s)
- AhRam Yoon
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Stephanie A Tammen
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA.; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA 02111, USA
| | - Soyoung Park
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Sung Nim Han
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.; Research Institute of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Sang-Woon Choi
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA.; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA 02111, USA.; Chaum Life Center, CHA University School of Medicine, Seoul 06062, Korea
| |
Collapse
|
699
|
Gaeini A, Department of Sport Physiology, School of Physical Education and Sport Sciences, University of Tehran, Tehran, Iran, Baghaban Eslaminejad M, Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran., Choobineh S, Department of Sport Physiology, School of Physical Education and Sport Sciences, University of Tehran, Tehran, Iran, Mousavi N, Department of Biochemistry and Nutrition, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran., Satarifard S, Department of Sport Physiology, School of Physical Education and Sport Sciences, University of Tehran, Tehran, Iran, Shafieineek L, Department of Sport Physiology, School of Physical Education and Sport Sciences, University of Tehran, Tehran, Iran. Effects of exercise prior or during pregnancy in high fat diet fed mice alter bone gene expression of female offspring: An experimental study. Int J Reprod Biomed 2017; 15:93-100. [DOI: 10.29252/ijrm.15.2.93] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
700
|
A guide to designing germline-dependent epigenetic inheritance experiments in mammals. Nat Methods 2017; 14:243-249. [DOI: 10.1038/nmeth.4181] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 01/08/2017] [Indexed: 12/13/2022]
|