1
|
Li Y, He R, Qin X, Zhu Q, Ma L, Liang X. Transcriptome analysis during 4-vinylcyclohexene diepoxide exposure-induced premature ovarian insufficiency in mice. PeerJ 2024; 12:e17251. [PMID: 38646488 PMCID: PMC11032656 DOI: 10.7717/peerj.17251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/26/2024] [Indexed: 04/23/2024] Open
Abstract
The occupational chemical 4-Vinylcyclohexene diepoxide (VCD) is a reproductively toxic environmental pollutant that causes follicular failure, leading to premature ovarian insufficiency (POI), which significantly impacts a woman's physical health and fertility. Investigating VCD's pathogenic mechanisms can offer insights for the prevention of ovarian impairment and the treatment of POI. This study established a mouse model of POI through intraperitoneal injection of VCD into female C57BL/6 mice for 15 days. The results were then compared with those of the control group, including a comparison of phenotypic characteristics and transcriptome differences, at two time points: day 15 and day 30. Through a comprehensive analysis of differentially expressed genes (DEGs), key genes were identified and validated some using RT-PCR. The results revealed significant impacts on sex hormone levels, follicle number, and the estrous cycle in VCD-induced POI mice on both day 15 and day 30. The DEGs and enrichment results obtained on day 15 were not as significant as those obtained on day 30. The results of this study provide a preliminary indication that steroid hormone synthesis, DNA damage repair, and impaired oocyte mitosis are pivotal in VCD-mediated ovarian dysfunction. This dysfunction may have been caused by VCD damage to the primordial follicular pool, impairing follicular development and aggravating ovarian damage over time, making it gradually difficult for the ovaries to perform their normal functions.
Collapse
Affiliation(s)
- Yi Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Ruifen He
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Xue Qin
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Qinying Zhu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Liangjian Ma
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaolei Liang
- Gansu Provincial Clinical Research Center for Gynecological Oncology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
2
|
Zhang JL, Xu MF, Chen J, Wei YL, She ZY. Kinesin-7 CENP-E mediates chromosome alignment and spindle assembly checkpoint in meiosis I. Chromosoma 2024; 133:149-168. [PMID: 38456964 DOI: 10.1007/s00412-024-00818-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 02/05/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
In eukaryotes, meiosis is the genetic basis for sexual reproduction, which is important for chromosome stability and species evolution. The defects in meiosis usually lead to chromosome aneuploidy, reduced gamete number, and genetic diseases, but the pathogenic mechanisms are not well clarified. Kinesin-7 CENP-E is a key regulator in chromosome alignment and spindle assembly checkpoint in cell division. However, the functions and mechanisms of CENP-E in male meiosis remain largely unknown. In this study, we have revealed that the CENP-E gene was highly expressed in the rat testis. CENP-E inhibition influences chromosome alignment and spindle organization in metaphase I spermatocytes. We have found that a portion of misaligned homologous chromosomes is located at the spindle poles after CENP-E inhibition, which further activates the spindle assembly checkpoint during the metaphase-to-anaphase transition in rat spermatocytes. Furthermore, CENP-E depletion leads to abnormal spermatogenesis, reduced sperm count, and abnormal sperm head structure. Our findings have elucidated that CENP-E is essential for homologous chromosome alignment and spindle assembly checkpoint in spermatocytes, which further contribute to chromosome stability and sperm cell quality during spermatogenesis.
Collapse
Affiliation(s)
- Jing-Lian Zhang
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China
| | - Meng-Fei Xu
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China
| | - Jie Chen
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China
| | - Ya-Lan Wei
- Medical Research Center, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Zhen-Yu She
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China.
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
3
|
Heddar A, Ogur C, Da Costa S, Braham I, Billaud-Rist L, Findikli N, Beneteau C, Reynaud R, Mahmoud K, Legrand S, Marchand M, Cedrin-Durnerin I, Cantalloube A, Peigne M, Bretault M, Dagher-Hayeck B, Perol S, Droumaguet C, Cavkaytar S, Nicolas-Bonne C, Elloumi H, Khrouf M, Rougier-LeMasle C, Fradin M, Le Boette E, Luigi P, Guerrot AM, Ginglinger E, Zampa A, Fauconnier A, Auger N, Paris F, Brischoux-Boucher E, Cabrol C, Brun A, Guyon L, Berard M, Riviere A, Gruchy N, Odent S, Gilbert-Dussardier B, Isidor B, Piard J, Lambert L, Hamamah S, Guedj AM, Brac de la Perriere A, Fernandez H, Raffin-Sanson ML, Polak M, Letur H, Epelboin S, Plu-Bureau G, Wołczyński S, Hieronimus S, Aittomaki K, Catteau-Jonard S, Misrahi M. Genetic landscape of a large cohort of Primary Ovarian Insufficiency: New genes and pathways and implications for personalized medicine. EBioMedicine 2022; 84:104246. [PMID: 36099812 PMCID: PMC9475279 DOI: 10.1016/j.ebiom.2022.104246] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/29/2022] Open
Abstract
Background Primary Ovarian Insufficiency (POI), a public health problem, affects 1-3.7% of women under 40 yielding infertility and a shorter lifespan. Most causes are unknown. Recently, genetic causes were identified, mostly in single families. We studied an unprecedented large cohort of POI to unravel its molecular pathophysiology. Methods 375 patients with 70 families were studied using targeted (88 genes) or whole exome sequencing with pathogenic/likely-pathogenic variant selection. Mitomycin-induced chromosome breakages were studied in patients’ lymphocytes if necessary. Findings A high-yield of 29.3% supports a clinical genetic diagnosis of POI. In addition, we found strong evidence of pathogenicity for nine genes not previously related to a Mendelian phenotype or POI: ELAVL2, NLRP11, CENPE, SPATA33, CCDC150, CCDC185, including DNA repair genes: C17orf53(HROB), HELQ, SWI5 yielding high chromosomal fragility. We confirmed the causal role of BRCA2, FANCM, BNC1, ERCC6, MSH4, BMPR1A, BMPR1B, BMPR2, ESR2, CAV1, SPIDR, RCBTB1 and ATG7 previously reported in isolated patients/families. In 8.5% of cases, POI is the only symptom of a multi-organ genetic disease. New pathways were identified: NF-kB, post-translational regulation, and mitophagy (mitochondrial autophagy), providing future therapeutic targets. Three new genes have been shown to affect the age of natural menopause supporting a genetic link. Interpretation We have developed high-performance genetic diagnostic of POI, dissecting the molecular pathogenesis of POI and enabling personalized medicine to i) prevent/cure comorbidities for tumour/cancer susceptibility genes that could affect life-expectancy (37.4% of cases), or for genetically-revealed syndromic POI (8.5% of cases), ii) predict residual ovarian reserve (60.5% of cases). Genetic diagnosis could help to identify patients who may benefit from the promising in vitro activation-IVA technique in the near future, greatly improving its success in treating infertility. Funding Université Paris Saclay, Agence Nationale de Biomédecine.
Collapse
Affiliation(s)
- Abdelkader Heddar
- Université Paris Saclay, Faculté de Médecine. Unité de Génétique Moléculaire des Maladies Métaboliques et de la Reproduction, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, AP-HP, Hôpitaux Universitaires Paris-Saclay, Le Kremlin-Bicêtre, France; UMR-S 1193, INSERM, Université Paris Saclay, Faculté de Médecine, Hôpital Paul Brousse, Villejuif, France
| | - Cagri Ogur
- Igenomix Turkey, İstanbul, Turkey; Institute of Science, Department of Bioengineering Yildiz Technical University, İstanbul, Turkey
| | - Sabrina Da Costa
- Service d'Endocrinologie Pédiatrique, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, CNR pathologies gynécologiques rares, 75015, Paris, France
| | - Inès Braham
- Service d'Endocrinologie et de Médicine de la Reproduction, Hôpital Universitaire de Nice, 06200, Nice, France
| | - Line Billaud-Rist
- Service d'Endocrinologie, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin/Port-Royal, 75005, Paris, France
| | - Necati Findikli
- Bahçeci Umut IVF Centre, Altunizade, İstanbul, Turkey; Faculty of Engineering and Architecture, Department of Biomedical Engineering, Beykent University, İstanbul, Turkey
| | - Claire Beneteau
- Service de Génétique Médicale, Centre Hospitalier Universitaire Nantes, 44000, Nantes, France
| | - Rachel Reynaud
- Aix Marseille Université, Assistance-Publique des Hôpitaux de Marseille (AP-HM), Service de Pédiatrie multidisciplinaire Hôpital de la Timone Enfants, 13385, Marseille Cedex 05, France
| | - Khaled Mahmoud
- Centre FERTILLIA de Médecine de la Reproduction- Clinique la ROSE, Tunis, Tunisie
| | - Stéphanie Legrand
- Centre de Fertilité - Clinique de l'Atlantique La Rochelle, 17000, La Rochelle, France
| | - Maud Marchand
- Service d'Endocrinologie Pédiatrique, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, CNR pathologies gynécologiques rares, 75015, Paris, France
| | - Isabelle Cedrin-Durnerin
- Service de Médecine de la Reproduction et Préservation de la Fertilité, hôpital Jean-Verdier, Assistance Publique-Hôpitaux de Paris, 93143 Bondy, France
| | - Adèle Cantalloube
- Service de Gynécologie et d'Obstétrique, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, AP-HP. Faculté de Médecine Pierre et Marie Curie. Université de la Sorbonne, Paris, France
| | - Maeliss Peigne
- Service de Médecine de la Reproduction et Préservation de la Fertilité, hôpital Jean-Verdier, Assistance Publique-Hôpitaux de Paris, 93143 Bondy, France
| | - Marion Bretault
- Service d'Endocrinologie, Hôpital Ambroise Paré, Assistance Publique-Hôpitaux de Paris, 92100, Boulogne Billancourt, France
| | - Benedicte Dagher-Hayeck
- Service de Médecine de la Reproduction et Préservation de la Fertilité, hôpital Jean-Verdier, Assistance Publique-Hôpitaux de Paris, 93143 Bondy, France
| | - Sandrine Perol
- Unité de gynécologie médicale, APHP, Hôpital Port-Royal Cochin, 27 Rue du Faubourg Saint-Jacques, Paris 75014, France
| | - Celine Droumaguet
- Service de Médecine Interne, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, 94000 Créteil, France
| | - Sabri Cavkaytar
- Bahçeci Umut IVF Centre, Altunizade, İstanbul, Turkey; Üsküdar University, Faculty of Medicine, Department of Obstetrics and Gynecology, İstanbul, Turkey
| | - Carole Nicolas-Bonne
- Service de Gynécologie et d'Obstétrique, Centre Hospitalier Alpes Léman, 74130, Contamine-Sur-Arve, France
| | - Hanen Elloumi
- Centre FERTILLIA de Médecine de la Reproduction- Clinique la ROSE, Tunis, Tunisie
| | - Mohamed Khrouf
- Centre FERTILLIA de Médecine de la Reproduction- Clinique la ROSE, Tunis, Tunisie
| | - Charlotte Rougier-LeMasle
- Service d'Endocrinologie et de Médicine de la Reproduction, Hôpital Universitaire de Nice, 06200, Nice, France
| | - Melanie Fradin
- Service de Génétique Clinique, Centre Hospitalier Universitaire de Rennes, Hôpital Sud, Univ Rennes, CNRS IGDR UMR 6290, Centre de référence Anomalies du développement CLAD-Ouest, ERN ITHACA, 35203, Rennes, France; Service de Génétique Médicale, Centre Hospitalier de Saint Brieuc, 22000, Saint-Brieuc, France
| | - Elsa Le Boette
- Service de Génétique Médicale, Centre Hospitalier de Saint Brieuc, 22000, Saint-Brieuc, France
| | - Perrine Luigi
- Service d'Endocrinologie-Diabétologie, Centre Hospitalier Antibes Juan Les Pins, 06600, Antibes, France
| | - Anne-Marie Guerrot
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Genetics and reference center for developmental disorders, FHU G4 Génomique, F-76000 Rouen, France
| | | | - Amandine Zampa
- Service de Génétique, Centre Hospitalier de Mulhouse, 68100, Mulhouse, France
| | - Anais Fauconnier
- Service d'Endocrinologie, Diabète et Maladies Métaboliques, Centre Hospitalier Universitaire de Saint-Etienne, 42270, Saint-Priest-en-Jarez, France
| | - Nathalie Auger
- Service de génétique des tumeurs. Institut Gustave Roussy, 94805, Villejuif, France
| | - Françoise Paris
- Département de Pédiatrie, Unité d'Endocrinologie-Gynécologie Pédiatrique, Hôpital A.-de-Villeneuve, Centre Hospitalier Universitaire Montpellier et Université Montpellier, 34090, Montpellier, France; Constitutif Sud, Centre de Référence Maladies Rares du Développement Génital, Hôpital Lapeyronie, Centre Hospitalier Universitaire Montpellier, Université de Montpellier, 34090 Montpellier, France; INSERM 1203, Développement Embryonnaire Fertilité Environnement, Université de Montpellier, 34090, Montpellier, France
| | - Elise Brischoux-Boucher
- Centre de Génétique Humaine, Université de Franche-Comté, Centre Hospitalier Universitaire de Besançon, 25000, Besançon, France
| | - Christelle Cabrol
- Centre de Génétique Humaine, Université de Franche-Comté, Centre Hospitalier Universitaire de Besançon, 25000, Besançon, France
| | - Aurore Brun
- Service de Génétique, Centre Hospitalier Universitaire de Poitiers, Université de Poitiers, 86021, Poitiers, France
| | - Laura Guyon
- Service de Génétique Médicale, Centre Hospitalier Universitaire Nantes, 44000, Nantes, France
| | - Melanie Berard
- Service de Génétique Clinique, Centre Hospitalier Régional Universitaire de Nancy, F-54000, Nancy, France
| | - Axelle Riviere
- Service de Génétique Clinique, Centre Hospitalier Régional Universitaire de Nancy, F-54000, Nancy, France
| | - Nicolas Gruchy
- Normandy University, UNICAEN, Caen University Hospital, Department of Genetics, EA 7450 BioTARGen, FHU G4 Genomics, Caen, France
| | - Sylvie Odent
- Service de Génétique Clinique, Centre Hospitalier Universitaire de Rennes, Hôpital Sud, Univ Rennes, CNRS IGDR UMR 6290, Centre de référence Anomalies du développement CLAD-Ouest, ERN ITHACA, 35203, Rennes, France
| | - Brigitte Gilbert-Dussardier
- Service de Génétique, Centre Hospitalier Universitaire de Poitiers, Université de Poitiers, 86021, Poitiers, France
| | - Bertrand Isidor
- Service de Génétique Médicale, Centre Hospitalier Universitaire Nantes, 44000, Nantes, France
| | - Juliette Piard
- Centre de Génétique Humaine, Université de Franche-Comté, Centre Hospitalier Universitaire de Besançon, 25000, Besançon, France
| | - Laetitia Lambert
- Service de Génétique Clinique, Centre Hospitalier Régional Universitaire de Nancy, F-54000, Nancy, France
| | - Samir Hamamah
- INSERM 1203, Développement Embryonnaire Fertilité Environnement, Université de Montpellier, 34090, Montpellier, France; Centre Hospitalier Universitaire de Montpellier, Département de Biologie de la Reproduction, Biologie de la Reproduction/DPI et CECOS, Université de Montpellier, Montpellier, France
| | - Anne Marie Guedj
- Service d'Endocrinologie et de Maladies Métaboliques, Centre Hospitalier Universitaire Nîmes, Université de Montpellier, 30029, Nîmes, France
| | - Aude Brac de la Perriere
- Fédération d'Endocrinologie, Centre de Référence des Maladies Rares du Développement Génital, Groupement Hospitalier Est, Hôpital Louis Pradel, 69002, Lyon, France
| | - Hervé Fernandez
- Service de Gynecologie et d'Obstétrique, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Faculté de médicine, Université Paris-Saclay, 94270 Le Kremlin Bicêtre, France; UVSQ, Inserm, CESP, Université Paris-Saclay, 94807 Villejuif, France
| | - Marie-Laure Raffin-Sanson
- Service d'Endocrinologie, Hôpital Ambroise Paré, Assistance Publique-Hôpitaux de Paris, 92100, Boulogne Billancourt, France
| | - Michel Polak
- Service d'Endocrinologie Pédiatrique, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, CNR pathologies gynécologiques rares, 75015, Paris, France
| | - Hélène Letur
- Service de Gynécologie Obstétrique et Médecine de la Reproduction, Hôpital Foch, 40 rue Worth 92 150 Suresnes, France; Service de Médecine de la Reproduction et Préservation de la Fertilité, Polyclinique de Navarre, 8, boulevard Hauterive, 64000 Pau, France
| | - Sylvie Epelboin
- Service de Gynécologie et d'Obstétrique, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, AP-HP. Faculté de Médecine Pierre et Marie Curie. Université de la Sorbonne, Paris, France
| | - Genevieve Plu-Bureau
- Unité de gynécologie médicale, APHP, Hôpital Port-Royal Cochin, 27 Rue du Faubourg Saint-Jacques, Paris 75014, France
| | - Sławomir Wołczyński
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Sylvie Hieronimus
- Service d'Endocrinologie et de Médicine de la Reproduction, Hôpital Universitaire de Nice, 06200, Nice, France
| | - Kristiina Aittomaki
- Department of Clinical Genetics, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Sophie Catteau-Jonard
- Service de gynécologie médicale, orthogénie et sexologie, Centre Hospitalier Universitaire de Lille, Université de Lille, 59000 Lille, France
| | - Micheline Misrahi
- Université Paris Saclay, Faculté de Médecine. Unité de Génétique Moléculaire des Maladies Métaboliques et de la Reproduction, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, AP-HP, Hôpitaux Universitaires Paris-Saclay, Le Kremlin-Bicêtre, France; UMR-S 1193, INSERM, Université Paris Saclay, Faculté de Médecine, Hôpital Paul Brousse, Villejuif, France.
| |
Collapse
|
4
|
Pan P, Huang X. The Clinical Application of Growth Hormone and Its Biological and Molecular Mechanisms in Assisted Reproduction. Int J Mol Sci 2022; 23:ijms231810768. [PMID: 36142677 PMCID: PMC9505823 DOI: 10.3390/ijms231810768] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/11/2022] [Accepted: 09/12/2022] [Indexed: 11/26/2022] Open
Abstract
Growth hormone (GH) has been used as a co-gonadotrophin in assisted reproduction, particularly in poor ovarian responders. The application of GH has been alleged to activate primordial follicles and improve oocyte quality, embryo quality, and steroidogenesis. However, the effects of GH on the live birth rate among women is controversial. Additionally, although the basic biological mechanisms that lead to the above clinical differences have been investigated, they are not yet well understood. The actions of GH are mediated by GH receptors (GHRs) or insulin-like growth factors (IGFs). GH regulates the vital signal transduction pathways that are involved in primordial follicular activation, steroidogenesis, and oocyte maturation. However, the therapeutic windows and duration of GH administration during assisted reproductive technology require further investigation. The review aimed to clarify the role of GH in human fertility from a molecular and biological point of view to provide evidence for proper GH administration.
Collapse
|
5
|
She ZY, Xu MF, Jiang SY, Wei YL. Kinesin-7 CENP-E is essential for chromosome alignment and spindle assembly of mouse spermatocytes. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119306. [PMID: 35680098 DOI: 10.1016/j.bbamcr.2022.119306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/28/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
Genome stability depends on chromosome congression and alignment during cell division. Kinesin-7 CENP-E is critical for kinetochore-microtubule attachment and chromosome alignment, which contribute to genome stability in mitosis. However, the functions and mechanisms of CENP-E in the meiotic division of male spermatocytes remain largely unknown. In this study, by combining the use of chemical inhibitors, siRNA-mediated gene knockdown, immunohistochemistry, and high-resolution microscopy, we have found that CENP-E inhibition results in chromosome misalignment and metaphase arrest in dividing spermatocyte during meiosis. Strikingly, we have revealed that CENP-E regulates spindle organization in metaphase I spermatocytes and cultured GC-2 spd cells. CENP-E depletion leads to spindle elongation, chromosome misalignment, and chromosome instability in spermatocytes. Together, these findings indicate that CENP-E mediates the kinetochore recruitment of BubR1, spindle assembly checkpoint and chromosome alignment in dividing spermatocytes, which finally contribute to faithful chromosome segregation and chromosome stability in the male meiotic division.
Collapse
Affiliation(s)
- Zhen-Yu She
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China; Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian 350122, China.
| | - Meng-Fei Xu
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China; Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian 350122, China
| | - Sun-Ying Jiang
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China; Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian 350122, China
| | - Ya-Lan Wei
- Fujian Obstetrics and Gynecology Hospital, Fuzhou, Fujian 350011, China; Medical Research Center, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, China
| |
Collapse
|
6
|
Toralova T, Kinterova V, Chmelikova E, Kanka J. The neglected part of early embryonic development: maternal protein degradation. Cell Mol Life Sci 2020; 77:3177-3194. [PMID: 32095869 PMCID: PMC11104927 DOI: 10.1007/s00018-020-03482-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 01/24/2020] [Accepted: 02/07/2020] [Indexed: 12/28/2022]
Abstract
The degradation of maternally provided molecules is a very important process during early embryogenesis. However, the vast majority of studies deals with mRNA degradation and protein degradation is only a very little explored process yet. The aim of this article was to summarize current knowledge about the protein degradation during embryogenesis of mammals. In addition to resuming of known data concerning mammalian embryogenesis, we tried to fill the gaps in knowledge by comparison with facts known about protein degradation in early embryos of non-mammalian species. Maternal protein degradation seems to be driven by very strict rules in terms of specificity and timing. The degradation of some maternal proteins is certainly necessary for the normal course of embryonic genome activation (EGA) and several concrete proteins that need to be degraded before major EGA have been already found. Nevertheless, the most important period seems to take place even before preimplantation development-during oocyte maturation. The defects arisen during this period seems to be later irreparable.
Collapse
Affiliation(s)
- Tereza Toralova
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - Veronika Kinterova
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic.
- Department of Veterinary Sciences, Czech University of Life Sciences in Prague, Prague, Czech Republic.
| | - Eva Chmelikova
- Department of Veterinary Sciences, Czech University of Life Sciences in Prague, Prague, Czech Republic
| | - Jiri Kanka
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| |
Collapse
|
7
|
She ZY, Yu KW, Zhong N, Xiao Y, Wei YL, Lin Y, Li YL, Lu MH. Kinesin-7 CENP-E regulates chromosome alignment and genome stability of spermatogenic cells. Cell Death Discov 2020; 6:25. [PMID: 32351712 PMCID: PMC7171076 DOI: 10.1038/s41420-020-0261-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/04/2020] [Accepted: 04/05/2020] [Indexed: 12/22/2022] Open
Abstract
Kinesin-7 CENP-E is an essential kinetochore motor required for chromosome alignment and congression. However, the specific functions of CENP-E in the spermatogenic cells during spermatogenesis remain unknown. In this study, we find that CENP-E proteins are expressed in the spermatogonia, spermatocytes, and the elongating spermatids. CENP-E inhibition by specific inhibitor GSK923295 results in the disruption of spermatogenesis and cell cycle arrest of spermatogenic cells. Both spermatogonia and spermatocytes are arrested in metaphase and several chromosomes are not aligned at the equatorial plate. We find that CENP-E inhibition leads to chromosome misalignment, the spindle disorganization, and the formation of the aneuploidy cells. Furthermore, the inhibition of CENP-E results in the defects in the formation of spermatids, including the sperm head condensation and the sperm tail formation. We have revealed that kinesin-7 CENP-E is essential for chromosome alignment and genome stability of the spermatogenic cells.
Collapse
Affiliation(s)
- Zhen-Yu She
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350122 China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, 350122 China
| | - Kai-Wei Yu
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350122 China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, 350122 China
| | - Ning Zhong
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350122 China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, 350122 China
| | - Yu Xiao
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350122 China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, 350122 China
| | - Ya-Lan Wei
- Fujian Obstetrics and Gynecology Hospital, Fuzhou, Fujian, 350001 China
- Medical Research Center, Fujian Provincial Children’s Hospital, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350001 China
| | - Yang Lin
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350122 China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, 350122 China
| | - Yue-Ling Li
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350122 China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, 350122 China
| | - Ming-Hui Lu
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350122 China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, 350122 China
| |
Collapse
|
8
|
Chu Q, Yao C, Qi X, Stripp BR, Tang N. STK11 is required for the normal program of ciliated cell differentiation in airways. Cell Discov 2019; 5:36. [PMID: 31636950 PMCID: PMC6796922 DOI: 10.1038/s41421-019-0104-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/06/2019] [Accepted: 05/31/2019] [Indexed: 01/07/2023] Open
Abstract
The functional properties of mucosal surfaces are dependent on establishing the correct proportions of specialized epithelial cell types. Multiciliated cells (also known as ciliated cells) are evolutionarily conserved and functionally indispensable epithelial cells, as suggested by the link between ciliated cell dysfunction and chronic human disease. Ciliated cell differentiation is an ordered process that involves initial cell fate determination and multiciliogenesis. STK11, a serine/threonine kinase, has been reported to be downregulated in human diseases associated with ciliopathies and functions as a tumor suppressor. Here, we show that STK11 is a physiological factor for the normal program of ciliated cell differentiation by phosphorylating MARK3, which directly suppresses ERK1/2 mediated pRB inactivation. Loss of Stk11 in airway progenitors impairs the differentiation of ciliated cells in both embryonic and adult airways. Our study establishes that STK11/MARK3/ERK1/2 signaling cascade is a key regulator to integrate ciliated cell fate commitment and the subsequent process of multiciliogenesis.
Collapse
Affiliation(s)
- Qiqi Chu
- College of Life Sciences, Beijing Normal University, 100875 Beijing, China
- National Institute of Biological Sciences, 102206 Beijing, China
| | - Changfu Yao
- Lung and Board of Governors Regenerative Medicine Institutes, Department of Medicine, Cedars-Sinai Medical Center, 90048 Los Angeles, CA USA
| | - Xiangbing Qi
- National Institute of Biological Sciences, 102206 Beijing, China
| | - Barry Raymond Stripp
- Lung and Board of Governors Regenerative Medicine Institutes, Department of Medicine, Cedars-Sinai Medical Center, 90048 Los Angeles, CA USA
| | - Nan Tang
- National Institute of Biological Sciences, 102206 Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, 100084 Beijing, China
| |
Collapse
|
9
|
Finding of bands of higher molecular weight than expected in three proteins in bovine preimplantation embryos. ZYGOTE 2019; 27:187-189. [PMID: 31182173 DOI: 10.1017/s0967199419000133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SummaryWe report here the existence of bands of higher molecular weight after western blot analysis in three proteins - Skp1, p27 and IκBα in bovine preimplantation embryos. This finding is specific to preimplantation embryos (from the 2-cell stage to the blastocyst stage) and not differentiated fibroblast cells in which these bands were of expected molecular weight. We suggest that these bands of higher molecular weight represent a complex of proteins that are characteristic of preimplantation embryos.
Collapse
|
10
|
Li Y, Liu H, Yu Q, Liu H, Huang T, Zhao S, Ma J, Zhao H. Growth Hormone Promotes in vitro Maturation of Human Oocytes. Front Endocrinol (Lausanne) 2019; 10:485. [PMID: 31396155 PMCID: PMC6667636 DOI: 10.3389/fendo.2019.00485] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/04/2019] [Indexed: 01/18/2023] Open
Abstract
Increasing the success rate of in vitro maturation (IVM) for human oocytes has a major clinical significance. Previous studies have shown that growth hormone (GH) added into IVM medium could promote IVM of oocytes from non-human beings. However, few studies on systematic IVM for human oocytes with GH have been reported. Human germinal vesicle (GV) oocytes collected for IVM were cultured with different concentrations of GH to optimize the concentration. Metaphase II (MII) stage oocytes obtained from IVM were fertilized by intracytoplasmic sperm injection (ICSI). Maturation rate, fertilization rate, and blastocyst rate were assessed after IVM with or without GH. Furthermore, gene expression profiles were compared in oocytes between the two groups using single-cell RNA-seq. The optimal concentration of GH for IVM was 200 ng/ml, and the maturation rate of this group reached 70% which was double that of the control group (35%, P = 0.004). The fertilization rate (73.1 vs. 60.3%) and blastocyst rate (25.0 vs. 15.5%) both had an increasing trend in the GH group compared to controls. Single-cell RNA-Seq and real-time PCR data showed that GH could significantly enhance the expression of genes associated with meiotic progression and embryo development, such as AURKA (aurora kinase A, P = 0.007), PDIA6 (protein disulfide isomerase family A member 6, P = 0.007), LINGO2 (leucine rich repeat and Ig domain containing 2, P = 0.007), and CENPJ (centromere protein J, P = 0.039). Taken together, GH could promote maturation of human oocytes, probably through accelerating meiotic progression, balancing redox homeostasis of cellular environment, and promoting oocyte developmental competence.
Collapse
Affiliation(s)
- Yue Li
- Center for Reproductive Medicine, Shandong University, Jinan, China
- The Key Laboratory of Reproductive Endocrinology of Ministry of Education, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Hui Liu
- Center for Reproductive Medicine, Shandong University, Jinan, China
- The Key Laboratory of Reproductive Endocrinology of Ministry of Education, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Qingqing Yu
- Center for Reproductive Medicine, Shandong University, Jinan, China
- The Key Laboratory of Reproductive Endocrinology of Ministry of Education, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Hongbin Liu
- Center for Reproductive Medicine, Shandong University, Jinan, China
- The Key Laboratory of Reproductive Endocrinology of Ministry of Education, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Tao Huang
- Center for Reproductive Medicine, Shandong University, Jinan, China
- The Key Laboratory of Reproductive Endocrinology of Ministry of Education, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Shigang Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, China
- The Key Laboratory of Reproductive Endocrinology of Ministry of Education, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- *Correspondence: Shigang Zhao
| | - Jinlong Ma
- Center for Reproductive Medicine, Shandong University, Jinan, China
- The Key Laboratory of Reproductive Endocrinology of Ministry of Education, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Han Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, China
- The Key Laboratory of Reproductive Endocrinology of Ministry of Education, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| |
Collapse
|
11
|
Camlin NJ, McLaughlin EA, Holt JE. Motoring through: the role of kinesin superfamily proteins in female meiosis. Hum Reprod Update 2017; 23:409-420. [PMID: 28431155 DOI: 10.1093/humupd/dmx010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 04/01/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The kinesin motor protein family consists of 14 distinct subclasses and 45 kinesin proteins in humans. A large number of these proteins, or their orthologues, have been shown to possess essential function(s) in both the mitotic and the meiotic cell cycle. Kinesins have important roles in chromosome separation, microtubule dynamics, spindle formation, cytokinesis and cell cycle progression. This article contains a review of the literature with respect to the role of kinesin motor proteins in female meiosis in model species. Throughout, we discuss the function of each class of kinesin proteins during oocyte meiosis, and where such data are not available their role in mitosis is considered. Finally, the review highlights the potential clinical importance of this family of proteins for human oocyte quality. OBJECTIVE AND RATIONALE To examine the role of kinesin motor proteins in oocyte meiosis. SEARCH METHODS A search was performed on the Pubmed database for journal articles published between January 1970 and February 2017. Search terms included 'oocyte kinesin' and 'meiosis kinesin' in addition to individual kinesin names with the terms oocyte or meiosis. OUTCOMES Within human cells 45 kinesin motor proteins have been discovered, with the role of only 13 of these proteins, or their orthologues, investigated in female meiosis. Furthermore, of these kinesins only half have been examined in mammalian oocytes, despite alterations occurring in gene transcripts or protein expression with maternal ageing, cryopreservation or behavioral conditions, such as binge drinking, for many of them. WIDER IMPLICATIONS Kinesin motor proteins have distinct and important roles throughout oocyte meiosis in many non-mammalian model species. However, the functions these proteins have in mammalian meiosis, particularly in humans, are less clear owing to lack of research. This review brings to light the need for more experimental investigation of kinesin motor proteins, particularly those associated with maternal ageing, cryopreservation or exposure to environmental toxicants.
Collapse
Affiliation(s)
- Nicole J Camlin
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.,Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Eileen A McLaughlin
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.,Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia.,School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Janet E Holt
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
12
|
De La Fuente R, Baumann C, Viveiros MM. Chromatin structure and ATRX function in mouse oocytes. Results Probl Cell Differ 2012; 55:45-68. [PMID: 22918800 DOI: 10.1007/978-3-642-30406-4_3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Differentiation of chromatin structure and function during oogenesis is essential to confer the mammalian oocyte with meiotic and developmental potential. Errors in chromosome segregation during female meiosis and subsequent transmission of an abnormal chromosome complement (aneuploidy) to the early conceptus are one of the leading causes of pregnancy loss in women. The chromatin remodeling protein ATRX (α-thalassemia mental retardation X-linked) has recently emerged as a critical factor involved in heterochromatin formation at mammalian centromeres during meiosis. In mammalian oocytes, ATRX binds to centromeric heterochromatin domains where it is required for accurate chromosome segregation. Loss of ATRX function induces abnormal meiotic chromosome morphology, reduces histone H3 phosphorylation, and promotes a high incidence of aneuploidy associated with severely reduced fertility. The presence of centromeric breaks during the transition to the first mitosis in the early embryo indicates that the role of ATRX in chromosome segregation is mediated through an epigenetic mechanism involving the maintenance of chromatin modifications associated with pericentric heterochromatin (PCH) formation and chromosome condensation. This is consistent with the existence of a potential molecular link between centromeric and PCH in the epigenetic control of centromere function and maintenance of chromosome stability in mammalian oocytes. Dissecting the molecular mechanisms of ATRX function during meiosis will have important clinical implications towards uncovering the epigenetic factors contributing to the onset of aneuploidy in the human oocyte.
Collapse
|
13
|
Brockmann C, Huarte J, Dugina V, Challet L, Rey E, Conne B, Swetloff A, Nef S, Chaponnier C, Vassalli JD. Beta- and gamma-cytoplasmic actins are required for meiosis in mouse oocytes. Biol Reprod 2011; 85:1025-39. [PMID: 21778137 DOI: 10.1095/biolreprod.111.091736] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
In mammals, female meiosis consists of two asymmetric cell divisions, which generate a large haploid oocyte and two small polar bodies. Asymmetric partitioning of the cytoplasm results from migration of the meiotic spindle toward the cortex and requires actin filaments. However, the subcellular localization and the role of the existing two cytoplasmic actin (CYA) isoforms, beta and gamma, have not been characterized. We show that beta- and gamma-CYA are differentially distributed in the maturing oocyte from late metaphase I as well as in preimplantation embryos. Gamma-CYA is preferentially enriched in oocyte cortices and is absent from all cell-cell contact areas from metaphase II until the blastocyst stage. Beta-CYA is enriched in contractile structures, at cytokinesis, at cell-cell contacts, and around the forming blastocoel. Alteration of beta- or gamma-CYA function by isoform-specific antibody microinjection suggests that gamma-CYA holds a major and specific role in the establishment and/or maintenance of asymmetry in meiosis I and in the maintenance of overall cortical integrity. In contrast, beta- and gamma-CYA, together, appear to participate in the formation and the cortical anchorage of the second meiotic spindle in waiting for fertilization. Finally, differences in gamma-CYA expression are amongst the earliest markers of cell fate determination in development.
Collapse
Affiliation(s)
- Céline Brockmann
- Departments of Genetic Medicine and Development and Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Kronja I, Kruljac-Letunic A, Caudron-Herger M, Bieling P, Karsenti E. XMAP215-EB1 interaction is required for proper spindle assembly and chromosome segregation in Xenopus egg extract. Mol Biol Cell 2009; 20:2684-96. [PMID: 19369422 DOI: 10.1091/mbc.e08-10-1051] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
In metaphase Xenopus egg extracts, global microtubule growth is mainly promoted by two unrelated microtubule stabilizers, end-binding protein 1 (EB1) and XMAP215. Here, we explore their role and potential redundancy in the regulation of spindle assembly and function. We find that at physiological expression levels, both proteins are required for proper spindle architecture: Spindles assembled in the absence of EB1 or at decreased XMAP215 levels are short and frequently multipolar. Moreover, the reduced density of microtubules at the equator of DeltaEB1 or DeltaXMAP215 spindles leads to faulty kinetochore-microtubule attachments. These spindles also display diminished poleward flux rates and, upon anaphase induction, they neither segregate chromosomes nor reorganize into interphasic microtubule arrays. However, EB1 and XMAP215 nonredundantly regulate spindle assembly because an excess of XMAP215 can compensate for the absence of EB1, whereas the overexpression of EB1 cannot substitute for reduced XMAP215 levels. Our data indicate that EB1 could positively regulate XMAP215 by promoting its binding to the microtubules. Finally, we show that disruption of the mitosis-specific XMAP215-EB1 interaction produces a phenotype similar to that of either EB1 or XMAP215 depletion. Therefore, the XMAP215-EB1 interaction is required for proper spindle organization and chromosome segregation in Xenopus egg extracts.
Collapse
Affiliation(s)
- Iva Kronja
- Department of Cell Biology and Biophysics, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
15
|
Otsuki J, Nagai Y, Chiba K. Association of spindle midzone particles with polo-like kinase 1 during meiosis in mouse and human oocytes. Reprod Biomed Online 2009; 18:522-8. [PMID: 19400994 DOI: 10.1016/s1472-6483(10)60129-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Polo-like kinase 1 (Plk1) has been reported to localize to the spindle midzone during meiosis in mouse oocytes. However, it has not been reported in human oocytes. In this study, the interaction of the meiotic structures and chromosome segregation in mouse and human oocytes were studied by time-lapse differential interference contrast microscopy. Using immunocytochemical studies, the localization of polo-like kinase 1 and its association with microtubules were examined during the extrusion of first and second polar bodies. It was found that Plk1 was localized in the spindle midzone in human oocytes at anaphase I and telophase I. Also, three-dimensional confocal laser microscopy showed that the meiotic spindle midzone contained numerous dot-like particles that were stained by anti-Plk1 antibody. These particles were aligned in the plane of the meiotic midzone in mouse and human oocytes.
Collapse
Affiliation(s)
- Junko Otsuki
- Nagai Clinic, 607-1 Kamihikona, Misato, Saitama, Japan.
| | | | | |
Collapse
|
16
|
Liu L, Keefe DL. Defective cohesin is associated with age-dependent misaligned chromosomes in oocytes. Reprod Biomed Online 2008; 16:103-12. [PMID: 18252055 DOI: 10.1016/s1472-6483(10)60562-7] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aneuploidy often results from chromosome misalignment at metaphases. Oocytes from senescence-accelerated mice (SAM) exhibit increased chromosome misalignment with age, which originates from nuclear factors. This work sought to further characterize the underlying defects of chromosome misalignments. Using immunofluorescence microscopy with specific antibodies, several specific components associated with spindles or chromosomes, including centrosomes, centromeres and cohesin complex were examined. No obvious differences were found in the distribution of centrosome focus at the spindle pole of oocytes from young and aged SAM, regardless of chromosome alignments, although cytoplasmic centrosome foci were significantly reduced in aged SAM (P < 0.0001). Oocytes from both young and aged SAM exhibited centromere-associated protein-E (CENP-E) at centromeres of all chromosomes, including misaligned chromosomes from aged SAM, demonstrating that CENP-E did not contribute to chromosome misalignments. Notably, both meiotic cohesin proteins located between sister chromatids, REC8 (recombinant 8), STAG3 (stromal antigen 3) and SMC1beta, were remarkably reduced in oocytes from aged SAM. Further, degradation of the cohesin was even more obvious in SAM than in hybrid F1 mice with age, which may explain why SAM are vulnerable to aneuploidy. This natural ageing mouse model shows that defective cohesin coincides with increased incidence of chromosome misalignment and precocious separations of sister chromatids.
Collapse
Affiliation(s)
- Lin Liu
- Department of Obstetrics and Gynecology, University of South Florida College of Medicine, Tampa, Florida 33612, USA.
| | | |
Collapse
|
17
|
Blank M, Lerenthal Y, Mittelman L, Shiloh Y. Condensin I recruitment and uneven chromatin condensation precede mitotic cell death in response to DNA damage. J Cell Biol 2006; 174:195-206. [PMID: 16847100 PMCID: PMC2064180 DOI: 10.1083/jcb.200604022] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2006] [Accepted: 06/16/2006] [Indexed: 11/22/2022] Open
Abstract
Mitotic cell death (MCD) is a prominent but poorly defined form of death that stems from aberrant mitosis. One of the early steps in MCD is premature mitosis and uneven chromatin condensation (UCC). The mechanism underlying this phenomenon is currently unknown. In this study, we show that DNA damage in cells with a compromised p53-mediated G2/M checkpoint triggers the unscheduled activation of cyclin-dependent kinase 1 (Cdk1), activation and chromatin loading of the condensin I complex, and UCC followed by the appearance of multimicronucleated cells, which is evidence of MCD. We demonstrate that these processes engage some of the players of normal mitotic chromatin packaging but not those that drive the apoptotic chromatin condensation. Our findings establish a link between the induction of DNA damage and mitotic abnormalities (UCC) through the unscheduled activation of Cdk1 and recruitment of condensin I. These results demonstrate a clear distinction between the mechanisms that drive MCD-associated and apoptosis-related chromatin condensation and provide mechanistic insights and new readouts for a major cell death process in treated tumors.
Collapse
Affiliation(s)
- Michael Blank
- The David and Inez Myers Laboratory for Genetic Research, Department of Molecular Genetics and Biochemistry, and Interdepartmental Core Facility, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | |
Collapse
|
18
|
Assou S, Anahory T, Pantesco V, Le Carrour T, Pellestor F, Klein B, Reyftmann L, Dechaud H, De Vos J, Hamamah S. The human cumulus--oocyte complex gene-expression profile. Hum Reprod 2006; 21:1705-19. [PMID: 16571642 PMCID: PMC2377388 DOI: 10.1093/humrep/del065] [Citation(s) in RCA: 218] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The understanding of the mechanisms regulating human oocyte maturation is still rudimentary. We have identified transcripts differentially expressed between immature and mature oocytes and cumulus cells. METHODS Using oligonucleotide microarrays, genome-wide gene expression was studied in pooled immature and mature oocytes or cumulus cells from patients who underwent IVF. RESULTS In addition to known genes, such as DAZL, BMP15 or GDF9, oocytes up-regulated 1514 genes. We show that PTTG3 and AURKC are respectively the securin and the Aurora kinase preferentially expressed during oocyte meiosis. Strikingly, oocytes overexpressed previously unreported growth factors such as TNFSF13/APRIL, FGF9, FGF14 and IL4 and transcription factors including OTX2, SOX15 and SOX30. Conversely, cumulus cells, in addition to known genes such as LHCGR or BMPR2, overexpressed cell-to-cell signalling genes including TNFSF11/RANKL, numerous complement components, semaphorins (SEMA3A, SEMA6A and SEMA6D) and CD genes such as CD200. We also identified 52 genes progressively increasing during oocyte maturation, including CDC25A and SOCS7. CONCLUSION The identification of genes that were up- and down-regulated during oocyte maturation greatly improves our understanding of oocyte biology and will provide new markers that signal viable and competent oocytes. Furthermore, genes found expressed in cumulus cells are potential markers of granulosa cell tumours.
Collapse
Affiliation(s)
- Said Assou
- IRB, Institut de recherche en biothérapie
CHRU MontpellierUniversité Montpellier IHôpital Saint-Eloi
34000 Montpellier,FR
- Immunopathologie des maladies tumorales et autoimmunes
INSERM : U475IFR76Institut de recherche en biothérapieUniversité Montpellier ICentre de Recherche Inserm
99, Rue Puech Villa
34197 MONTPELLIER CEDEX 5,FR
| | - Tal Anahory
- UFR Médecine
Université Montpellier IMontpellier,FR
| | - Véronique Pantesco
- Immunopathologie des maladies tumorales et autoimmunes
INSERM : U475IFR76Institut de recherche en biothérapieUniversité Montpellier ICentre de Recherche Inserm
99, Rue Puech Villa
34197 MONTPELLIER CEDEX 5,FR
| | - Tanguy Le Carrour
- IRB, Institut de recherche en biothérapie
CHRU MontpellierUniversité Montpellier IHôpital Saint-Eloi
34000 Montpellier,FR
| | - Franck Pellestor
- UFR Médecine
Université Montpellier IMontpellier,FR
- IGH, Institut de génétique humaine
CNRS : UPR1142institut de Génétique humaine
141 Rue de la Cardonille
34396 MONTPELLIER CEDEX 5,FR
| | - Bernard Klein
- IRB, Institut de recherche en biothérapie
CHRU MontpellierUniversité Montpellier IHôpital Saint-Eloi
34000 Montpellier,FR
- Immunopathologie des maladies tumorales et autoimmunes
INSERM : U475IFR76Institut de recherche en biothérapieUniversité Montpellier ICentre de Recherche Inserm
99, Rue Puech Villa
34197 MONTPELLIER CEDEX 5,FR
| | - Lionel Reyftmann
- Service de gynécologie-obstétrique et médecine de la reproduction
CHRU MontpellierHôpital Arnaud de VilleneuveUniversité Montpellier IFR
| | - Hervé Dechaud
- Service de gynécologie-obstétrique et médecine de la reproduction
CHRU MontpellierHôpital Arnaud de VilleneuveUniversité Montpellier IFR
| | - John De Vos
- IRB, Institut de recherche en biothérapie
CHRU MontpellierUniversité Montpellier IHôpital Saint-Eloi
34000 Montpellier,FR
- Immunopathologie des maladies tumorales et autoimmunes
INSERM : U475IFR76Institut de recherche en biothérapieUniversité Montpellier ICentre de Recherche Inserm
99, Rue Puech Villa
34197 MONTPELLIER CEDEX 5,FR
- * Correspondence should be adressed to: John De Vos
| | - Samir Hamamah
- IRB, Institut de recherche en biothérapie
CHRU MontpellierUniversité Montpellier IHôpital Saint-Eloi
34000 Montpellier,FR
- Immunopathologie des maladies tumorales et autoimmunes
INSERM : U475IFR76Institut de recherche en biothérapieUniversité Montpellier ICentre de Recherche Inserm
99, Rue Puech Villa
34197 MONTPELLIER CEDEX 5,FR
- UFR Médecine
Université Montpellier IMontpellier,FR
- * Correspondence should be adressed to: Samir Hamamah
| |
Collapse
|
19
|
Abstract
For more than 30 yr, Xenopus laevis has been the animal of choice for studying the biochemical regulation of the meiotic and early mitotic vertebrate cell cycles. Attracted by its diploid genome, several laboratories have begun using the similar, although evolutionarily distinct, frog Xenopus tropicalis for studies of vertebrate development. Comparisons between the two species indicate that their development is similar in most respects. Both frogs share many advantages, including their amenability to manipulation and their ability to produce large numbers of high-quality oocytes and eggs year round. In addition, X. tropicalis possesses several advantages that, when combined with its potential for genetic studies, makes it an attractive, complementary model for vertebrate developmental biology. In this chapter, we note some of these advantages and describe in detail techniques we have adapted for the study of meiosis in X. tropicalis.
Collapse
Affiliation(s)
- Jean-François L Bodart
- Laboratoire de Biologie du Développement, UPRES EA3, Université des sciences et technologies de Lille, France
| | | |
Collapse
|
20
|
Bodart JFL, Baert FY, Sellier C, Duesbery NS, Flament S, Vilain JP. Differential roles of p39Mos-Xp42Mpk1 cascade proteins on Raf1 phosphorylation and spindle morphogenesis in Xenopus oocytes. Dev Biol 2005; 283:373-83. [PMID: 15913594 DOI: 10.1016/j.ydbio.2005.04.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2004] [Revised: 04/12/2005] [Accepted: 04/15/2005] [Indexed: 10/25/2022]
Abstract
Fully-grown G2-arrested Xenopus oocytes resume meiosis upon hormonal stimulation. Resumption of meiosis is characterized by germinal vesicle breakdown, chromosome condensation, and organization of a bipolar spindle. These cytological events are accompanied by activation of MPF and the p39(Mos)-MEK1-Xp42(Mpk1)-p90(Rsk) pathways. The latter cascade is activated upon p39(Mos) accumulation. Using U0126, a MEK1 inhibitor, and p39(Mos) antisense morpholino and phosphorothioate oligonucleotides, we have investigated the role of the members of the p39(Mos)-MEK1-Xp42(Mpk1)-p90(Rsk) in spindle morphogenesis. First, we have observed at a molecular level that prevention of p39(Mos) accumulation always led to MEK1 phosphorylation defects, even when meiosis was stimulated through the insulin Ras-dependent pathway. Moreover, we have observed that Raf1 phosphorylation that occurs during meiosis resumption was dependent upon the activity of MEK1 or Xp42(Mpk1) but not p90(Rsk). Second, inhibition of either p39(Mos) accumulation or MEK1 inhibition led to the formation of a cytoplasmic aster-like structure that was associated with condensed chromosomes. Spindle morphogenesis rescue experiments using constitutively active Rsk and purified murine Mos protein suggested that p39(Mos) or p90(Rsk) alone failed to promote meiotic spindle organization. Our results indicate that activation of the p39(Mos)-MEK1-Xp42(Mpk1)-p90(Rsk) pathway is required for bipolar organization of the meiotic spindle at the cortex.
Collapse
Affiliation(s)
- J-F L Bodart
- Laboratoire de Biologie du Développement, UPRES EA 1033, Université des Sciences et Technologies de Lille, SN3, Villeneuve d'Ascq, France.
| | | | | | | | | | | |
Collapse
|
21
|
Lan GC, Han D, Wu YG, Han ZB, Ma SF, Liu XY, Chang CL, Tan JH. Effects of duration, concentration, and timing of ionomycin and 6-dimethylaminopurine (6-DMAP) treatment on activation of goat oocytes. Mol Reprod Dev 2005; 71:380-8. [PMID: 15806561 DOI: 10.1002/mrd.20267] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The protocol of ionomycin followed by 6-dimethylaminopurine (6-DMAP) is commonly used for activation of oocytes and reconstituted embryos. Since numerous abnormalities and impaired development were observed when oocytes were activated with 6-DMAP, this protocol needs optimization. Effects of concentration and treatment duration of both drugs on activation and development of goat oocytes were examined in this study. The best oocyte activation (87-95%), assessed by pronuclear formation, was obtained when oocytes matured in vitro for 27 hr were treated with 0.625-20 microM ionomycin for 1 min before 6-hr incubation in 2 mM 6-DMAP. Progressional reduction of time for 6-DMAP-exposure showed that the duration of 6-DMAP treatment can be reduced to 1 hr from the second up to the fourth hour after ionomycin, to produce activation rates greater than 85%. Activation rates of oocytes in vitro matured for 27, 30, and 33 hr were higher (P < 0.05) than that of oocytes matured for 24 hr when treated with ionomycin plus 1-hr (the third hour) 6-DMAP, but a 4-hr incubation in 6-DMAP enhanced activation of the 24-hr oocytes. Goat activated oocytes began pronuclear formation at 3 hr and completed it by 5-hr post ionomycin. An extended incubation in 6-DMAP (a) impaired the development of goat parthenotes, (b) quickened both the release from metaphase arrest and the pronuclear formation, and (c) inhibited the chromosome movement at anaphase II (A-II) and telophase II (T-II), leading to the formation of one pronucleus without extrusion of PB2. In conclusion, duration, concentration, and timing of ionomycin and 6-DMAP treatment had marked effects on goat oocyte activation, and to obtain better activation and development, goat oocytes matured in vitro for 27 hr should be activated by 1 min exposure to 2.5 microM ionomycin followed by 2 mM 6-DMAP treatment for the third hour.
Collapse
Affiliation(s)
- Guo-Cheng Lan
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Ito J, Shimada M. Timing of MAP kinase inactivation effects on emission of polar body in porcine oocytes activated by Ca2+ ionophore. Mol Reprod Dev 2005; 70:64-9. [PMID: 15515060 DOI: 10.1002/mrd.20182] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Artificial activation is required for successful intracytoplasmic sperm injection (ICSI) to induce haploidy pronuclear formation with extraction of second polar body. The present study showed that an additional treatment with Phorbol 12-myristate 13-acetate (PMA) followed by Ca(2+) ionophore treatment improved the rate of pronuclear formation, however, these oocytes had more than two pronuclei because of the suppression of polar body emission. The cultivation with MEK inhibitor U0126 followed by Ca(2+) ionophore also increased the rate of pronuclear formation but suppressed the emission of second polar body. These results suggested that the decrease of MAP kinase activity at early stage of artificial activation, concomitantly with decreasing p34(cdc2) kinase activity, prevented the second polar body extraction. We investigated that the timing of MAP kinase inactivation affected the extraction of the polar body and pronuclear formation rate. The addition of PMA 8 hr after Ca(2+) ionophore treatment induced the delay of MAP kinase inactivation, which resulted in haploidy pronuclear formation with emission of polar body. These results demonstrated for the first time that the delay of MAP kinase inactivation induced by PMA improved pronuclear formation with the extraction of second polar body in porcine oocytes activated by Ca(2+) ionophore. This method can be available for successfully ICSI in low response species of oocyte activation to Ca(2+) ionophore including pig.
Collapse
Affiliation(s)
- Junya Ito
- Department of Applied Animal Science, Graduate School of Biosphere Sciences, Hiroshima University, Higashi-Hiroshima, Japan
| | | |
Collapse
|
23
|
Mailhes JB, Mastromatteo C, Fuseler JW. Transient exposure to the Eg5 kinesin inhibitor monastrol leads to syntelic orientation of chromosomes and aneuploidy in mouse oocytes. Mutat Res 2004; 559:153-67. [PMID: 15066583 DOI: 10.1016/j.mrgentox.2004.01.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2003] [Revised: 01/12/2004] [Accepted: 01/14/2004] [Indexed: 11/17/2022]
Abstract
Aneuploidy may result from abnormalities in the biochemical pathways and cellular organelles associated with chromosome segregation. Monastrol is a reversible, cell-permeable, non-tubulin interacting inhibitor of the mitotic kinesin Eg5 motor protein which is required for assembling and maintaining the mitotic spindle. Monastrol can also impair centrosome separation and induce monoastral spindles in mammalian somatic cells. The ability of monastrol to alter kinesin Eg5 and centrosome activities and spindle geometry may lead to abnormal chromosome segregation. Mouse oocytes were exposed to 0 (control), 15, 30, and 45 microg/ml monastrol in vitro for 6 h during meiosis I and subsequently cultured for 17 h in monastrol-free media prior to cytogenetic analysis of metaphase II oocytes. A subset of oocytes was cultured for 5 h prior to processing cells for meiotic I spindle analysis. Monastrol retarded oocyte maturation by significantly (P < 0.05) decreasing germinal vesicle breakdown and increasing the frequencies of arrested metaphase I oocytes. Also, significant (P < 0.05) increases in the frequencies of monoastral spindles and chromosome displacement from the metaphase plate were found in oocytes during meiosis I. In metaphase II oocytes, monastrol significantly (P < 0.05) increased the frequencies of premature centromere separation and aneuploidy. These findings suggest that abnormal meiotic spindle geometry predisposes oocytes to aneuploidy.
Collapse
Affiliation(s)
- John B Mailhes
- Department of Obstetrics and Gynecology, Louisiana State University Health Sciences Center, P.O. Box 33932, Shreveport, LA 71130, USA.
| | | | | |
Collapse
|
24
|
Eichenlaub-Ritter U, Vogt E, Yin H, Gosden R. Spindles, mitochondria and redox potential in ageing oocytes. Reprod Biomed Online 2004; 8:45-58. [PMID: 14759287 DOI: 10.1016/s1472-6483(10)60497-x] [Citation(s) in RCA: 179] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Studies of human oocytes obtained from women of advanced reproductive age revealed that spindles are frequently aberrant, with chromosomes sometimes failing to align properly at the equator during meiosis I and II. Chromosomal analyses of donated and spare human oocytes and cytogenetic and molecular studies on the origin of trisomies collectively suggest that errors in chromosome segregation during oogenesis increase with advancing maternal age and as the menopause approaches. Disturbances in the fidelity of chromosome segregation, especially at anaphase I, leading to aneuploidy are prime causes of reduced developmental competence of embryos in assisted reproduction, as well as being responsible for the genesis of genetic disease. This review provides an overview of spindle formation and chromosome behaviour in mammalian oocytes. Evidence of a link between abnormal mitochondrial function in oocytes and somatic follicular cells, and finally disturbances in chromosome cohesion and segregation, and cell cycle control in aged mammalian oocytes, are also discussed.
Collapse
Affiliation(s)
- U Eichenlaub-Ritter
- Institute of Gene Technology/Microbiology, Faculty of Biology, University of Bielefeld, D-33501 Bielefeld, Germany.
| | | | | | | |
Collapse
|
25
|
Tunquist BJ, Maller JL. Under arrest: cytostatic factor (CSF)-mediated metaphase arrest in vertebrate eggs. Genes Dev 2003; 17:683-710. [PMID: 12651887 DOI: 10.1101/gad.1071303] [Citation(s) in RCA: 188] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Brian J Tunquist
- The Howard Hughes Medical Institute and Department of Pharmacology, University of Colorado School of Medicine, Denver, CO 80262, USA
| | | |
Collapse
|
26
|
Bodart JFL, Gutierrez DV, Nebreda AR, Buckner BD, Resau JR, Duesbery NS. Characterization of MPF and MAPK activities during meiotic maturation of Xenopus tropicalis oocytes. Dev Biol 2002; 245:348-61. [PMID: 11977986 DOI: 10.1006/dbio.2002.0647] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Resumption of meiosis in oocytes of Xenopus tropicalis required translation but not transcription, and was marked by the appearance of a white spot and a dark ring, coincident with entry into metaphase I and the onset of anaphase I, respectively. Cyclin B(2)/p34(cdc2) activity increased prior to the first meiotic division, declined at the onset of anaphase I, and subsequently increased again. The capacity of egg cytoplasm to induce germinal vesicle breakdown (GVBD) was inhibited by cycloheximide, despite the fact that these oocytes contained cyclin B(2)/p34(cdc2) complexes. However, cycloheximide-treated oocytes underwent GVBD following injection of constitutively active mitogen-activated protein kinase (MAPK) kinase 2 (MEK2), p33(Ringo), or Delta 90 cyclin B. MAPK activity increased just prior to the first meiotic division and remained stable thereafter. Although injection of constitutively active MEK2 induced GVBD, treatment with the MEK inhibitors U0126 or anthrax lethal factor delayed GVBD and prevented spindle formation. Interestingly, the ability of egg cytoplasm to induce GVBD was unaffected by the inhibition of MEK activity. Our results indicate that the synthesis of a novel or short-lived protein(s) which acts in a MEK-independent fashion is required in order for egg cytoplasm to induce GVBD in X. tropicalis oocytes.
Collapse
Affiliation(s)
- Jean-Francois L Bodart
- Van Andel Research Institute, Laboratory of Developmental Cell Biology, Special Program in Analytical, Cellular, and Molecular Microscopy, 333 Bostwick NE, Grand Rapids, Michigan 49503, USA
| | | | | | | | | | | |
Collapse
|
27
|
Maller JL, Schwab MS, Gross SD, Taieb FE, Roberts BT, Tunquist BJ. The mechanism of CSF arrest in vertebrate oocytes. Mol Cell Endocrinol 2002; 187:173-8. [PMID: 11988325 DOI: 10.1016/s0303-7207(01)00695-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A cytoplasmic activity in mature oocytes responsible for second meiotic metaphase arrest was identified over 30 years ago in amphibian oocytes. In Xenopus oocytes cytostatic factor (CSF) activity is initiated by the progesterone-dependent synthesis of Mos, a MAPK kinase kinase that activates the MAPK pathway. CSF arrest is mediated by a sole MAPK target, the protein kinase p90(Rsk). Rsk phosphorylates and activates the Bub1 protein kinase, which may cause metaphase arrest due to inhibition of the anaphase-promoting complex (APC) by a conserved mechanism defined genetically in yeast and mammalian cells. CSF arrest in vertebrate oocytes by p90(Rsk) provides a link between the MAPK pathway and the spindle assembly checkpoint in the cell cycle.
Collapse
Affiliation(s)
- James L Maller
- The Howard Hughes Medical Institute and Department of Pharmacology, University of Colorado School of Medicine, 4200 E. 9th Avenue, Box C236, Denver, CO 80262, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Eaker S, Pyle A, Cobb J, Handel MA. Evidence for meiotic spindle checkpoint from analysis of spermatocytes from Robertsonian-chromosome heterozygous mice. J Cell Sci 2001; 114:2953-65. [PMID: 11686299 DOI: 10.1242/jcs.114.16.2953] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mice heterozygous for Robertsonian centric fusion chromosomal translocations frequently produce aneuploid sperm. In this study RBJ/Dn× C57BL/6J F1 males, heterozygous for four Robertsonian translocations (2N=36), were analyzed to determine effects on germ cells of error during meiosis. Analysis of sperm by three color fluorescence in situ hybridization revealed significantly elevated aneuploidy, thus validating Robertsonian heterozygous mice as a model for production of chromosomally abnormal gametes. Primary spermatocytes from heterozygous males exhibited abnormalities of chromosome pairing in meiotic prophase and metaphase. In spite of prophase abnormalities, the prophase/metaphase transition occurred. However, an increased frequency of cells with misaligned condensed chromosomes was observed. Cytological analysis of both young and adult heterozygous mice revealed increased apoptosis in spermatocytes during meiotic metaphase I. Metaphase spermatocytes with misaligned chromosomes accounted for a significant proportion of the apoptotic spermatocytes, suggesting that a checkpoint process identifies aberrant meioses. Immunofluorescence staining revealed that kinetochores of chromosomes that failed to align on the spindle stained more intensely for kinetochore antigens CENP-E and CENP-F than did aligned chromosomes. Taken together, these observations are consistent with detection of malattached chromosomes by a meiotic spindle checkpoint mechanism that monitors attachment and/or congression of homologous chromosome pairs. However, the relatively high frequency of gametic aneuploidy suggests that the checkpoint mechanism does not efficiently eliminate all germ cells with chromosomal abnormalities.
Collapse
Affiliation(s)
- S Eaker
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, USA
| | | | | | | |
Collapse
|
29
|
Duesbery NS, Resau J, Webb CP, Koochekpour S, Koo HM, Leppla SH, Vande Woude GF. Suppression of ras-mediated transformation and inhibition of tumor growth and angiogenesis by anthrax lethal factor, a proteolytic inhibitor of multiple MEK pathways. Proc Natl Acad Sci U S A 2001; 98:4089-94. [PMID: 11259649 PMCID: PMC31184 DOI: 10.1073/pnas.061031898] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2001] [Indexed: 02/07/2023] Open
Abstract
Lethal factor is a protease, one component of Bacillus anthracis exotoxin, which cleaves many of the mitogen-activated protein kinase kinases (MEKs). Given the importance of MEK signaling in tumorigenesis, we assessed the effects of anthrax lethal toxin (LeTx) on tumor cells. LeTx was very effective in inhibiting mitogen-activated protein kinase activation in V12 H-ras-transformed NIH 3T3 cells. In vitro, treatment of transformed cells with LeTx caused them to revert to a nontransformed morphology, and inhibited their abilities to form colonies in soft agar and to invade Matrigel without markedly affecting cell proliferation. In vivo, LeTx inhibited growth of ras-transformed cells implanted in athymic nude mice (in some cases causing tumor regression) at concentrations that caused no apparent animal toxicity. Unexpectedly, LeTx also greatly decreased tumor neovascularization. These results demonstrate that LeTx potently inhibits ras-mediated tumor growth and is a potential antitumor therapeutic.
Collapse
Affiliation(s)
- N S Duesbery
- Van Andel Research Institute, 333 Bostwick Avenue, NE, Grand Rapids, MI 49503, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
While animal eggs await fertilization, their cell cycle needs to be halted. The molecule responsible for this arrest--the cytostatic factor--was first described in 1971. But its identity was not revealed until 1989, and even now questions remain about this elusive factor.
Collapse
Affiliation(s)
- Y Masui
- Department of Zoology, University of Toronto, 25 Harbord Street, Toronto, Ontario, Canada M5S 3G5.
| |
Collapse
|
31
|
Yao X, Abrieu A, Zheng Y, Sullivan KF, Cleveland DW. CENP-E forms a link between attachment of spindle microtubules to kinetochores and the mitotic checkpoint. Nat Cell Biol 2000; 2:484-91. [PMID: 10934468 DOI: 10.1038/35019518] [Citation(s) in RCA: 292] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Here we show that suppression of synthesis of the microtubule motor CENP-E (centromere-associated protein E), a component of the kinetochore corona fibres of mammalian centromeres, yields chromosomes that are chronically mono-orientated, with spindles that are flattened along the plane of the substrate. Despite apparently normal microtubule numbers and the continued presence at kinetochores of other microtubule motors, spindle poles fragment in the absence of CENP-E, which implicates this protein in delivery of components from kinetochores to poles. CENP-E represents a link between attachment of spindle microtubules and the mitotic checkpoint signalling cascade, as depletion of this motor leads to profound checkpoint activation, whereas immunoprecipitation reveals a nearly stoichiometric association of CENP-E with the checkpoint kinase BubR1 during mitosis.
Collapse
Affiliation(s)
- X Yao
- Department of Physiology, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | | | | | | | |
Collapse
|
32
|
Lee J, Miyano T, Dai Y, Wooding P, Yen TJ, Moor RM. Specific regulation of CENP-E and kinetochores during meiosis I/meiosis II transition in pig oocytes. Mol Reprod Dev 2000; 56:51-62. [PMID: 10737967 DOI: 10.1002/(sici)1098-2795(200005)56:1<51::aid-mrd7>3.0.co;2-n] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
To understand the mechanisms which regulate meiosis-specific cell cycle and chromosome distribution in mammalian oocytes, the level and the localization of CENP-E and the kinetochore number and direction on a half bivalent were examined during pig oocyte maturation. CENP-E is a kinetochore motor protein whose intracellular level and localization are strictly regulated in the somatic cell cycle. The localizations of CENP-E on meiotic chromosomes from diakinesis stage to anaphase I and at the spindle midzone at telophase I were shown by immunofluorescent confocal microscopy to be similar to those in somatic cells of pig and other species. Further, ultrastructural analysis revealed the presence of CENP-E on fibrous corona and outer plate of kinetochores of the meiotic chromosomes. However, unlike mitosis, CENP-E staining was continuously detected either at the spindle midzone or on the kinetochores of segregated chromosomes during the first polar body emission. Consistent with this, immunoblot analysis revealed that CENP-E level remained high during meiosis I/meiosis II (MI/MII) transition and that some of CENP-E survived through the transition even in cycloheximide-treated oocytes in which cyclin B1 was completely degraded. Furthermore, examinations of CENP-E signals in confocal microscopy and kinetochores in electron microscopy in MI and MII oocytes provide the cytological evidence in mammalian oocytes which suggests that each sister chromatid in a pair has its own kinetochore which localizes side-by-side so that two sister chromatids on a half bivalent are oriented toward and connected to the same pole in MI.
Collapse
Affiliation(s)
- J Lee
- Laboratory of Protein Function, The Babraham Institute, Cambridge, United Kingdom.
| | | | | | | | | | | |
Collapse
|
33
|
Goldstein LS, Philp AV. The road less traveled: emerging principles of kinesin motor utilization. Annu Rev Cell Dev Biol 1999; 15:141-83. [PMID: 10611960 DOI: 10.1146/annurev.cellbio.15.1.141] [Citation(s) in RCA: 196] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Proteins of the kinesin superfamily utilize a conserved catalytic motor domain to generate movements in a wide variety of cellular processes. In this review, we discuss the rapid expansion in our understanding of how eukaryotic cells take advantage of these proteins to generate force and movement in diverse functional contexts. We summarize several recent examples revealing that the simplest view of a kinesin motor protein binding to and translocating a cargo along a microtubule track is inadequate. In fact, this paradigm captures only a small subset of the many ways in which cells harness force production of the generation of intracellular movements and functions. We also highlight several situations where the catalytic kinesin motor domain may not be used to generate movement, but instead may be used in other biochemical and functional contexts. Finally, we review some recent ideas about kinesin motor regulation, redundancy, and cargo attachment strategies.
Collapse
Affiliation(s)
- L S Goldstein
- Howard Hughes Medical Institute, Department of Pharmacology, University of California San Diego, La Jolla 92093-0683, USA.
| | | |
Collapse
|
34
|
Matthies HJ, Messina LG, Namba R, Greer KJ, Walker M, Hawley RS. Mutations in the alpha-tubulin 67C gene specifically impair achiasmate segregation in Drosophila melanogaster. J Cell Biol 1999; 147:1137-44. [PMID: 10601329 PMCID: PMC2168102 DOI: 10.1083/jcb.147.6.1137] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Drosophila melanogaster oocytes heterozygous for mutations in the alpha-tubulin 67C gene (alphatub67C) display defects in centromere positioning during prometaphase of meiosis I. The centromeres do not migrate to the poleward edges of the chromatin mass, and the chromatin fails to stretch during spindle lengthening. These results suggest that the poleward forces acting at the kinetochore are compromised in the alphatub67C mutants. Genetic studies demonstrate that these mutations also strongly and specifically decrease the fidelity of achiasmate chromosome segregation. Proper centromere orientation, chromatin elongation, and faithful segregation can all be restored by a decrease in the amount of the Nod chromokinesin. These results suggest that the accurate segregation of achiasmate chromosomes requires the proper balancing of forces acting on the chromosomes during prometaphase.
Collapse
Affiliation(s)
- Heinrich J.G. Matthies
- Department of Genetics, Section of Molecular and Cellular Biology, University of California at Davis, Davis, California 95616
| | - Lisa G. Messina
- Department of Genetics, Section of Molecular and Cellular Biology, University of California at Davis, Davis, California 95616
| | - Ruria Namba
- Department of Genetics, Section of Molecular and Cellular Biology, University of California at Davis, Davis, California 95616
| | - Kimberly J. Greer
- Department of Genetics, Section of Molecular and Cellular Biology, University of California at Davis, Davis, California 95616
| | - M.Y. Walker
- Department of Genetics, Section of Molecular and Cellular Biology, University of California at Davis, Davis, California 95616
| | - R. Scott Hawley
- Department of Genetics, Section of Molecular and Cellular Biology, University of California at Davis, Davis, California 95616
| |
Collapse
|
35
|
Hirao Y, Eppig JJ. Analysis of the mechanism(s) of metaphase I-arrest in strain LT mouse oocytes: delay in the acquisition of competence to undergo the metaphase I/anaphase transition. Mol Reprod Dev 1999; 54:311-8. [PMID: 10497353 DOI: 10.1002/(sici)1098-2795(199911)54:3<311::aid-mrd12>3.0.co;2-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Fully grown oocytes of most laboratory mice progress without interruption from the germinal vesicle (GV) stage to metaphase II, where meiosis is arrested until fertilization. In contrast, many oocytes of strain LT mice arrest precociously at metaphase I and often undergo subsequent spontaneous parthenogenetic activation. Cytostatic factor (CSF), which prevents the degradation of cyclin B and maintains high maturation-promoting factor (MPF) activity, is required for maintenance of metaphase I-arrest in LT oocytes, similar to its requirement for maintaining metaphase II-arrest in normal oocytes. However, CSF does not instigate metaphase I-arrest since a temporary metaphase I-arrest occurs in MOS-null LT oocytes. This paper addresses the mechanism(s) that may instigate metaphase I-arrest and tests the hypothesis that there may be one or more defects in LT oocytes that delay their acquisition of competence to trigger the cascade of processes that normally drive entry into and progression through anaphase I. To test this hypothesis, MPF activity was artificially abrogated by treating oocytes with a general protein kinase inhibitor, 6-DMAP, at various times during the progression of meiosis I. This allowed a comparison of the time at which LT and normal oocytes become competent to undergo the metaphase I/anaphase transition even if oocytes were arrested at metaphase I when 6-DMAP-treatment was begun. There were no differences between LT and control oocytes in the kinetics of MPF suppression by 6-DMAP. However, it was found that LT oocytes do not acquire competence to undergo the metaphase I/anaphase transition in response to 6-DMAP until 50-60 min after normal oocytes. A similar delay was observed in strain CX8-4 oocytes, which also have a high incidence of metaphase I-arrest, but not in strain CX8-11 oocytes, which exhibit a low incidence of metaphase I-arrest. MOS-null LT oocytes also exhibit a delay in acquisition of competence to undergo the metaphase I/anaphase transition. Thus, a delay in competence to undergo the metaphase I/anaphase transition in response to 6-DMAP-treatment correlates with metaphase I-arrest. It is therefore hypothesized that the observed delay in acquisition of competence to enter anaphase I may instigate the sustained metaphase I-arrest in LT oocytes by allowing CSF activity to rise to a level that prevents cyclin B degradation and maintains high MPF activity before anaphase can be initiated by normal triggering mechanisms.
Collapse
Affiliation(s)
- Y Hirao
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | |
Collapse
|
36
|
Liu L, Yang X. Interplay of maturation-promoting factor and mitogen-activated protein kinase inactivation during metaphase-to-interphase transition of activated bovine oocytes. Biol Reprod 1999; 61:1-7. [PMID: 10377024 DOI: 10.1095/biolreprod61.1.1] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The objective of the present study was to examine the activity changes in histone H1 kinase (also known as maturation-promoting factor [MPF]) and mitogen-activated protein kinase (MAPK) and their constituent proteins in in vitro-matured bovine oocytes after in vitro fertilization (IVF) or after parthenogenetic activation induced by calcium ionophore A23187 alone or by the ionophore followed by either 6-dimethylaminopurine (6-DMAP) or cycloheximide (CHX). Inactivation of both H1 kinase and MAPK occurred after both A23187+6-DMAP treatment and IVF; inactivation of H1 kinase preceded inactivation of MAPK. However, MAPK was inactivated much earlier in 6-DMAP-treated oocytes. Further analysis of constituent cell cycle proteins of these kinases by Western blot showed that A23187 alone could not induce changes in cdc2, cdc25, or ERK2 but induced reduction of cyclin B1. IVF and A23187+CHX induced similar changes: cyclin B1 was destroyed shortly after activation followed by accumulation of cyclin B1, phosphorylation of cdc2, and dephosphorylation of ERK2 at pronuclear formation 15 h after activation. No change in cdc25 was observed at this time. In contrast, A23187+6-DMAP treatment resulted in earlier phosphorylation of cdc2 and dephosphorylation of ERK2 at 4 h after treatment when the pronucleus formed. Moreover, accumulation of both cdc25 and cyclin B1 was detected at 15 h. Microinjection of ERK2 antibody into A23187-treated oocytes resulted in pronuclear formation. In conclusion, activation of bovine oocytes with 6-DMAP led to earlier inactivation of MAPK, while CHX induced inactivation of MAPK parallel to that following sperm-induced oocyte activation. Destruction of cyclin B is responsible for inactivation of MPF, while phosphorylation of cdc2 is likely responsible for maintaining its low activity. Inactivation of MAPK is closely associated with pronuclear development regardless of the activation protocol used.
Collapse
Affiliation(s)
- L Liu
- Department of Animal Science, University of Connecticut, Storrs, Connecticut 06269, USA
| | | |
Collapse
|
37
|
Perry AC, Wakayama T, Yanagimachi R. A novel trans-complementation assay suggests full mammalian oocyte activation is coordinately initiated by multiple, submembrane sperm components. Biol Reprod 1999; 60:747-55. [PMID: 10026126 DOI: 10.1095/biolreprod60.3.747] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
To initiate normal embryonic development, an egg must receive a signal to become activated at fertilization. We here report that the ability of demembranated sperm heads to activate is abolished after incubation over the range 20-44 degreesC and is sensitive to reducing agents. On the basis of this observation, we have developed a microinjection-based, trans-complementation assay in order to dissect the heat-inactivated sperm-borne oocyte-activating factor(s) (SOAF). We demonstrate that the failure of heat-inactivated sperm heads to activate an egg is rescued by coinjection with dithiothreitol-solubilized SOAF from demembranated sperm heads. The solubilized SOAF (SOAFs) is trypsin sensitive and is liberated from demembranated heads in a temperature-dependent manner that inversely correlates with the ability of sperm heads to activate. This argues that SOAFs is a proteinaceous molecular species required to initiate activation. Injection of oocytes with mouse or hamster sperm cytosolic factors, but not SOAFs alone, induced resumption of meiosis, further suggesting that these cytosolic factors and SOAF are distinct. Collectively, these data strongly suggest that full mammalian oocyte activation is initiated by the coordinated action of one or more heat-sensitive protein constituents of the perinuclear matrix and at least one heat-stable submembrane component.
Collapse
Affiliation(s)
- A C Perry
- Department of Anatomy and Reproductive Biology, University of Hawaii School of Medicine, Honolulu, Hawaii 96822, USA
| | | | | |
Collapse
|
38
|
Schumacher JM, Golden A, Donovan PJ. AIR-2: An Aurora/Ipl1-related protein kinase associated with chromosomes and midbody microtubules is required for polar body extrusion and cytokinesis in Caenorhabditis elegans embryos. J Cell Biol 1998; 143:1635-46. [PMID: 9852156 PMCID: PMC2132979 DOI: 10.1083/jcb.143.6.1635] [Citation(s) in RCA: 251] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
An emerging family of kinases related to the Drosophila Aurora and budding yeast Ipl1 proteins has been implicated in chromosome segregation and mitotic spindle formation in a number of organisms. Unlike other Aurora/Ipl1-related kinases, the Caenorhabditis elegans orthologue, AIR-2, is associated with meiotic and mitotic chromosomes. AIR-2 is initially localized to the chromosomes of the most mature prophase I-arrested oocyte residing next to the spermatheca. This localization is dependent on the presence of sperm in the spermatheca. After fertilization, AIR-2 remains associated with chromosomes during each meiotic division. However, during both meiotic anaphases, AIR-2 is present between the separating chromosomes. AIR-2 also remains associated with both extruded polar bodies. In the embryo, AIR-2 is found on metaphase chromosomes, moves to midbody microtubules at anaphase, and then persists at the cytokinesis remnant. Disruption of AIR-2 expression by RNA- mediated interference produces entire broods of one-cell embryos that have executed multiple cell cycles in the complete absence of cytokinesis. The embryos accumulate large amounts of DNA and microtubule asters. Polar bodies are not extruded, but remain in the embryo where they continue to replicate. The cytokinesis defect appears to be late in the cell cycle because transient cleavage furrows initiate at the proper location, but regress before the division is complete. Additionally, staining with a marker of midbody microtubules revealed that at least some of the components of the midbody are not well localized in the absence of AIR-2 activity. Our results suggest that during each meiotic and mitotic division, AIR-2 may coordinate the congression of metaphase chromosomes with the subsequent events of polar body extrusion and cytokinesis.
Collapse
Affiliation(s)
- J M Schumacher
- Cell Biology of Development and Differentiation Group, ABL-Basic Research Program, NCI-Frederick Cancer Research and Development Center, Frederick, Maryland 21702, USA
| | | | | |
Collapse
|
39
|
Abstract
OBJECTIVES Correlations between parental age, aneuploidy in germ cells and recent findings on aetiological factors in mammalian trisomy formation are reviewed. METHODS Data from observations in human oocytes, molecular studies on the origin of extra chromosomes in trisomies, experiments in a mouse model system, and transgenic approaches are shown. RESULTS Errors in chromosome segregation are most frequent in meiosis I of oogenesis in mammals and predominantly predispose specific chromosomes and susceptible chiasmate configurations to maternal age-related nondisjunction. Studies on spindle structure, cell cycle and chromosome behaviour in oocytes of the CBA/Ca mouse used as a model for the maternal age-effect suggest that hormonal homeostasis and size of the follicle pool influence the quality, maturation competence and spindle size of the mammalian oocyte. Predisposition to errors in chromosome segregation are critically dependent on altered cell cycles. Compromised protein synthesis and mitochondrial function affect maturation kinetics and spindle formation, and cause untimely segregation of chromosomes (predivision), mimicking an aged phenotype. CONCLUSIONS Altered cell cycles and untimely resolution of chiasmata but also nondisjunction of late segregating homologues caused by asynchrony in cytoplasmic and nuclear maturation appear to be causal to errors in chromosome segregation with advanced maternal age. Oocytes appear to lack checkpoints guarding against untimely chromosome segregation. Genes and exposures affecting pool size, hormonal homeostasis and interactions between oocytes and their somatic compartment and thus quality of follicles and oocytes have the potential to critically influence chromosome distribution in female meiosis and affect fertility in humans and other mammals.
Collapse
|
40
|
Starr DA, Williams BC, Hays TS, Goldberg ML. ZW10 helps recruit dynactin and dynein to the kinetochore. J Cell Biol 1998; 142:763-74. [PMID: 9700164 PMCID: PMC2148168 DOI: 10.1083/jcb.142.3.763] [Citation(s) in RCA: 208] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/1998] [Revised: 07/01/1998] [Indexed: 02/08/2023] Open
Abstract
Mutations in the Drosophila melanogaster zw10 gene, which encodes a conserved, essential kinetochore component, abolish the ability of dynein to localize to kinetochores. Several similarities between the behavior of ZW10 protein and dynein further support a role for ZW10 in the recruitment of dynein to the kinetochore: (a) in response to bipolar tension across the chromosomes, both proteins mostly leave the kinetochore at metaphase, when their association with the spindle becomes apparent; (b) ZW10 and dynein both bind to functional neocentromeres of structurally acentric minichromosomes; and (c) the localization of both ZW10 and dynein to the kinetochore is abolished in cells mutant for the gene rough deal. ZW10's role in the recruitment of dynein to the kinetochore is likely to be reasonably direct, because dynamitin, the p50 subunit of the dynactin complex, interacts with ZW10 in a yeast two-hybrid screen. Since in zw10 mutants no defects in chromosome behavior are observed before anaphase onset, our results suggest that dynein at the kinetochore is essential for neither microtubule capture nor congression to the metaphase plate. Instead, dynein's role at the kinetochore is more likely to be involved in the coordination of chromosome separation and/or poleward movement at anaphase onset.
Collapse
Affiliation(s)
- D A Starr
- Section of Genetics and Development, Cornell University, Ithaca, New York 14853-2703, USA
| | | | | | | |
Collapse
|
41
|
Duesbery NS, Webb CP, Leppla SH, Gordon VM, Klimpel KR, Copeland TD, Ahn NG, Oskarsson MK, Fukasawa K, Paull KD, Vande Woude GF. Proteolytic inactivation of MAP-kinase-kinase by anthrax lethal factor. Science 1998; 280:734-7. [PMID: 9563949 DOI: 10.1126/science.280.5364.734] [Citation(s) in RCA: 786] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Anthrax lethal toxin, produced by the bacterium Bacillus anthracis, is the major cause of death in animals infected with anthrax. One component of this toxin, lethal factor (LF), is suspected to be a metalloprotease, but no physiological substrates have been identified. Here it is shown that LF is a protease that cleaves the amino terminus of mitogen-activated protein kinase kinases 1 and 2 (MAPKK1 and MAPKK2) and that this cleavage inactivates MAPKK1 and inhibits the MAPK signal transduction pathway. The identification of a cleavage site for LF may facilitate the development of LF inhibitors.
Collapse
Affiliation(s)
- N S Duesbery
- Advanced BioScience Laboratories-Basic Research Program, National Cancer Institute-Frederick Cancer Research and Development Center, Post Office Box B, Frederick, MD 21702
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Wood KW, Sakowicz R, Goldstein LS, Cleveland DW. CENP-E is a plus end-directed kinetochore motor required for metaphase chromosome alignment. Cell 1997; 91:357-66. [PMID: 9363944 DOI: 10.1016/s0092-8674(00)80419-5] [Citation(s) in RCA: 332] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Mitosis requires dynamic attachment of chromosomes to spindle microtubules. This interaction is mediated largely by kinetochores. During prometaphase, forces exerted at kinetochores, in combination with polar ejection forces, drive congression of chromosomes to the metaphase plate. A major question has been whether kinetochore-associated microtubule motors play an important role in congression. Using immunodepletion from and antibody addition to Xenopus egg extracts, we show that the kinetochore-associated kinesin-like motor protein CENP-E is essential for positioning chromosomes at the metaphase plate. We further demonstrate that CENP-E powers movement toward microtubule plus ends in vitro. These findings support a model in which CENP-E functions in congression to tether kinetochores to dynamic microtubule plus ends.
Collapse
Affiliation(s)
- K W Wood
- Laboratory of Cell Biology, Ludwig Institute for Cancer Research, University of California at San Diego, La Jolla 92093-0660, USA
| | | | | | | |
Collapse
|
43
|
Yao X, Anderson KL, Cleveland DW. The microtubule-dependent motor centromere-associated protein E (CENP-E) is an integral component of kinetochore corona fibers that link centromeres to spindle microtubules. J Cell Biol 1997; 139:435-47. [PMID: 9334346 PMCID: PMC2139792 DOI: 10.1083/jcb.139.2.435] [Citation(s) in RCA: 182] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/1997] [Revised: 07/31/1997] [Indexed: 02/05/2023] Open
Abstract
Centromere-associated protein E (CENP-E) is a kinesin-related microtubule motor protein that is essential for chromosome congression during mitosis. Using immunoelectron microscopy, CENP-E is shown to be an integral component of the kinetochore corona fibers that tether centromeres to the spindle. Immediately upon nuclear envelope fragmentation, an associated plus end motor trafficks cytoplasmic CENP-E toward chromosomes along astral microtubules that enter the nuclear volume. Before or concurrently with initial lateral attachment of spindle microtubules, CENP-E targets to the outermost region of the developing kinetochores. After stable attachment, throughout chromosome congression, at metaphase, and throughout anaphase A, CENP-E is a constituent of the corona fibers, extending at least 50 nm away from the kinetochore outer plate and intertwining with spindle microtubules. In congressing chromosomes, CENP-E is preferentially associated with (or accessible at) the stretched, leading kinetochore known to provide the primary power for chromosome movement. Taken together, this evidence strongly supports a model in which CENP-E functions in congression to tether kinetochores to the disassembling microtubule plus ends.
Collapse
Affiliation(s)
- X Yao
- Laboratory of Cell Biology, Ludwig Institute for Cancer Research, School of Medicine, University of California, La Jolla, CA 92093-0660, USA
| | | | | |
Collapse
|