Volume 67, Issue 6, Pages e2 (September 2017)

Slides:



Advertisements
Similar presentations
Volume 14, Issue 4, Pages (May 2004)
Advertisements

Volume 15, Issue 1, Pages (July 2014)
Mira Park, Ph. D. , Dae-Shik Suh, M. D. , Kangseok Lee, Ph. D
Volume 8, Issue 3, Pages (March 2005)
Volume 21, Issue 6, Pages (December 2011)
Volume 53, Issue 4, Pages (February 2014)
Volume 11, Issue 11, Pages (June 2015)
L. L. Sun, Z. Y. Sun, P. Zhang, X. W. Zhai, J. Tang, Q. J. Pan, Q. H
Prevention of chemotherapy-induced ovarian damage
Volume 48, Issue 3, Pages (November 2012)
VEGF Gene Delivery to Muscle
Telomere-Driven Tetraploidization Occurs in Human Cells Undergoing Crisis and Promotes Transformation of Mouse Cells  Teresa Davoli, Titia de Lange  Cancer.
Volume 14, Issue 2, Pages (February 2008)
Volume 26, Issue 13, Pages (July 2016)
Quantitative Live Cell Imaging Reveals a Gradual Shift between DNA Repair Mechanisms and a Maximal Use of HR in Mid S Phase  Ketki Karanam, Ran Kafri,
Richard C. Centore, Stephanie A. Yazinski, Alice Tse, Lee Zou 
Volume 26, Issue 9, Pages (May 2016)
Volume 20, Issue 3, Pages e4 (March 2017)
The Mouse Spo11 Gene Is Required for Meiotic Chromosome Synapsis
Oocytes Progress beyond Prophase in the Presence of DNA Damage
Nikhila S. Tanneti, Kathryn Landy, Eric F. Joyce, Kim S. McKim 
Shi Ying Jin, Ph. D. , Lei Lei, Ph. D. , Ariella Shikanov, Ph. D
Volume 23, Issue 2, Pages (August 2012)
Volume 14, Issue 1, Pages (January 2004)
Wenqian Hu, Bingbing Yuan, Harvey F. Lodish  Developmental Cell 
Volume 37, Issue 1, Pages (July 2012)
Role of the Notch Ligand Delta1 in Embryonic and Adult Mouse Epidermis
Ryang Hwa Lee, Nara Yoon, John C. Reneau, Darwin J. Prockop 
Volume 25, Issue 1, Pages (January 2017)
A Super-Assembly of Whi3 Encodes Memory of Deceptive Encounters by Single Cells during Yeast Courtship  Fabrice Caudron, Yves Barral  Cell  Volume 155,
Volume 21, Issue 2, Pages (January 2006)
Volume 54, Issue 2, Pages (April 2007)
Large Cytoplasm Is Linked to the Error-Prone Nature of Oocytes
Jungmook Lyu, Vicky Yamamoto, Wange Lu  Developmental Cell 
Volume 130, Issue 5, Pages (September 2007)
The Shortest Telomere, Not Average Telomere Length, Is Critical for Cell Viability and Chromosome Stability  Michael T Hemann, Margaret A Strong, Ling-Yang.
Volume 17, Issue 4, Pages (October 2016)
Leah Vardy, Terry L. Orr-Weaver  Developmental Cell 
Volume 5, Issue 3, Pages (March 2000)
Sex, Not Genotype, Determines Recombination Levels in Mice
Volume 21, Issue 6, Pages (December 2011)
Volume 34, Issue 1, Pages (July 2015)
Volume 48, Issue 3, Pages (November 2012)
Cell-Nonautonomous Regulation of C. elegans Germ Cell Death by kri-1
Volume 35, Issue 4, Pages (August 2009)
Volume 11, Issue 11, Pages (June 2015)
Applying “Gold Standards” to In-Vitro-Derived Germ Cells
Polo Kinases Establish Links between Meiotic Chromosomes and Cytoskeletal Forces Essential for Homolog Pairing  Sara Labella, Alexander Woglar, Verena.
Telomere-Driven Tetraploidization Occurs in Human Cells Undergoing Crisis and Promotes Transformation of Mouse Cells  Teresa Davoli, Titia de Lange  Cancer.
Retinoic Acid Antagonizes Testis Development in Mice
Volume 31, Issue 1, Pages (July 2008)
Volume 39, Issue 1, Pages (July 2010)
A Role for Retrotransposon LINE-1 in Fetal Oocyte Attrition in Mice
Volume 47, Issue 4, Pages (August 2012)
Florence Couteau, Monique Zetka  Developmental Cell 
Volume 16, Issue 2, Pages (February 2009)
Volume 18, Issue 6, Pages (December 2010)
Volume 20, Issue 23, Pages (December 2010)
Volume 19, Issue 9, Pages (May 2017)
FGF9 Suppresses Meiosis and Promotes Male Germ Cell Fate in Mice
Volume 24, Issue 12, Pages (September 2018)
The Meiosis-Specific Hop2 Protein of S
Masakazu Hashimoto, Hiroshi Sasaki
Volume 26, Issue 5, Pages (March 2016)
Multiple Rad5 Activities Mediate Sister Chromatid Recombination to Bypass DNA Damage at Stalled Replication Forks  Eugen C. Minca, David Kowalski  Molecular.
Volume 1, Issue 5, Pages (April 1998)
Volume 39, Issue 6, Pages (September 2010)
CRISPR Immunological Memory Requires a Host Factor for Specificity
Patterns of Meiotic Recombination in Human Fetal Oocytes
Meiotic sex chromosome inactivation
Presentation transcript:

Volume 67, Issue 6, Pages 1026-1036.e2 (September 2017) The DNA Damage Checkpoint Eliminates Mouse Oocytes with Chromosome Synapsis Failure  Vera D. Rinaldi, Ewelina Bolcun-Filas, Hiroshi Kogo, Hiroki Kurahashi, John C. Schimenti  Molecular Cell  Volume 67, Issue 6, Pages 1026-1036.e2 (September 2017) DOI: 10.1016/j.molcel.2017.07.027 Copyright © 2017 Elsevier Inc. Terms and Conditions

Molecular Cell 2017 67, 1026-1036.e2DOI: (10.1016/j.molcel.2017.07.027) Copyright © 2017 Elsevier Inc. Terms and Conditions

Figure 1 CHK2 Is Required for Efficient Elimination of Asynaptic Spo11−/− Mouse Oocytes (A) H&E-stained histological sections of 8-week-old ovaries. Black arrowheads indicate antral follicles. The presence of corpora lutea (CL) is indicative of prior rounds of ovulation. Shown at the bottom are higher-magnification images of an ovarian cortical region, where primordial follicles reside. Yellow arrows and asterisks indicate primordial and primary follicles, respectively. (B) Follicle counts from ovaries of the indicated genotypes at 3 and 8 weeks pp, respectively. Each data point is from a single ovary, each from a different animal. Total, all follicle types. Horizontal hashes denote mean and SD. Littermate controls included animals with the following genotypes: Spo11+/+Chk2+/+, Spo11+/− Chk2+/−, and Spo11+/+Chk2+/−. ˆ, the values obtained for the 3-week follicles/ovaries counts are not comparable with the 8-week ones (see STAR Methods). ∗p = 0.005–0.05, ∗∗p = 0.001–0.005 and ∗∗∗ p ≤ 0.001 derived from a non-parametric, one-way ANOVA test (Kruskal-Wallis). Molecular Cell 2017 67, 1026-1036.e2DOI: (10.1016/j.molcel.2017.07.027) Copyright © 2017 Elsevier Inc. Terms and Conditions

Figure 2 Synapsis-Competent Trip13Gt/Gt Oocytes Are Eliminated in a HORMAD2-Dependent Manner (A) H&E-stained histological sections of 8-week-old ovaries of the indicated genotypes. Black arrowheads indicate antral follicles. Shown at the bottom are higher-magnification images of cortical regions of ovaries. Yellow arrows and asterisks indicate primordial and primary follicles, respectively. (B) Follicle quantification of 8-week-old ovaries. Each data point is from a single ovary, each from a different animal. Total, all follicle types. Horizontal hashes denote mean and SD. The statistic used was a Kruskal-Wallis test. ∗p = 0.002. (C) Graphed are mean litter sizes. n ≥ 3 females tested for fertility per genotypic group. Control matings were between mice with the genotypes Trip13Gt/+ and Trip13Gt/+ Hormad2+/−. Error bars represent SD, and ∗∗p ≤ 0.005, derived from the Kruskal-Wallis test. Molecular Cell 2017 67, 1026-1036.e2DOI: (10.1016/j.molcel.2017.07.027) Copyright © 2017 Elsevier Inc. Terms and Conditions

Figure 3 HORMAD2 and CHK2 Are Not in the Same Checkpoint Pathway (A) H&E-stained histological sections of cortical regions of 8-week-old mutant mouse ovaries, where primordial follicles are concentrated. Histologies of whole ovaries of these genotypes are presented in Figure S2. Primordial follicles, which constitute the oocyte reserve, are indicated by yellow arrows, and a primary follicle by an asterisk. Residual Dmc1−/− ovaries are not represented because they are completely devoid of oocytes (Pittman et al., 1998). (B) Follicle counts from ovaries of the indicated genotypes at 8 weeks of age. Total, all types of follicles. Data points represent follicle counts derived from one ovary, each ovary originating from a different animal. ∗p ≤ 0.05 (Kruskal-Wallis test). Molecular Cell 2017 67, 1026-1036.e2DOI: (10.1016/j.molcel.2017.07.027) Copyright © 2017 Elsevier Inc. Terms and Conditions

Figure 4 Depletion of HORMAD2 Accelerates DSB Repair during Early Stages of Meiotic Prophase I (A) Representative images of meiotic chromosome spreads from oocytes at different substages of meiotic prophase I, probed with antibodies for SYCP3 (SC axis protein) and the DSB marker RAD51. Oocytes were isolated from female embryos ranging from 15.5 DPC to newborns. See Figure S1 for HORMAD2 localization in meiotic mutants. (B) Numbers of RAD51 foci in the specified meiotic prophase I substage of the indicated mutants. Only RAD51 foci present on SYCP3-stained axes were scored. Each data point represents one cell. In each genotypic group, at each stage, the counts are derived from at least three animals. Horizontal hashes in summary statistic plots denote mean and SD. Values of the mixed-model calculation can be found in Table S1. Colors correspond to genotypes. Asterisks indicate statistical significant differences between groups in terms of the least square means of RAD51 foci: ∗∗∗p ≤ 0.001, ∗∗p ≤ 0.005, ∗p ≤ 0.05 (Tukey HSD). See Table S1 for associated raw data and statistical calculations. Molecular Cell 2017 67, 1026-1036.e2DOI: (10.1016/j.molcel.2017.07.027) Copyright © 2017 Elsevier Inc. Terms and Conditions

Figure 5 Depletion of HORMAD2 Accelerates Repair of Induced DSBs in Oocytes (A) Immunolabeling of surface-spread chromosomes from oocytes after exposure to ionizing radiation (IR). Fetal ovaries were collected at 15.5 DPC, cultured for 24 hr, exposed to 2 Gy of IR, and then cultured for an additional 4-8 hr. Shown are those recovered 8 hr after IR. See Figure S4 for single Hormad2−/− single-mutant results. (B) Quantification of RAD51 foci. Each data point represents one oocyte. The graphs include mean and SD and are color-coded according to genotypic group. The 4- and 8-hr unirradiated samples were combined. Data were derived from at least two different animals per condition. See Table S2 for associated raw data and statistical calculations. Molecular Cell 2017 67, 1026-1036.e2DOI: (10.1016/j.molcel.2017.07.027) Copyright © 2017 Elsevier Inc. Terms and Conditions

Figure 6 DNA Damage Threshold Required to Trigger Oocyte Death and Evidence for HORMAD-Mediated Inhibition of IS Repair (A) Linear regression for conversion of radiation dosages to RAD51 focus counts. Meiotic surface spreads were made from WT neonatal ovaries 2.5 hr after IR. Plotted are means with SD. Each IR dose has focus counts from ∼25 pachytene oocytes derived from a total of 18 animals. See Figure S5 for single-cell focus counts and numerical values. (B) Chk2−/− oocytes are highly IR-resistant. Shown are immunofluorescence images of ovarian sections labeled with nuclear and cytoplasmic germ cell markers (p63 and MVH, respectively). (C) RAD51 focus counts from newborn oocyte spreads. Only oocytes with discrete patterns of RAD51 foci were scored, as defined in Figure S3. Data points represent individual oocytes, derived from at least five different animals from each genotypic group. Horizontal hashes denote means and SD calculated using a mixed model (see STAR Methods). Asterisks indicate statistically significant differences between groups: ∗∗∗p ≤ 0.001, ∗∗p ≤ 0.005, ∗p ≤ 0.05 (Tukey HSD). See Table S3 for raw data and statistical calculations. (D) Model for pachytene checkpoint activation in mouse oocytes. Oocytes with many unsynapsed chromosomes (green) ultimately accumulate DSBs, which cannot be repaired because of block to IS recombination imposed by HORMADs on asynapsed axes. Failure of DSB repair leads to activation of CHK2 and downstream effector proteins (p53/TAp63) that trigger apoptosis. Few asynapsed chromosomes (red) lead to inactivation of essential genes by MSUC, thereby causing oocyte death. Molecular Cell 2017 67, 1026-1036.e2DOI: (10.1016/j.molcel.2017.07.027) Copyright © 2017 Elsevier Inc. Terms and Conditions