The Mouse Spo11 Gene Is Required for Meiotic Chromosome Synapsis

Slides:



Advertisements
Similar presentations
Tumor spectrum analysis in p53-mutant mice
Advertisements

Imprinting-Mutation Mechanisms in Prader-Willi Syndrome
Volume 93, Issue 3, Pages (May 1998)
Volume 28, Issue 3, Pages (November 2007)
Joyce Tay, Joel D. Richter  Developmental Cell 
Volume 93, Issue 5, Pages (May 1998)
Volume 9, Issue 20, Pages S1-S2 (October 1999)
Comparative gene expression profiling of adult mouse ovary-derived oogonial stem cells supports a distinct cellular identity  Anthony N. Imudia, M.D.,
Volume 2, Issue 4, Pages (October 1998)
Late mitotic failure in mice lacking Sak, a polo-like kinase
Redundant and Unique Roles of Two Enhancer Elements in the TCRγ Locus in Gene Regulation and γδ T Cell Development  Na Xiong, Chulho Kang, David H Raulet 
Volume 4, Issue 4, Pages (April 2003)
Volume 12, Issue 4, Pages (April 2007)
Volume 11, Issue 4, Pages (April 2003)
Volume 7, Issue 3, Pages (March 2001)
Volume 67, Issue 6, Pages e2 (September 2017)
Volume 48, Issue 2, Pages (October 2005)
Volume 25, Issue 6, Pages (June 2017)
Nikhila S. Tanneti, Kathryn Landy, Eric F. Joyce, Kim S. McKim 
Analysis of Mouse Keratin 6a Regulatory Sequences in Transgenic Mice Reveals Constitutive, Tissue-Specific Expression by a Keratin 6a Minigene  Donna.
Barbara R. Migeon, Catherine H. Lee, Ashis K
Douglas J Guarnieri, G.Steven Dodson, Michael A Simon  Molecular Cell 
A Novel Family of Candidate Pheromone Receptors in Mammals
Volume 10, Issue 8, Pages (April 2000)
Volume 2, Issue 6, Pages (June 2002)
Nicholas S. Sokol, Lynn Cooley  Current Biology 
Naokazu Inoue, Ph. D. , Takao Nishikawa, M. S. , Masahito Ikawa, Ph. D
Mutation in the Mismatch Repair Gene Msh6 Causes Cancer Susceptibility
Ahnak/Desmoyokin Is Dispensable for Proliferation, Differentiation, and Maintenance of Integrity in Mouse Epidermis  Michiyoshi Kouno, Gen Kondoh, Kyoji.
Correlations between Synaptic Initiation and Meiotic Recombination: A Study of Humans and Mice  Jennifer R. Gruhn, Nasser Al-Asmar, Rachael Fasnacht,
Volume 27, Issue 2, Pages (October 2013)
Volume 5, Issue 1, Pages (January 2000)
Volume 30, Issue 2, Pages (May 2001)
Volume 17, Issue 4, Pages (October 2016)
Both E12 and E47 Allow Commitment to the B Cell Lineage
Targeted Myostatin Gene Editing in Multiple Mammalian Species Directed by a Single Pair of TALE Nucleases  Li Xu, Piming Zhao, Andrew Mariano, Renzhi.
Volume 4, Issue 4, Pages (April 1996)
Volume 1, Issue 5, Pages (April 1998)
Volume 123, Issue 4, Pages (November 2005)
Volume 3, Issue 4, Pages (April 1999)
Volume 10, Issue 6, Pages (December 2002)
Volume 19, Issue 5, Pages (November 2003)
Sex-Linked period Genes in the Silkmoth, Antheraea pernyi
Volume 2, Issue 4, Pages (October 1998)
Targeted disruption of the Nijmegen breakage syndrome gene NBS1 leads to early embryonic lethality in mice  Jie Zhu, Simone Petersen, Lino Tessarollo,
Β-globin Gene Switching and DNase I Sensitivity of the Endogenous β-globin Locus in Mice Do Not Require the Locus Control Region  M.A Bender, Michael.
Yuji Owada, Ichiro Suzuki, Ryoji Suzuki, Hisatake Kondo 
Volume 86, Issue 5, Pages (September 1996)
Multiple Developmental Stage–Specific Enhancers Regulate CD8 Expression in Developing Thymocytes and in Thymus-Independent T Cells  Wilfried Ellmeier,
Volume 10, Issue 4, Pages (April 1999)
Targeted Stimulation of Meiotic Recombination
Penelope R Chua, G.Shirleen Roeder  Cell 
Volume 20, Issue 23, Pages (December 2010)
The Meiosis-Specific Hop2 Protein of S
Fig. 4. Targeted disruption of STK35 transcripts in mouse.
Fig. 1. Generation of WNK3 knockout mice
Petrice W. Brown, LuAnn Judis, E
Securin is not required for cellular viability, but is required for normal growth of mouse embryonic fibroblasts  Junjie Mei, Xingxu Huang, Pumin Zhang 
Volume 21, Issue 9, Pages (May 2011)
Volume 10, Issue 8, Pages (April 2000)
Gene Amplification as a Developmental Strategy
Long-Term and Therapeutic-Level Hepatic Gene Expression of Human Factor IX after Naked Plasmid Transfer in Vivo  Carol H. Miao, Arthur R. Thompson, Keith.
Mutation of the Ca2+ Channel β Subunit Gene Cchb4 Is Associated with Ataxia and Seizures in the Lethargic (lh) Mouse  Daniel L Burgess, Julie M Jones,
Sertoli cell signaling by Desert hedgehog regulates the male germline
Volume 1, Issue 5, Pages (April 1998)
Volume 84, Issue 2, Pages (January 1996)
Volume 13, Issue 11, Pages (December 2015)
Volume 103, Issue 5, Pages (November 2000)
Targeting expression of a transgene to the airway surface epithelium using a ciliated cell-specific promoter  Lawrence E Ostrowski, James R Hutchins,
Volume 13, Issue 5, Pages (November 2007)
Presentation transcript:

The Mouse Spo11 Gene Is Required for Meiotic Chromosome Synapsis Peter J Romanienko, R.Daniel Camerini-Otero  Molecular Cell  Volume 6, Issue 5, Pages 975-987 (November 2000) DOI: 10.1016/S1097-2765(00)00097-6

Figure 1 Generation of Spo11-Deficient Mice (A) Gene disruption of the Spo11 locus. The targeting construct and genomic locus of Spo11 are shown. The EcoRI site in intron 1 was disrupted, and another EcoRI site was created at the junction of the neo cassette and exon 1 of Spo11 to facilitate identification of targeted clones. The probe was derived from the Spo11 cDNA encompassing exons 10–13. An EcoRI digest would give a 13 kb band from wild-type genomic DNA and a 16 kb band from a targeted allele. (B) Southern blot analysis of representative genotypes of Spo11 knockout mice. Homozygous knockout, −/−; heterozygous, +/−; wild-type, +/+. The digest was done with EcoRI and probed with Spo11 cDNA, exons 10–13. The arrows indicate positions of bands and their sizes are indicated. (C) PCR genotyping of Spo11 knockout mice by PCR. Mice used in experiments were genotyped by PCR. The same mice were used for Southern blot analysis in (B), indicating the method is consistent with results from Southern blotting. The primer locations are indicated in (A). Targeting at the Spo11 locus removes the binding site for one Spo11-specific primer. (D) Northern analysis of Spo11 knockout mice. Poly(A)+ RNA isolated from testes of mice with the indicated genotypes was probed with the mouse coding sequence cDNA, and the same blot was later probed with the mouse β-actin cDNA. The Spo11 transcript (1.8 kb) was absent in −/− testes and reduced in the heterozygote. The β-actin signal is not relevant to the phenotype and serves as a loading standard. Postmeiotic α-actin was absent in −/− testes. Molecular Cell 2000 6, 975-987DOI: (10.1016/S1097-2765(00)00097-6)

Figure 2 Histological Examination of Adult Testes from Spo11−/− Mice (A) Reduced testicular size in Spo11−/− mice. Wild-type, +/+; heterozygote, +/−; and homozygote knockout, −/− testes were compared from 6-week-old male sibs. (B) Hematoxylin- and eosin-stained cross section of a seminiferous tubule from a wild-type adult mouse. Magnification is 400×. (C) Hematoxylin- and eosin-stained cross sections of seminiferous tubules from a Spo11−/− adult mouse. Magnification is 400×. (D) Fluorescent TUNEL assay in wild-type mouse testis section; labeling was detected in some tubules near the basal lamina (not shown). Magnification is 200×. (E) Fluorescent TUNEL assay in Spo11−/− mouse testis section; labeled cells (green fluorescence within tubules) were detected in about 20% of tubules. Magnification is 200×; the arrow indicates positive tubule. Molecular Cell 2000 6, 975-987DOI: (10.1016/S1097-2765(00)00097-6)

Figure 3 Histological Examination of Adult and Fetal Ovaries from Spo11−/− Mice (A) Hematoxylin- and eosin-stained adult wild-type ovary. Arrow indicates maturing follicle. Eight to twelve follicles were generally seen per section. Magnification is 200×. (B) Hematoxylin- and eosin-stained adult Spo11−/− ovary. Arrow indicates follicle, but note absence of any other follicles. No more than one or two follicles were seen per section. Magnification is 200×. (C) Hematoxylin- and eosin-stained wild-type fetal ovary, 15 dpc. The two female fetal mice were from the same dam. Arrows denote cells with condensed chromatin. Magnification is 1000×. (D) Hematoxylin- and eosin-stained Spo11−/− fetal ovary, 15 dpc. Note fewer cells with condensed chromatin morphology as compared to (C). The number of cells with this morphology was half of those seen in wild type. Magnification is 1000×. Molecular Cell 2000 6, 975-987DOI: (10.1016/S1097-2765(00)00097-6)

Figure 4 Indirect Immunofluorescence of Spo11−/− Spermatocytes (A) Leptotene stage Spo11−/− spermatocyte. Merged image of αScp3 (red) and the centromere antisera CREST (green). All images initially viewed at 1000× magnification. Scale bar, 10 μm. Overlap of red and green signals results in yellow signal. (B) Zygotene-like stage of arrest in Spo11−/− spermatocytes. Merged image of αScp3 (red) and the centromere antisera CREST (green). Three nuclei are shown. (C) Zygotene wild-type spermatocyte; merged image of αScp3 (red) and the αRad51 (green). (D) Zygotene-like Spo11−/− spermatocyte; merged image of αScp3 (red) and the αRad51 (green). (E) Zygotene wild-type spematocyte; merged image of αScp3 (red) and the αDmc1 (green). (F) Zygotene-like Spo11−/− spermatocyte; merged image of αScp3 (red) and the αDmc1 (green). (G) Zygotene-like Spo11−/− spematocyte showing extent of synapsis, indicated by arrows. Stained with αScp3 (red). Molecular Cell 2000 6, 975-987DOI: (10.1016/S1097-2765(00)00097-6)

Figure 5 Spo11 Localization in Meiotic Chromosome Spreads (A) Spo11−/− spermatocyte; merged image of αScp3 (red) and the αSpo11 (green). Note absence of Spo11 signal. All images were initially viewed at 1000× magnification. Scale bar, 10 μm. Overlap of red and green signals results in yellow signal. (B) Leptotene spermatocyte; merged image of αScp3 (red) and the αSpo11 (green). (C) Zygotene spermatocyte; merged image of αScp3 (red) and the αSpo11 (green). Arrows indicate regions of synapsis. (D) Pachytene spermatocyte; merged image of αScp3 (red) and the αSpo11 (green). Arrow indicates the pseudoautosomal region (PAR). (E) Magnification of (C); arrow indicates PAR. (F) Pachytene spermatocyte; merged image of αScp1 (red) and the αSpo11 (green). Arrow indicates PAR. (G) Magnification of (E); arrow indicates PAR. Molecular Cell 2000 6, 975-987DOI: (10.1016/S1097-2765(00)00097-6)

Figure 6 Spo11 Localization and Expression in Mouse Meiotic Mutants (A) RT-PCR analysis of Spo11 expression in testes of mutant mice. The two forms of Spo11 (α and β) are indicated by arrows in the top panel, and the bottom panel shows control amplification of Aop2 (Pittman et al. 1998). The mutants analyzed are listed across the top. 13.5 dpp (days postpartum), amplification from testis cDNA from a juvenile mouse; H20, water control in amplification reaction. α and β forms of Spo11 are shown with primer location relative to exon 2, active site region, and Toprim domain. The sizes of putative proteins are indicated on the right. aa, amino acids. (B) Localization of Spo11 in Dmc1−/− spermatocyte. Merged image of αScp3 (red) and αSpo11 (green). Arrowhead indicates Spo11 staining in synapsed region, arrows indicate ends of axial elements of homologous chromosome participating in homologous synapsis, and asterisks indicate synapsis between nonhomologs. Images were initially viewed at 1000× magnification. The scale bar is 10 μm. Overlap of red and green signals results in yellow signal. (C) Localization of Spo11 in mei1 spermatocytes. Merged image of αScp3 (red) and αSpo11 (green). Arrow indicates Spo11 staining of fully synapsed homologs, and asterisks indicate synapsis between nonhomologs. Two nuclei are shown. Molecular Cell 2000 6, 975-987DOI: (10.1016/S1097-2765(00)00097-6)

Figure 7 Partial Rescue of Meiotic Arrest in Spo11−/− Spermatocytes (A) Induction of Rad51 foci in Spo11−/− spermatocytes 3 days after cisplatin administration. Merged image of αScp3 (red) and αRad51 (green). Cells were in zygotene-like stage. The images were initially viewed at 1000× magnification. Four nuclei are shown; the scale bar is 10 μm. Overlap of red and green signals results in yellow signal. (B) Induction of Dmc1 foci in Spo11−/− spermatocytes 3 days after cisplatin administration. Merged image of αScp3 (red) and αDmc1 (green). Cells were in zygotene-like stage; six nuclei are shown. (C) Colocalization of Rad51 and Dmc1 foci in Spo11−/− spermatocyte 3 days after cisplatin administration. Merged image of αRad51 (red) and αDmc1 (green). (D) Graphic representation of increased synapsis seen in Spo11−/− spermatocytes after cisplatin administration. One −/− mouse was treated with cisplatin for 3 days, the other was injected with saline. The mice were from the same litter and ≥5 weeks old when injected. One hundred nuclei were analyzed for each sample, and the number of CREST signals (centromeres) were recorded. (E) Colocalization of Scp1 and centromeres (CREST antisera) in Spo11−/− spermatocytes 3 days after cisplatin administration. Merged image of αScp1 (red) and CREST (green). Three nuclei are shown, (a) has 33 centromeres; (b), 30, and (c), 28. In many instances, synapsis extends away from the centromere. Note more αScp1 staining coincident with fewer centromeres. Molecular Cell 2000 6, 975-987DOI: (10.1016/S1097-2765(00)00097-6)