About   Help   FAQ
Phenotypes associated with this allele
Allele Symbol
Allele Name
Allele ID
Tg(Stra8-icre)1Reb
transgene insertion 1, Robert E Braun
MGI:3779079
Summary 56 genotypes
Jump to Allelic Composition Genetic Background Genotype ID
cn1
G3bp2em1Smoc/G3bp2em1.1Smoc
Tg(Stra8-icre)1Reb/0
B6.Cg-G3bp2em1Smoc/G3bp2em1.1Smoc Tg(Stra8-icre)1Reb MGI:7333229
cn2
Hellstm1c(EUCOMM)Wtsi/Hellstm1d(EUCOMM)Wtsi
Tg(Stra8-icre)1Reb/0
(B6(Cg)-Hellstm1c(EUCOMM)Wtsi/BakrJ x B6.Cg-Hellstm1d(EUCOMM)Wtsi Tg(Stra8-icre)1Reb/Bakr)F1 MGI:6389564
cn3
Tg(Stra8-icre)1Reb/0
Upf3atm1.1Wilk/Upf3a+
involves: 129 * 129S4/SvJaeSor * C57BL/6 * FVB/NJ MGI:6117762
cn4
Upf3atm1.1Wilk/Upf3atm1.1Wilk
Tg(Stra8-icre)1Reb/0
involves: 129 * 129S4/SvJaeSor * C57BL/6 * FVB/NJ MGI:6117761
cn5
Ift172tm1.1Rama/Ift172tm1.1Rama
Tg(Stra8-icre)1Reb/0
involves: 129 * 129S4/SvJaeSor * C57BL/6 * FVB/NJ MGI:6860531
cn6
Cdk1tm3Kald/Cdk1+
Tg(Stra8-icre)1Reb/0
involves: 129 * C57BL/6 * FVB/NJ MGI:6201526
cn7
Golga2tm1.1Baos/Golga2tm1.2Baos
Tg(Stra8-icre)1Reb/0
involves: 129 * C57BL/6 * FVB/NJ MGI:6885893
cn8
Aldh1a1tm1Gdu/Aldh1a1tm1Gdu
Aldh1a2tm1.1Mbp/Aldh1a2tm1.2Mbp
Tg(Stra8-icre)1Reb/0
involves: 129 * C57BL/6 * FVB/NJ MGI:7327642
cn9
Fbxo43tm1.1Kald/Fbxo43tm1.1Kald
Tg(Stra8-icre)1Reb/0
involves: 129 * C57BL/6 * FVB/NJ * SJL MGI:6201523
cn10
Cops5tm1Rpar/Cops5tm1Rpar
Tg(Stra8-icre)1Reb/0
involves: 129 * C57BL/6 * FVB/NJ * SJL MGI:6858076
cn11
Carm1tm1.1Mtb/Carm1tm1.1Mtb
Tg(Stra8-icre)1Reb/0
involves: 129 * C57BL/6J * FVB/NJ MGI:6283447
cn12
Mfn1tm2Dcc/Mfn1tm2Dcc
Tg(Stra8-icre)1Reb/0
involves: 129 * C57BL/6J * FVB/NJ MGI:7448527
cn13
Mfn2tm3Dcc/Mfn2tm3Dcc
Tg(Stra8-icre)1Reb/0
involves: 129 * C57BL/6J * FVB/NJ MGI:7448528
cn14
Mfn1tm2Dcc/Mfn1tm2Dcc
Mfn2tm3Dcc/Mfn2tm3Dcc
Tg(Stra8-icre)1Reb/0
involves: 129 * C57BL/6J * FVB/NJ MGI:7448532
cn15
Rhoxtm1Wilk/Rhoxtm1Wilk
Tg(RNU6-RNAi:Rhox3)2Wilk/0
Tg(Stra8-icre)1Reb/0
involves: 129 * FVB/NJ MGI:5763277
cn16
Piwil2tm2.1Doca/Piwil2tm2.1Doca
Tg(Stra8-icre)1Reb/0
involves: 129P2/OlaHsd * 129S4/SvJaeSor * C57BL/6J * FVB/NJ MGI:5521297
cn17
Piwil2tm1.1Doca/Piwil2tm2.1Doca
Tg(Stra8-icre)1Reb/0
involves: 129P2/OlaHsd * 129S4/SvJaeSor * C57BL/6J * FVB/NJ MGI:5521299
cn18
Kdm6btm1.1Iwam/Kdm6btm1.2Iwam
Tg(Stra8-icre)1Reb/0
involves: 129P2/OlaHsd * C57BL/6J * FVB/NJ MGI:5551619
cn19
Mgat1tm2Jxm/Mgat1tm2Jxm
Tg(Stra8-icre)1Reb/0
involves: 129S1/Sv * 129X1/SvJ * FVB/NJ MGI:6888114
cn20
Agbl5tm1.1Ics/Agbl5tm1.1Ics
Tg(Stra8-icre)1Reb/0
involves: 129S2/SvPas * C57BL/6N MGI:6887929
cn21
Csnk2btm1.1Bb/Csnk2btm1.2Bb
Tg(Stra8-icre)1Reb/0
involves: 129S2/SvPasCrl * C57BL/6 * FVB/NJ MGI:6845052
cn22
Ift27tm1c(EUCOMM)Hmgu/Ift27tm1c(EUCOMM)Hmgu
Tg(Stra8-icre)1Reb/0
involves: 129S4/SvJae * C57BL/6J * C57BL/6N * FVB/NJ MGI:6861833
cn23
Ift20tm1.1Gjp/Ift20tm1.2Gjp
Tg(Stra8-icre)1Reb/0
involves: 129S4/SvJaeSor * 129S7/SvEvBrd * FVB/NJ MGI:6865740
cn24
Mettl3tm1.1Jhha/Mettl3tm1.1Jhha
Tg(Stra8-icre)1Reb/0
involves: 129S4/SvJaeSor * BALB/c * C57BL/6 * FVB/NJ MGI:6491901
cn25
Ift74tm1c(EUCOMM)Hmgu/Ift74tm1c(EUCOMM)Hmgu
Tg(Stra8-icre)1Reb/0
involves: 129S4/SvJaeSor * C57BL/6N * FVB/NJ MGI:6864146
cn26
Fis1tm1Dcc/Fis1tm1.1Dcc
Gt(ROSA)26Sortm1.1(CAG-COX8A/Dendra2)Dcc/Gt(ROSA)26Sor+
Tg(Stra8-icre)1Reb/0
involves: 129S6/SvEvTac * C57BL/6 * FVB/NJ MGI:6755500
cn27
Huwe1tm1Alas/Y
Tg(Stra8-icre)1Reb/0
involves: 129S7/SvEvBrd * C57BL/6 * FVB/NJ MGI:7286183
cn28
Rxfp2tm1Aia/Rxfp2tm1c(EUCOMM)Wtsi
Tg(Stra8-icre)1Reb/0
involves: 129S7/SvEvBrd * C57BL/6N * FVB/NJ MGI:5473319
cn29
Spata19tm1Lij/Spata19tm1Lij
Tg(Stra8-icre)1Reb/0
involves: 129S7/SvEvBrd * FVB/NJ MGI:6510020
cn30
Ppp1cctm1Lex/Ppp1cctm1Lex
Tg(Stra8-icre)1Reb/0
involves: 129S/SvEvBrd * FVB/NJ MGI:5559510
cn31
Ppp4cem1Qys/Ppp4cem1Qys
Tg(Stra8-icre)1Reb/0
involves: 129S/SvEv * C57BL/6 * FVB/NJ MGI:7281440
cn32
Atg5tm1Myok/Atg5tm1Myok
Tg(Stra8-icre)1Reb/0
involves: 129S/SvEv * FVB/NJ MGI:6883592
cn33
Rdh10tm1c(KOMP)Wtsi/Rdh10tm1c(KOMP)Wtsi
Tg(Amh-cre)8815Reb/0
Tg(RARE-Hspa1b/lacZ)12Jrt/0
Tg(Stra8-icre)1Reb/0
involves: C57BL/6 * C57BL/6N * CD-1 * FVB/N * SJL MGI:5475537
cn34
Ift81tm1c(EUCOMM)Wtsi/Ift81tm1c(EUCOMM)Wtsi
Tg(Stra8-icre)1Reb/0
involves: C57BL/6 * C57BL/6N * FVB/NJ MGI:6864067
cn35
Rdh10tm1c(KOMP)Wtsi/Rdh10tm1c(KOMP)Wtsi
Tg(Stra8-icre)1Reb/0
involves: C57BL/6 * C57BL/6N * FVB/NJ * SJL MGI:5475535
cn36
Rdh10tm1c(KOMP)Wtsi/Rdh10tm1c(KOMP)Wtsi
Tg(Amh-cre)8815Reb/0
Tg(Stra8-icre)1Reb/0
involves: C57BL/6 * C57BL/6N * FVB/N * SJL MGI:5475536
cn37
Racgap1tm1Mahi/Racgap1tm1Mahi
Tg(Stra8-icre)1Reb/0
involves: C57BL/6 * FVB/NJ MGI:6864268
cn38
Hnrnpktm1.1Cya/Hnrnpktm1.1Cya
Tg(Stra8-icre)1Reb/0
involves: C57BL/6 * FVB/NJ MGI:7287839
cn39
Tardbptm1.1Pcw/Tardbptm1.1Pcw
Tg(Stra8-icre)1Reb/0
involves: C57BL/6 * FVB/NJ * SJL MGI:7266812
cn40
Ewsr1tm1c(EUCOMM)Wtsi/Ewsr1tm1d(EUCOMM)Wtsi
Tg(Stra8-icre)1Reb/0
involves: C57BL/6J MGI:6830505
cn41
Pramex1tm1c(KOMP)Wtsi/Y
Tg(Stra8-icre)1Reb/0
involves: C57BL/6J * C57BL/6N MGI:6457270
cn42
Slc9a8tm1c(KOMP)Wtsi/Slc9a8tm1c(KOMP)Wtsi
Tg(Stra8-icre)1Reb/0
involves: C57BL/6J * C57BL/6N * FVB/NJ MGI:6887371
cn43
Mfn2em1Cya/Mfn2em1Cya
Tg(Stra8-icre)1Reb/0
involves: C57BL/6J * FVB/NJ MGI:7327651
cn44
Ranbp9tm1Wyan/Ranbp9tm1Wyan
Tg(Stra8-icre)1Reb/0
involves: C57BL/6J * FVB/NJ MGI:5702983
cn45
Rbm46em3Rpc/Rbm46em3Rpc
Tg(Stra8-icre)1Reb/0
involves: C57BL/6J * FVB/NJ * SJL/J MGI:7356612
cn46
Cep250tm1a(EUCOMM)Wtsi/Cep250tm1a(EUCOMM)Wtsi
Tg(Stra8-icre)1Reb/0
involves: C57BL/6N MGI:7444373
cn47
Tdrd5tm1c(EUCOMM)Hmgu/Tdrd5tm1c(EUCOMM)Hmgu
Tg(Stra8-icre)1Reb/0
involves: C57BL/6N * FVB/NJ MGI:6156877
cn48
Katnal2tm1c(EUCOMM)Wtsi/Katnal2tm1c(EUCOMM)Wtsi
Tg(Stra8-icre)1Reb/0
involves: C57BL/6N * FVB/NJ MGI:6274241
cn49
Ift140tm1c(KOMP)Wtsi/Ift140tm1c(KOMP)Wtsi
Tg(Stra8-icre)1Reb/0
involves: C57BL/6N * FVB/NJ MGI:6857985
cn50
Crisp2tm1c(KOMP)Mbp/Crisp2tm1c(KOMP)Mbp
Tg(Stra8-icre)1Reb/0
involves: C57BL/6N * FVB/NJ MGI:6369095
cn51
Phf7tm1c(EUCOMM)Hmgu/Phf7tm1c(EUCOMM)Hmgu
Tg(Stra8-icre)1Reb/0
involves: C57BL/6N * FVB/NJ MGI:6690603
cn52
Cenpvtm1c(EUCOMM)Hmgu/Cenpvtm1c(EUCOMM)Hmgu
Tg(Stra8-icre)1Reb/0
involves: C57BL/6N * FVB/NJ MGI:6838391
cn53
Tg(Stra8-icre)1Reb/0
Zscan21tm1.1Jiwa/Zscan21tm1.1Jiwa
involves: FVB/NJ MGI:5904161
cn54
Tg(RNU6-RNAi:Rhox3)2Wilk/0
Tg(Stra8-icre)1Reb/0
involves: FVB/NJ MGI:5763275
cn55
Cxxc1tm2.1Skk/Cxxc1tm2.1Skk
Tg(Stra8-icre)1Reb/0
involves: FVB/NJ MGI:7507026
tg56
Tg(Stra8-icre)1Reb/0 involves: FVB MGI:3850155


Genotype
MGI:7333229
cn1
Allelic
Composition
G3bp2em1Smoc/G3bp2em1.1Smoc
Tg(Stra8-icre)1Reb/0
Genetic
Background
B6.Cg-G3bp2em1Smoc/G3bp2em1.1Smoc Tg(Stra8-icre)1Reb
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
G3bp2em1.1Smoc mutation (0 available); any G3bp2 mutation (58 available)
G3bp2em1Smoc mutation (0 available); any G3bp2 mutation (58 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• male mice exhibit normal spermatogenesis and fertility

cellular




Genotype
MGI:6389564
cn2
Allelic
Composition
Hellstm1c(EUCOMM)Wtsi/Hellstm1d(EUCOMM)Wtsi
Tg(Stra8-icre)1Reb/0
Genetic
Background
(B6(Cg)-Hellstm1c(EUCOMM)Wtsi/BakrJ x B6.Cg-Hellstm1d(EUCOMM)Wtsi Tg(Stra8-icre)1Reb/Bakr)F1
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Hellstm1c(EUCOMM)Wtsi mutation (1 available); any Hells mutation (58 available)
Hellstm1d(EUCOMM)Wtsi mutation (0 available); any Hells mutation (58 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
cellular
• At 12 days of age spermatocytes show a decrease in epigenomic modification at recombination hotspots, H3K4me3 and chromatin accessibility sites at recombination hotspots are decreased

endocrine/exocrine glands

reproductive system
• At 12 days of age spermatocytes show a decrease in epigenomic modification at recombination hotspots, H3K4me3 and chromatin accessibility sites at recombination hotspots are decreased
• Incomplete DNA repair at double stranded breaks in pachytene spermatocytes and the genomic locations of the double stranded breaks is abnormal
• arrest at the late-zygotene to early pachytene stage, with an increased proportion of leptotene and zygotene and absence of normal pachytene and diplotene spermatocytes, with the pachytene cells having H2AX phosphorylation and incomplete synapsis
• these males fail to sire offspring




Genotype
MGI:6117762
cn3
Allelic
Composition
Tg(Stra8-icre)1Reb/0
Upf3atm1.1Wilk/Upf3a+
Genetic
Background
involves: 129 * 129S4/SvJaeSor * C57BL/6 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Tg(Stra8-icre)1Reb mutation (2 available)
Upf3atm1.1Wilk mutation (0 available); any Upf3a mutation (22 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
cellular
• reduced number of pachytene spermatocytes amongst FACS sorted cells at age 4 weeks
• reduced number of cells expressing spermatocyte-specific markers amongst purified spermatocytes
• reduced number of pachytene spermatocytes and postmeiotic germ cells amongst FACS sorted cells at age 4 weeks
• reduced number of cells expressing spermatocyte-specific markers amongst purified spermatocytes
• reduced number of cells expressing spermatogenic markers at age 4 weeks

reproductive system
• reduced number of pachytene spermatocytes amongst FACS sorted cells at age 4 weeks
• reduced number of cells expressing spermatocyte-specific markers amongst purified spermatocytes
• reduced number of pachytene spermatocytes and postmeiotic germ cells amongst FACS sorted cells at age 4 weeks
• reduced number of cells expressing spermatocyte-specific markers amongst purified spermatocytes
• reduced number of cells expressing spermatogenic markers at age 4 weeks




Genotype
MGI:6117761
cn4
Allelic
Composition
Upf3atm1.1Wilk/Upf3atm1.1Wilk
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129 * 129S4/SvJaeSor * C57BL/6 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Tg(Stra8-icre)1Reb mutation (2 available)
Upf3atm1.1Wilk mutation (0 available); any Upf3a mutation (22 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
cellular
N
• number of apoptotic testicular cells at age 1 and 2 weeks
• reduced number of cells expressing spermatid-specific markers Tp2 and Prm2 amongst dissociated cells from purified seminiferous tubules
• reduced fraction of seminiferous tubules containing elongating spermatids
• 6-fold fewer pachytene spermatocytes compared to wild-type
• significant reduction in pachytene spermatocytes amongst dissociated cells from purified seminiferous tubules
• reduced number of cells expressing spermatocyte-specific markers Spo11 and Sycp3 amongst dissociated cells from purified seminiferous tubules
• reduced number of cells expressing spermatocyte-specific markers Stra8, Spo11 and Sycp3 amongst FACS sorted 4N spermatocytes
• reduced number of cells expressing germ cell marker Ldhc amongst dissociated cells from purified seminiferous tubules
• around 6-fold fewer round and elongated spermatids than wild-type
• 6-fold fewer pachytene spermatocytes compared to wild-type
• significant reduction in pachytene spermatocytes amongst dissociated cells from purified seminiferous tubules
• reduced fraction of seminiferous tubules containing elongating spermatids
• reduced number of cells expressing Ldhc (germ cell marker), Spo11 and Sycp3 (spermatocyte-specific markers), and Tp2 and Prm2 (spermatid-specific markers) amongst dissociated cells from purified seminiferous tubules
• normal number of leptotene/zygotene spermatocytes
• normal number of spermatogonia at age 1 and 2 weeks
• normal number of apoptotic testicular cells at age 1 and 2 weeks
• sperm count reduced 10-fold to around 10% of wild-type
• reduced expression of NMD (nonsense-mediated decay) target transcripts Atf4, Cdh11, Ern2, Smad5 and Smad7 in FACS sorted spermatocytes from 4-week old mice
• around 3-fold increase in apoptotic male germ cells per tubule at age 4 weeks

growth/size/body
N
• body weight and overall morphology

reproductive system
• reduced number of cells expressing spermatid-specific markers Tp2 and Prm2 amongst dissociated cells from purified seminiferous tubules
• reduced fraction of seminiferous tubules containing elongating spermatids
• 6-fold fewer pachytene spermatocytes compared to wild-type
• significant reduction in pachytene spermatocytes amongst dissociated cells from purified seminiferous tubules
• reduced number of cells expressing spermatocyte-specific markers Spo11 and Sycp3 amongst dissociated cells from purified seminiferous tubules
• reduced number of cells expressing spermatocyte-specific markers Stra8, Spo11 and Sycp3 amongst FACS sorted 4N spermatocytes
• reduced number of cells expressing germ cell marker Ldhc amongst dissociated cells from purified seminiferous tubules
• around 6-fold fewer round and elongated spermatids than wild-type
• 6-fold fewer pachytene spermatocytes compared to wild-type
• significant reduction in pachytene spermatocytes amongst dissociated cells from purified seminiferous tubules
• reduced fraction of seminiferous tubules containing elongating spermatids
• reduced number of cells expressing Ldhc (germ cell marker), Spo11 and Sycp3 (spermatocyte-specific markers), and Tp2 and Prm2 (spermatid-specific markers) amongst dissociated cells from purified seminiferous tubules
• normal number of leptotene/zygotene spermatocytes
• normal number of spermatogonia at age 1 and 2 weeks
• normal number of apoptotic testicular cells at age 1 and 2 weeks
• sperm count reduced 10-fold to around 10% of wild-type
• around 3-fold increase in apoptotic male germ cells per tubule at age 4 weeks
• significant reduction in pachytene spermatocytes amongst dissociated cells from purified seminiferous tubules
• 6-fold fewer pachytene spermatocytes compared to wild-type




Genotype
MGI:6860531
cn5
Allelic
Composition
Ift172tm1.1Rama/Ift172tm1.1Rama
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129 * 129S4/SvJaeSor * C57BL/6 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Ift172tm1.1Rama mutation (1 available); any Ift172 mutation (75 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• number of sperm collected from the cauda epididymis is significantly reduced
• >80% of total sperm exhibit morphological abnormalities
• some sperm exhibit vacuoles in the flagellum
• some epididymal sperm display a reduction in the outer dense fiber (ODF) subunits in the midpiece
• some epididymal sperm display abnormal core axonemes and accessory structures, including missing or disorganized 9 + 2 arrangement of the microtubules
• some epididymal sperm display a reduction in the outer dense fiber (ODF) subunits
• failure of mitochondria and ODFs to assemble properly into the flagellum is also observed
• however, the fibrous sheath structure appears normal
• most epididymal spermatozoa show bent flagella at the midpiece
• elongating spermatids exhibit abnormal head formation/shapes
• localization of an acrosome marker (Sp56) is normal in round spermatids but appears to be abnormal in some elongating spermatids
• elongating spermatids with misshapen heads also show distorted acrosomes, suggesting that this might be a secondary effect
• elongating spermatids exhibit a mislocalized and longer manchette; those with abnormally developed heads also show distorted acrosomes
• manchette is mislocalized in testicular germ cells
• isolated testicular germ cells exhibit a longer manchette than control cells
• seminiferous tubules have very small lumens and contain only few spermatid tails in the spermiogenesis phase
• cauda epididymis shows a low concentration of sperm in the lumen, with non-aligned sperm and numerous abnormalities
• abnormal sperm heads and tails are observed, along with sloughed round bodies of residual spermatid cytoplasm; an occasional sloughed spermatocyte nucleus is observed
• fewer sperm are detected in the lumen of seminiferous tubules in the spermiogenesis phase
• fewer sperm are present in the cauda epididymal lumen and round bodies and cytoplasmic debris are also present
• males produce significantly fewer pups per litter than control males (4.57 +/- 2.5 versus 8.67 +/- 1.2, respectively)
• after mating with wild-type females of known fertility for over 2 months, less than 50% of adult males are fertile and produce offspring with decreased litter sizes
• however, testis/body weight ratio is similar to that in control males
• expression levels of ODF2 (a component of the outer dense fiber), AKAP4 (a component of fibrous sheath), and two IFT components IFT25 and IFT57 are dramatically reduced in epididymal sperm
• sperm motility measured as curvilinear velocity (VCL) is significantly decreased

cellular
• number of sperm collected from the cauda epididymis is significantly reduced
• >80% of total sperm exhibit morphological abnormalities
• some sperm exhibit vacuoles in the flagellum
• some epididymal sperm display a reduction in the outer dense fiber (ODF) subunits in the midpiece
• some epididymal sperm display abnormal core axonemes and accessory structures, including missing or disorganized 9 + 2 arrangement of the microtubules
• some epididymal sperm display a reduction in the outer dense fiber (ODF) subunits
• failure of mitochondria and ODFs to assemble properly into the flagellum is also observed
• however, the fibrous sheath structure appears normal
• most epididymal spermatozoa show bent flagella at the midpiece
• elongating spermatids exhibit abnormal head formation/shapes
• localization of an acrosome marker (Sp56) is normal in round spermatids but appears to be abnormal in some elongating spermatids
• elongating spermatids with misshapen heads also show distorted acrosomes, suggesting that this might be a secondary effect
• elongating spermatids exhibit a mislocalized and longer manchette; those with abnormally developed heads also show distorted acrosomes
• manchette is mislocalized in testicular germ cells
• isolated testicular germ cells exhibit a longer manchette than control cells
• immunofluorescence staining of gamma-tubulin (a centrosome marker) showed that ~22.5% of isolated male germ cells exhibit multiple centrosomal signals versus less than 1% in control cells, suggesting centrosome fragmentation
• sperm motility measured as curvilinear velocity (VCL) is significantly decreased

endocrine/exocrine glands
• seminiferous tubules have very small lumens and contain only few spermatid tails in the spermiogenesis phase




Genotype
MGI:6201526
cn6
Allelic
Composition
Cdk1tm3Kald/Cdk1+
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129 * C57BL/6 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Cdk1tm3Kald mutation (0 available); any Cdk1 mutation (21 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• tubules contain only Sertoli cells with only rare spermatogonia

endocrine/exocrine glands

cellular
• tubules contain only Sertoli cells with only rare spermatogonia




Genotype
MGI:6885893
cn7
Allelic
Composition
Golga2tm1.1Baos/Golga2tm1.2Baos
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129 * C57BL/6 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Golga2tm1.1Baos mutation (0 available); any Golga2 mutation (53 available)
Golga2tm1.2Baos mutation (0 available); any Golga2 mutation (53 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
cellular
• Mitotracker-positive mitochondria are abnormally found in the sperm head surrounding the deformed nucleus instead of the flagellar midpiece, suggesting improper mitochondrial sheath assembly
• Mitotracker staining revealed complete absence of the mitochondrial sheath in the midpiece of sperm tails
• Mitotracker-positive mitochondria are abnormally found in the sperm head surrounding the nucleus
• at 2 months of age, sperm heads in the cauda epididymides are round
• acrosome is absent, as shown by the absence of Afaf signal in most germ cells and of acrosome-specific protein ZP3R (aka SP56) in sperm

reproductive system
• Mitotracker-positive mitochondria are abnormally found in the sperm head surrounding the deformed nucleus instead of the flagellar midpiece, suggesting improper mitochondrial sheath assembly
• Mitotracker staining revealed complete absence of the mitochondrial sheath in the midpiece of sperm tails
• Mitotracker-positive mitochondria are abnormally found in the sperm head surrounding the nucleus
• at 2 months of age, sperm heads in the cauda epididymides are round
• acrosome is absent, as shown by the absence of Afaf signal in most germ cells and of acrosome-specific protein ZP3R (aka SP56) in sperm
• defects in spermiogenesis are observed at 2 months of age
• however, seminiferous tubule morphology is grossly normal




Genotype
MGI:7327642
cn8
Allelic
Composition
Aldh1a1tm1Gdu/Aldh1a1tm1Gdu
Aldh1a2tm1.1Mbp/Aldh1a2tm1.2Mbp
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129 * C57BL/6 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Aldh1a1tm1Gdu mutation (1 available); any Aldh1a1 mutation (37 available)
Aldh1a2tm1.1Mbp mutation (0 available); any Aldh1a2 mutation (36 available)
Aldh1a2tm1.2Mbp mutation (0 available); any Aldh1a2 mutation (36 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• male mice exhibit normal spermatogenesis




Genotype
MGI:6201523
cn9
Allelic
Composition
Fbxo43tm1.1Kald/Fbxo43tm1.1Kald
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129 * C57BL/6 * FVB/NJ * SJL
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Fbxo43tm1.1Kald mutation (0 available); any Fbxo43 mutation (29 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• at stage XII (pachytene/diplotene)
• in prophase in meiosis I

cellular
• at stage XII (pachytene/diplotene)




Genotype
MGI:6858076
cn10
Allelic
Composition
Cops5tm1Rpar/Cops5tm1Rpar
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129 * C57BL/6 * FVB/NJ * SJL
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Cops5tm1Rpar mutation (0 available); any Cops5 mutation (23 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• TUNEL staining revealed a dramatic increase in the presence of apoptotic germ cells in the seminiferous tubules
• Western blot analysis showed that PARP1 and FANK1 expression levels are dramatically reduced, whereas Caspase-3 expression level is significantly increased
• less than 10% of sperm are motile; however, their curvilinear velocity is significantly reduced
• most sperm display no mobility
• seminiferous tubule structure is totally disrupted
• large vacuoles are present in the epithelium and these are likely the spaces remaining after Sertoli cell phagocytosis of apoptotic cells
• adult testis size is markedly reduced
• testis weight and testis weight/body weight are reduced to about one third of controls
• spermatogenesis is severely impaired: a number of degenerating germ cells are observed in the lumen of the seminiferous epithelium, with very few developed elongated spermatids
• round bodies (possibly sloughed germ cells) and cellular debris are seen in the cauda epididymis lumen
• cauda epididymal sperm counts are severely reduced
• >80% of sperm exhibit abnormal morphology
• few surviving spermatids exhibit little to no staining with acrosome marker peanut-lectin (PNA) while expression level of SPACA1 (another acrosomal component) is significantly reduced
• testicular IFT20 expression level is significantly reduced; although IFT20 is still found in the Golgi apparatus of spermatocytes, it is no longer localized in the acrosome of round spermatids and, instead, it is present as individual vesicles in the cytoplasm
• acrosome formation is absent in most round spermatids; in other spermatids, an abnormal acrosome is formed, or a very thin acrosome is attached to the Sertoli cell membrane by an abnormal ectoplasmic specialization
• few germ cells that develop into elongating spermatids exhibit abnormal nuclear structures
• elongating spermatids are rarely observed
• few germ cells that develop into elongating spermatids exhibit abnormal nuclei
• when bred with adult wild-type females of known fertility, none of the 6-wk-old or adult males tested sired any pups/litters during a 2-month breeding period, indicating complete male sterility
• however, mating behavior and vaginal plug formation are normal

cellular
• cauda epididymal sperm counts are severely reduced
• >80% of sperm exhibit abnormal morphology
• few surviving spermatids exhibit little to no staining with acrosome marker peanut-lectin (PNA) while expression level of SPACA1 (another acrosomal component) is significantly reduced
• testicular IFT20 expression level is significantly reduced; although IFT20 is still found in the Golgi apparatus of spermatocytes, it is no longer localized in the acrosome of round spermatids and, instead, it is present as individual vesicles in the cytoplasm
• acrosome formation is absent in most round spermatids; in other spermatids, an abnormal acrosome is formed, or a very thin acrosome is attached to the Sertoli cell membrane by an abnormal ectoplasmic specialization
• few germ cells that develop into elongating spermatids exhibit abnormal nuclear structures
• elongating spermatids are rarely observed
• few germ cells that develop into elongating spermatids exhibit abnormal nuclei
• TUNEL staining revealed a dramatic increase in the presence of apoptotic germ cells in the seminiferous tubules
• Western blot analysis showed that PARP1 and FANK1 expression levels are dramatically reduced, whereas Caspase-3 expression level is significantly increased
• less than 10% of sperm are motile; however, their curvilinear velocity is significantly reduced
• most sperm display no mobility

endocrine/exocrine glands
• seminiferous tubule structure is totally disrupted
• large vacuoles are present in the epithelium and these are likely the spaces remaining after Sertoli cell phagocytosis of apoptotic cells
• adult testis size is markedly reduced
• testis weight and testis weight/body weight are reduced to about one third of controls




Genotype
MGI:6283447
cn11
Allelic
Composition
Carm1tm1.1Mtb/Carm1tm1.1Mtb
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129 * C57BL/6J * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Carm1tm1.1Mtb mutation (0 available); any Carm1 mutation (93 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• percentage of sperm with a crooked/bent midpiece is significantly increased
• most cauda epididymal sperm exhibit deformed head morphology
• percentage of acephalic/headless sperm is significantly increased
• a significant drop in the number of late stage spermatids is observed, starting from step 7
• a significant drop in the number of late stage spermatids is observed, starting from step 7
• at 6 weeks of age, average sperm count in the cauda epididymis is severely reduced
• a dramatic decrease in sperm motility is observed
• a slight but significant decrease in testis weight is observed at 3 months of age
• however, testis size and weight are normal at 6 weeks of age
• development of late stage haploid spermatids is impaired
• cauda epididymis shows an accumulation of immature germ cells and aberrant sperm
• subfertile males produced an average of 1.7 pups per litter
• half of the 2-month-old males examined were completely sterile
• half of the 2-month-old males examined produced very small litters

cellular
• percentage of sperm with a crooked/bent midpiece is significantly increased
• most cauda epididymal sperm exhibit deformed head morphology
• percentage of acephalic/headless sperm is significantly increased
• a significant drop in the number of late stage spermatids is observed, starting from step 7
• a significant drop in the number of late stage spermatids is observed, starting from step 7
• at 6 weeks of age, average sperm count in the cauda epididymis is severely reduced
• a dramatic decrease in sperm motility is observed

endocrine/exocrine glands
• a slight but significant decrease in testis weight is observed at 3 months of age
• however, testis size and weight are normal at 6 weeks of age




Genotype
MGI:7448527
cn12
Allelic
Composition
Mfn1tm2Dcc/Mfn1tm2Dcc
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129 * C57BL/6J * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Mfn1tm2Dcc mutation (2 available); any Mfn1 mutation (44 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• TEM analysis of adult testes showed mitochondrial swelling and fragmentation in male germ cells
• male germ cells exhibit increased mitochondrial fission
• seminiferous tubules exhibit severe vacuolization defects as early as P28
• adult testis size is grossly smaller than that in controls
• testis weight/body weight ratio is significantly decreased from P25 to P180
• adult epididymis size is grossly smaller than that in controls

cellular
N
• immunofluorescent images of ATP5A (an outer mitochondria membrane marker) showed normal mitochondrial distribution in testicular sections at P25
• calreticulin -- a mitochondria-associated ER membranes (MAMs) and ER marker protein -- does not exhibit any abnormally diffused signal pattern in spermatocytes
• TEM analysis of adult testes showed mitochondrial swelling and fragmentation in male germ cells
• TEM analysis of adult testes showed mitochondrial swelling and fragmentation in male germ cells
• adult testes show a significant increase in mitochondrial DNA copy number at P60
• cytochrome c oxidase (COX, complex IV) activity is increased in adult testis sections
• in contrast, succinate dehydrogenase (SDH, complex II) activity is normal
• male germ cells exhibit increased mitochondrial fission
• male germ cells exhibit increased mitochondrial fission

endocrine/exocrine glands
• seminiferous tubules exhibit severe vacuolization defects as early as P28
• adult testis size is grossly smaller than that in controls
• testis weight/body weight ratio is significantly decreased from P25 to P180




Genotype
MGI:7448528
cn13
Allelic
Composition
Mfn2tm3Dcc/Mfn2tm3Dcc
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129 * C57BL/6J * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Mfn2tm3Dcc mutation (2 available); any Mfn2 mutation (26 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• at P42, late-stage spermatocytes and round spermatids appear to gradually vacuolize
• TEM analysis of adult testes showed mitochondrial swelling and fragmentation in male germ cells
• at P42, late-stage spermatocytes and round spermatids are decreased in number
• adult males show a dramatic reduction in the number of spermatozoa retrieved from the cauda epididymis
• TUNEL assays show a significantly increased number of apoptotic cells in the testes at P35 and P56
• however, testis histology is normal at P7-P28 and no increase in apoptosis is noted at P14 and P28
• male germ cells exhibit increased mitochondrial fission
• from P35, most male germ cells are gradually lost by apoptosis
• as adult males age, the number of severely atrophic seminiferous tubules is increased
• as adult males age, seminiferous tubules become severely atrophic and vacuolated (P60 to P120)
• testes are significantly smaller at P56
• testis weight/body weight ratio is significantly decreased from P35 to P120
• males show age-dependent spermatogenic defects, with germ cells starting to decline after P35
• only ~2% of epididymal sperm display normal morphology versus ~80% in wild-type controls
• ~14% of sperm exhibit a coiled tail
• ~31% of sperm exhibit a bent head
• ~53% of sperm are acephalic
• at P42, round spermatids are decreased in number and appear to gradually vacuolize
• analysis of the mitochondrial aspect ratio (AR) distribution (where AR is ratio of the major axis to the minor axis of mitochondria) showed an increased % of short mitochondria in round spermatids
• the distance between mitochondria and endoplasmic reticulum (ER) is increased by ~16% while the % of mitochondria-ER contacts is reduced by almost 50% and the ER-mitochondria contact coefficient (ERMICC) is reduced by >30% in round spermatids
• at P42, late-stage spermatocytes are decreased in number and appear to gradually vacuolize
• at P60, pachytene spermatocytes show a significantly increased thickness of the inter-mitochondrial cement (IMC)
• at P18, immunostaining of calreticulin -- a mitochondria-associated ER membranes (MAMs) and ER marker protein -- showed a diffused granular pattern in the cytoplasm of spermatocytes instead of the continuous perinuclear localization seen in controls; at P60, calreticulin signals appear to be reduced and show diffused granular distribution
• epididymis is significantly smaller at P56
• all males fail to produce offspring after mating with fertility-proven adult wild-type females for 5 months

cellular
N
• immunofluorescent images of ATP5A (an outer mitochondria membrane marker) showed normal mitochondrial distribution in testicular sections at P25
• at P42, late-stage spermatocytes and round spermatids appear to gradually vacuolize
• TEM analysis of adult testes showed mitochondrial swelling and fragmentation in male germ cells
• only ~2% of epididymal sperm display normal morphology versus ~80% in wild-type controls
• ~14% of sperm exhibit a coiled tail
• ~31% of sperm exhibit a bent head
• ~53% of sperm are acephalic
• at P42, round spermatids are decreased in number and appear to gradually vacuolize
• analysis of the mitochondrial aspect ratio (AR) distribution (where AR is ratio of the major axis to the minor axis of mitochondria) showed an increased % of short mitochondria in round spermatids
• the distance between mitochondria and endoplasmic reticulum (ER) is increased by ~16% while the % of mitochondria-ER contacts is reduced by almost 50% and the ER-mitochondria contact coefficient (ERMICC) is reduced by >30% in round spermatids
• at P42, late-stage spermatocytes are decreased in number and appear to gradually vacuolize
• at P60, pachytene spermatocytes show a significantly increased thickness of the inter-mitochondrial cement (IMC)
• at P18, immunostaining of calreticulin -- a mitochondria-associated ER membranes (MAMs) and ER marker protein -- showed a diffused granular pattern in the cytoplasm of spermatocytes instead of the continuous perinuclear localization seen in controls; at P60, calreticulin signals appear to be reduced and show diffused granular distribution
• at P42, late-stage spermatocytes and round spermatids are decreased in number
• adult males show a dramatic reduction in the number of spermatozoa retrieved from the cauda epididymis
• at P60, round spermatids exhibit a fragmented ER structure instead of the tube-like cisternae structures seen in control spermatids
• TEM analysis of adult testes showed mitochondrial swelling and fragmentation in male germ cells
• at P60, pachytene spermatocytes show a significantly increased thickness of the inter-mitochondrial cement (IMC)
• analysis of the mitochondrial aspect ratio (AR) distribution (where AR is ratio of the major axis to the minor axis of mitochondria) showed an increased % of short mitochondria in round spermatids
• the distance between mitochondria and endoplasmic reticulum (ER) is increased by ~16% while mitochondria-ER contacts are reduced by almost 50% and the ER-mitochondria contact coefficient (ERMICC) is reduced by >30% in round spermatids
• at P18, immunostaining of calreticulin -- a mitochondria-associated ER membranes (MAMs) and ER marker protein -- showed a diffused granular pattern in the cytoplasm of spermatocytes instead of the continuous perinuclear localization seen in controls; at P60, calreticulin signals appear to be reduced and show diffused granular distribution
• adult testes show a significant increase in mitochondrial DNA copy number at P60
• TUNEL assays show a significantly increased number of apoptotic cells in the testes at P35 and P56
• however, testis histology is normal at P7-P28 and no increase in apoptosis is noted at P14 and P28
• cytochrome c oxidase (COX, complex IV) activity is increased in adult testis sections
• in contrast, succinate dehydrogenase (SDH, complex II) activity is normal
• male germ cells exhibit increased mitochondrial fission
• from P35, most male germ cells are gradually lost by apoptosis
• male germ cells exhibit increased mitochondrial fission

endocrine/exocrine glands
• TUNEL assays show a significantly increased number of apoptotic cells in the testes at P35 and P56
• however, testis histology is normal at P7-P28 and no increase in apoptosis is noted at P14 and P28
• as adult males age, the number of severely atrophic seminiferous tubules is increased
• as adult males age, seminiferous tubules become severely atrophic and vacuolated (P60 to P120)
• testes are significantly smaller at P56
• testis weight/body weight ratio is significantly decreased from P35 to P120




Genotype
MGI:7448532
cn14
Allelic
Composition
Mfn1tm2Dcc/Mfn1tm2Dcc
Mfn2tm3Dcc/Mfn2tm3Dcc
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129 * C57BL/6J * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Mfn1tm2Dcc mutation (2 available); any Mfn1 mutation (44 available)
Mfn2tm3Dcc mutation (2 available); any Mfn2 mutation (26 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• almost no round spermatids are detected in testes at P25
• diameter of seminiferous tubules is significantly decreased at P14, P18, P25 and P60
• adult testes show a significantly higher number of atrophic and vacuolated seminiferous tubules
• adult testis size is much smaller than that in either single conditional knockout
• adult testis weight/body weight ratio is significantly lower than that in either single conditional knockout
• at P25 and P56, double conditional mutant males exhibit much more severe spermatogenic defects than either single conditional knockout
• almost no round spermatids are detected in testes at P25
• at P25, mitochondria are abnormally aggregated to one side of the cytoplasm in spermatocytes, unlike in either single conditional knockout where mitochondria distribution is normal
• males fail to complete meiosis; almost no post-meiotic round spermatids are found at P25
• adult epididymis size is much smaller than that in either single conditional knockout

cellular
• almost no round spermatids are detected in testes at P25
• at P25, mitochondria are abnormally aggregated to one side of the cytoplasm in spermatocytes, unlike in either single conditional knockout where mitochondria distribution is normal
• almost no round spermatids are detected in testes at P25
• at P25, mitochondria are abnormally aggregated to one side of the cytoplasm in spermatocytes, unlike in either single conditional knockout where mitochondria distribution is normal

endocrine/exocrine glands
• diameter of seminiferous tubules is significantly decreased at P14, P18, P25 and P60
• adult testes show a significantly higher number of atrophic and vacuolated seminiferous tubules
• adult testis size is much smaller than that in either single conditional knockout
• adult testis weight/body weight ratio is significantly lower than that in either single conditional knockout




Genotype
MGI:5763277
cn15
Allelic
Composition
Rhoxtm1Wilk/Rhoxtm1Wilk
Tg(RNU6-RNAi:Rhox3)2Wilk/0
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Rhoxtm1Wilk mutation (1 available); any Rhox mutation (1 available)
Tg(RNU6-RNAi:Rhox3)2Wilk mutation (0 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• dramatic reduction of post-meiotic germ cells
• dramatic
• 80% reduction

endocrine/exocrine glands
• 80% reduction

cellular
• dramatic reduction of post-meiotic germ cells
• dramatic




Genotype
MGI:5521297
cn16
Allelic
Composition
Piwil2tm2.1Doca/Piwil2tm2.1Doca
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129P2/OlaHsd * 129S4/SvJaeSor * C57BL/6J * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Piwil2tm2.1Doca mutation (1 available); any Piwil2 mutation (54 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• especially in meiotic cells

cellular
• round spermatids fail to exhibit L1 transposon silencing unlike wild-type cells
• especially in meiotic cells

endocrine/exocrine glands




Genotype
MGI:5521299
cn17
Allelic
Composition
Piwil2tm1.1Doca/Piwil2tm2.1Doca
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129P2/OlaHsd * 129S4/SvJaeSor * C57BL/6J * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Piwil2tm1.1Doca mutation (1 available); any Piwil2 mutation (54 available)
Piwil2tm2.1Doca mutation (1 available); any Piwil2 mutation (54 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• especially in meiotic cells

cellular
• round spermatids fail to exhibit L1 transposon silencing unlike wild-type cells
• especially in meiotic cells

endocrine/exocrine glands




Genotype
MGI:5551619
cn18
Allelic
Composition
Kdm6btm1.1Iwam/Kdm6btm1.2Iwam
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129P2/OlaHsd * C57BL/6J * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Kdm6btm1.1Iwam mutation (0 available); any Kdm6b mutation (65 available)
Kdm6btm1.2Iwam mutation (0 available); any Kdm6b mutation (65 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype

Enlarged testes in Kdm6btm1.1Iwam/Kdm6btm1.2Iwam Tg(Stra8-icre)1Reb/0 males

cellular
• increased Asingle spermatogonia in seminiferous tubules
• frequent spermatogonial dedifferentiation

reproductive system
• increased Asingle spermatogonia in seminiferous tubules
• frequent spermatogonial dedifferentiation
• male mice sire pups much longer without a change in litter size compared with wild-type mice
• however, litter production rate is normal
• male mice sire pups much longer without a change in litter size compared with wild-type mice
• however, litter production rate is normal

endocrine/exocrine glands




Genotype
MGI:6888114
cn19
Allelic
Composition
Mgat1tm2Jxm/Mgat1tm2Jxm
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S1/Sv * 129X1/SvJ * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Mgat1tm2Jxm mutation (1 available); any Mgat1 mutation (18 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• at 28 dpp, no mature spermatozoa are present in testis sections
• at 25 dpp, few elongated spermatids are observed
• at 24 and 25 dpp, fusion of cells adjacent to the lumen is observed in a few tubules
• at 28 dpp, ~14% of tubules contain multinucleated cells composed of fused spermatids
• starting at 24 and 25 dpp, fusion of cells adjacent to the lumen is observed in a few tubules
• at 28 dpp, number of tubules with elongated spermatids is significantly decreased and multinucleated germ cells, but no mature spermatozoa, are present
• premature upregulation of genes normally expressed later in spermatogenesis and spermiogenesis is observed at 22 and 23 dpp
• EGFR and PDGFRA transcripts and ERK1/2 signaling are decreased in 22 dpp male germ cells
• however, the number of Sertoli cells, spermatogonia, spermatocytes and stage VI and beyond tubules are relatively normal at 24-26 dpp

cellular
• at 28 dpp, no mature spermatozoa are present in testis sections
• at 25 dpp, few elongated spermatids are observed
• at 24 and 25 dpp, fusion of cells adjacent to the lumen is observed in a few tubules
• at 28 dpp, ~14% of tubules contain multinucleated cells composed of fused spermatids




Genotype
MGI:6887929
cn20
Allelic
Composition
Agbl5tm1.1Ics/Agbl5tm1.1Ics
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S2/SvPas * C57BL/6N
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Agbl5tm1.1Ics mutation (0 available); any Agbl5 mutation (42 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• male mice are fertile, unlike male Agbl5tm1.2Ics homozygotes
• splayed axonemes are observed
• epididymal sperm show malformed sperm tails with hairpin bending (back-folded initial segments)
• caudal epididymal sperm count is markedly reduced
• abnormally-shaped sperm nuclei and nuclear inclusions are observed
• round and early elongating spermatids exhibit highly glutamylated microtubules
• elongating spermatids have curly flagella and misshapen nuclei
• number of elongating spermatids with supernumerary (>2) basal bodies is increased
• absence of the perinuclear ring
• spermatids are mislocalized at different stages throughout the different regions of the seminiferous tubules
• a reduced number of mature spermatozoa are found in the lumen of the tubules
• however, seminiferous tubule size is normal
• testis tissue is disordered with spermatids of different developmental stages found mislocalized
• various defects in spermiogenesis are observed at all stages from the early elongating spermatid stage to the condensed spermatid stage, similar to the ultrastructural phenotypes seen in male Agbl5tm1.2Ics homozygotes
• vacuolization of the cytoplasm and abnormal accumulation of cytoplasm in sperm cells with highly condensed nucleus is observed
• however, assembly of sperm flagella is observed indicating partial maturation of sperm cells, unlike in male Agbl5tm1.2Ics homozygotes
• disordered manchette microtubules and absence of the perinuclear ring and circumferential groove are observed
• a decreased number of mature spermatozoa and an increased number of precursor cells are found in the cauda epididymis

cellular
• splayed axonemes are observed
• epididymal sperm show malformed sperm tails with hairpin bending (back-folded initial segments)
• caudal epididymal sperm count is markedly reduced
• abnormally-shaped sperm nuclei and nuclear inclusions are observed
• round and early elongating spermatids exhibit highly glutamylated microtubules
• elongating spermatids have curly flagella and misshapen nuclei
• number of elongating spermatids with supernumerary (>2) basal bodies is increased
• disordered manchette microtubules and absence of the perinuclear ring and circumferential groove are observed
• absence of the perinuclear ring

endocrine/exocrine glands
• spermatids are mislocalized at different stages throughout the different regions of the seminiferous tubules
• a reduced number of mature spermatozoa are found in the lumen of the tubules
• however, seminiferous tubule size is normal




Genotype
MGI:6845052
cn21
Allelic
Composition
Csnk2btm1.1Bb/Csnk2btm1.2Bb
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S2/SvPasCrl * C57BL/6 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Csnk2btm1.1Bb mutation (0 available); any Csnk2b mutation (13 available)
Csnk2btm1.2Bb mutation (0 available); any Csnk2b mutation (13 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• no round or elongated spermatids are detected in seminiferous tubules at 2 months of age
• numbers of leptotene, zygotene and pachytene spermatocytes are significantly reduced in juvenile males
• numbers of zygotene, pachytene and diplotene spermatocytes are significantly decreased at 2 months of age
• however, numbers of spermatogonia and leptotene spermatocytes are normal at 2 months of age
• number of germ cells per seminiferous tubule is significantly lower than that in controls at 12 dpp, 15 dpp and 17 dpp (corresponding to leptotene, zygotene and pachytene spermatocytes, respectively), and remains almost unchanged from 12 dpp to 17 dpp, unlike in control testes
• number of MVH-positive germ cells is dramatically reduced at 2 months of age
• no mature spermatozoa are present in the cauda epididymal lumens at 2 months of age
• number of TUNEL+ germ cells per seminiferous tubule is significantly increased at 2 months of age
• more than 50% of the seminiferous tubules are TUNEL+ compared with less than 20% in control males
• seminiferous tubules contain only a few spermatocytes at 12 dpp, 15 dpp and 17 dpp
• only a few germ cells and no round or elongated spermatids are detected in seminiferous tubules at 2 months of age
• seminiferous tubule diameter is reduced to half of that in control males at 2 months of age
• testis size is markedly smaller at 2 months of age
• testis weight-to-body weight ratio shows only small changes from day 12 to month 12 after birth and remains lower than that in controls at all ages examined
• males develop testicular atrophy that does not change with increasing age after adulthood
• first wave of spermatogenesis is severely blocked
• 8-week-old male mice fail to produce offspring during a 6-month mating period with wild-type females of known fertility

cellular
• no round or elongated spermatids are detected in seminiferous tubules at 2 months of age
• numbers of leptotene, zygotene and pachytene spermatocytes are significantly reduced in juvenile males
• numbers of zygotene, pachytene and diplotene spermatocytes are significantly decreased at 2 months of age
• however, numbers of spermatogonia and leptotene spermatocytes are normal at 2 months of age
• number of germ cells per seminiferous tubule is significantly lower than that in controls at 12 dpp, 15 dpp and 17 dpp (corresponding to leptotene, zygotene and pachytene spermatocytes, respectively), and remains almost unchanged from 12 dpp to 17 dpp, unlike in control testes
• number of MVH-positive germ cells is dramatically reduced at 2 months of age
• no mature spermatozoa are present in the cauda epididymal lumens at 2 months of age
• number of TUNEL+ germ cells per seminiferous tubule is significantly increased at 2 months of age
• more than 50% of the seminiferous tubules are TUNEL+ compared with less than 20% in control males

endocrine/exocrine glands
• more than 50% of the seminiferous tubules are TUNEL+ compared with less than 20% in control males
• seminiferous tubules contain only a few spermatocytes at 12 dpp, 15 dpp and 17 dpp
• only a few germ cells and no round or elongated spermatids are detected in seminiferous tubules at 2 months of age
• seminiferous tubule diameter is reduced to half of that in control males at 2 months of age
• testis size is markedly smaller at 2 months of age
• testis weight-to-body weight ratio shows only small changes from day 12 to month 12 after birth and remains lower than that in controls at all ages examined
• males develop testicular atrophy that does not change with increasing age after adulthood

homeostasis/metabolism
• Csnk2a2 mRNA expression and CSNK2A2 protein level is significantly decreased in testes




Genotype
MGI:6861833
cn22
Allelic
Composition
Ift27tm1c(EUCOMM)Hmgu/Ift27tm1c(EUCOMM)Hmgu
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S4/SvJae * C57BL/6J * C57BL/6N * FVB/NJ
Cell Lines HEPD0653_7_E10
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Ift27tm1c(EUCOMM)Hmgu mutation (0 available); any Ift27 mutation (18 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• males exhibit no significant differences in testis weight or testis/body weight ratio relative to control males
• only ~2% of epididymal sperm are motile but show a dramatic loss of progressive motility
• only ~2% of epididymal sperm are motile but their motility, calculated as curvilinear velocity (VCL), is severely reduced
• vast majority of epididymal sperm are immotile
• testicular expression levels of IFT25 (the IFT27 binding partner) and IFT81 (a component of IFT-B complex) are significantly reduced
• adult males show multiple signs of spermatogenesis failure during spermiogenesis
• epididymal sperm count is severely reduced
• sperm collected from the cauda epididymides show multiple abnormalities, including short, bent tails and round heads
• however, sperm lipid raft distribution appears to be normal
• very few sperm appear to have a smooth tail; different light densities are observed along the whole tail
• most sperm have vesicles/vacuoles in some area along the flagella
• some epididymal sperm flagella have a distorted/damaged cell membrane
• however, branched sperm flagella are never observed
• almost all epididymal sperm have misplaced or missing outer dense fibers (ODFs)
• TEM analysis showed that almost all epididymal sperm show a disrupted 9 + 2 core axoneme structure and a disorganized axoneme microtubule array
• very few axoneme structures are detected in the seminiferous tubules
• some sperm exhibit swollen tail tips
• uneven thickness of sperm tails in some areas
• TEM analysis of epididymal sperm showed that the mitochondria sheath is disorganized
• TEM analysis of epididymal sperm showed that the fibrous sheath is disorganized
• bent sperm tails are observed
• round and other misshapen sperm heads are observed
• round sperm heads are observed
• very few developed sperm are found in the lumen of seminiferous tubules
• cauda epididymis lumen is filled with abnormal sperm heads, cytoplasm bodies, sloughed round spermatids, sloughed aggregates of elongating spermatids, numerous detached sperm heads and abnormal short sperm tails that appear disorganized
• when bred with adult wild-type females of known fertility, none of the 6-wk-old or adult males tested sired any pups/litters during a 2-month breeding period, indicating complete male sterility

cellular
• epididymal sperm count is severely reduced
• sperm collected from the cauda epididymides show multiple abnormalities, including short, bent tails and round heads
• however, sperm lipid raft distribution appears to be normal
• very few sperm appear to have a smooth tail; different light densities are observed along the whole tail
• most sperm have vesicles/vacuoles in some area along the flagella
• some epididymal sperm flagella have a distorted/damaged cell membrane
• however, branched sperm flagella are never observed
• almost all epididymal sperm have misplaced or missing outer dense fibers (ODFs)
• TEM analysis showed that almost all epididymal sperm show a disrupted 9 + 2 core axoneme structure and a disorganized axoneme microtubule array
• very few axoneme structures are detected in the seminiferous tubules
• some sperm exhibit swollen tail tips
• uneven thickness of sperm tails in some areas
• TEM analysis of epididymal sperm showed that the mitochondria sheath is disorganized
• TEM analysis of epididymal sperm showed that the fibrous sheath is disorganized
• bent sperm tails are observed
• round and other misshapen sperm heads are observed
• round sperm heads are observed
• some epididymal sperm flagella have a distorted /damaged cell membrane
• only ~2% of epididymal sperm are motile but show a dramatic loss of progressive motility
• only ~2% of epididymal sperm are motile but their motility, calculated as curvilinear velocity (VCL), is severely reduced
• vast majority of epididymal sperm are immotile

endocrine/exocrine glands
• testicular expression levels of IFT25 (the IFT27 binding partner) and IFT81 (a component of IFT-B complex) are significantly reduced




Genotype
MGI:6865740
cn23
Allelic
Composition
Ift20tm1.1Gjp/Ift20tm1.2Gjp
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S4/SvJaeSor * 129S7/SvEvBrd * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Ift20tm1.1Gjp mutation (1 available); any Ift20 mutation (20 available)
Ift20tm1.2Gjp mutation (0 available); any Ift20 mutation (20 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• adult males exhibit no differences in testis-to-body weight ratio relative to control males
• only a small % of sperm are motile and show forward motility, and flagellar movement is limited to a slow, erratic waveform with a very low amplitude
• only about 10% of epididymal sperm are motile but their motility, calculated as curvilinear velocity (VCL), is significantly reduced
• only about 10% of epididymal sperm are motile versus 80% in control males
• very few flagellar axonemes or mature lysosomes are present in the seminiferous tubules; instead, cell debris, including degenerating chromatin and vacuoles are observed
• very few germ cells complete spermatogenesis
• however, testis histology revealed normal mitosis and meiosis
• cauda epididymal sperm count is severely reduced
• SEM analysis of epididymal sperm showed a variety of abnormal sperm morphologies, including short and kinked tails and round heads
• some epididymal sperm with kinked tails show excess retention of cytoplasm in the kinked area
• missing outer dense fibers (ODFs) are observed
• abnormal axonemal structures are frequently observed in epididymal sperm, including a disrupted 9 + 2 pattern and missing outer dense fibers (ODFs) in the axoneme
• many sperm tails are kinked
• many sperm tails are shortened
• some sperm display round, swollen heads
• caudal epididymis shows a very low sperm concentration, with a high frequency of sperm anomalies, sloughed round germ cells, residual bodies, sperm with short tails, and sperm tails without heads
• in testis, stage IX shows abnormal step 16 elongated spermatids that are not released into the lumen and residual bodies being sloughed into the lumen
• stages I-III show abnormal elongated spermatids along with some normal heads and cytoplasm but spermatid tails do not appear in the lumen
• stage X shows step 16 spermatid heads remaining in the seminiferous epithelium and residual bodies remaining at the lumen rather than being phagocytized
• stage XI shows abnormal elongating spermatids and sloughed residual bodies remaining from prior stages
• stage X shows failure of spermiation with step 16 spermatid heads remaining in the seminiferous epithelium
• first litter size was significantly reduced in 37.5% of the 6-wk-old males that sired pups (2 to 5 pups/litter versus 7 to 11 pups/litter for age-matched control males)
• none of the 14 adult males tested produced any litters after 3 months of breeding with adult wild-type females of known fertility
• however, sexual behavior is normal and vaginal plugs are observed in paired females
• 6 of 16 (37.5%) of the 6-wk-old males tested were able to sire a single litter of significantly reduced size, but failed to sire pups again after the first litter

cellular
• cauda epididymal sperm count is severely reduced
• SEM analysis of epididymal sperm showed a variety of abnormal sperm morphologies, including short and kinked tails and round heads
• some epididymal sperm with kinked tails show excess retention of cytoplasm in the kinked area
• missing outer dense fibers (ODFs) are observed
• abnormal axonemal structures are frequently observed in epididymal sperm, including a disrupted 9 + 2 pattern and missing outer dense fibers (ODFs) in the axoneme
• many sperm tails are kinked
• many sperm tails are shortened
• some sperm display round, swollen heads
• only a small % of sperm are motile and show forward motility, and flagellar movement is limited to a slow, erratic waveform with a very low amplitude
• only about 10% of epididymal sperm are motile but their motility, calculated as curvilinear velocity (VCL), is significantly reduced
• only about 10% of epididymal sperm are motile versus 80% in control males

endocrine/exocrine glands
• very few flagellar axonemes or mature lysosomes are present in the seminiferous tubules; instead, cell debris, including degenerating chromatin and vacuoles are observed




Genotype
MGI:6491901
cn24
Allelic
Composition
Mettl3tm1.1Jhha/Mettl3tm1.1Jhha
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S4/SvJaeSor * BALB/c * C57BL/6 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Mettl3tm1.1Jhha mutation (0 available); any Mettl3 mutation (39 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
cellular
• 75% reduction in sperm count in cauda epididymis

endocrine/exocrine glands
• mild degeneration
• significant reduction in testis volume

reproductive system
• 75% reduction in sperm count in cauda epididymis
• mild degeneration
• significant reduction in testis volume
• 75% reduction in sperm count in cauda epididymis




Genotype
MGI:6864146
cn25
Allelic
Composition
Ift74tm1c(EUCOMM)Hmgu/Ift74tm1c(EUCOMM)Hmgu
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S4/SvJaeSor * C57BL/6N * FVB/NJ
Cell Lines HEPD0774_6_H07
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Ift74tm1c(EUCOMM)Hmgu mutation (0 available); any Ift74 mutation (44 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• adult males show no differences in testis-to-body weight ratio relative to control males
• epididymal sperm count is dramatically reduced
• all of the few sperm collected from the cauda epididymides show a variety of abnormalities, including very short tails and mostly abnormally shaped heads
• spermatids show a variety of axonemal abnormalities in the seminiferous tubules, including a complete absence of the core axoneme, disorganized microtubules, clusters of microtubules, and abnormally formed axonemes without the central microtubules
• abnormally formed axonemes without the central microtubules are observed
• step 11 and 13 elongating spermatids lack tails
• all epididymal sperm have very short tails
• most epididymal sperm have grossly abnormal heads
• elongating spermatids show round and distorted heads
• however, acrosome formation appears to be normal
• elongating spermatids show round and distorted heads and absence of tails
• only ~2% of epididymal sperm are motile but their motility, calculated as curvilinear velocity (VCL), is significantly reduced
• vast majority of epididymal sperm are immotile
• seminiferous tubule lumen is almost empty, unlike in control tubules where sperm are found in the lumen
• testicular expression of IFT81 protein (a major IFT74 binding partner) and components of other IFT complex proteins (IFT27, IFT57, IFT88, and IFT140) are significantly reduced
• AKAP4 (a processed form of the major component of the fibrous sheath) is also significantly reduced
• stage XI seminiferous tubules show abnormal step 11 spermatids with abnormally shaped heads and absence of tails
• step 13 elongating spermatids lack tails while excess cytoplasm appears to be sloughed into the lumen
• cauda epididymal lumen is filled with massive amounts of large abnormal cytoplasmic bodies, residual bodies, sloughed round cells, and abnormal spermatids with short or absent tails
• failure of spermiation in stage VIII, with abnormal step 16 spermatids being phagocytized, although step 8 round spermatids and pachytene spermatocytes look normal in the same tubule
• residual bodies are not formed but small pieces of germ cell cytoplasm are retained at the luminal border
• when bred with adult wild-type females of known fertility for at least 2 months, none of the adult males tested sired any pups/litters, indicating complete male sterility

cellular
• epididymal sperm count is dramatically reduced
• all of the few sperm collected from the cauda epididymides show a variety of abnormalities, including very short tails and mostly abnormally shaped heads
• spermatids show a variety of axonemal abnormalities in the seminiferous tubules, including a complete absence of the core axoneme, disorganized microtubules, clusters of microtubules, and abnormally formed axonemes without the central microtubules
• abnormally formed axonemes without the central microtubules are observed
• step 11 and 13 elongating spermatids lack tails
• all epididymal sperm have very short tails
• most epididymal sperm have grossly abnormal heads
• elongating spermatids show round and distorted heads
• however, acrosome formation appears to be normal
• elongating spermatids show round and distorted heads and absence of tails
• only ~2% of epididymal sperm are motile but their motility, calculated as curvilinear velocity (VCL), is significantly reduced
• vast majority of epididymal sperm are immotile

endocrine/exocrine glands
• seminiferous tubule lumen is almost empty, unlike in control tubules where sperm are found in the lumen
• testicular expression of IFT81 protein (a major IFT74 binding partner) and components of other IFT complex proteins (IFT27, IFT57, IFT88, and IFT140) are significantly reduced
• AKAP4 (a processed form of the major component of the fibrous sheath) is also significantly reduced




Genotype
MGI:6755500
cn26
Allelic
Composition
Fis1tm1Dcc/Fis1tm1.1Dcc
Gt(ROSA)26Sortm1.1(CAG-COX8A/Dendra2)Dcc/Gt(ROSA)26Sor+
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S6/SvEvTac * C57BL/6 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Fis1tm1.1Dcc mutation (0 available); any Fis1 mutation (47 available)
Fis1tm1Dcc mutation (0 available); any Fis1 mutation (47 available)
Gt(ROSA)26Sortm1.1(CAG-COX8A/Dendra2)Dcc mutation (1 available); any Gt(ROSA)26Sor mutation (942 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• no spermatozoa in epididymides
• impaired acrosome development and failure to produce elongated spermatids
• with increased mitochondria content
• 4-fold increase in tubules

cellular
• no spermatozoa in epididymides
• impaired acrosome development and failure to produce elongated spermatids
• with increased mitochondria content
• in round spermatids and giant cells
• in round spermatids and giant cells with aberrant accumulation of autophagic structures
• 4-fold increase in tubules

homeostasis/metabolism
• in round spermatids and giant cells with aberrant accumulation of autophagic structures

endocrine/exocrine glands




Genotype
MGI:7286183
cn27
Allelic
Composition
Huwe1tm1Alas/Y
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S7/SvEvBrd * C57BL/6 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Huwe1tm1Alas mutation (0 available); any Huwe1 mutation (63 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• at 48 days post retinoic acid injection (48dpRA), synchronized adult testes contain fewer Stra8+ pre-leptotene spermatocytes (located away from the basement membrane of the tubules), indicating a defect in spermatogonial differentiation
• at 8 days post retinoic acid injection (8dpRA), synchronized testes show clear loss of pre-leptotene/leptotene spermatocytes (germ cells located away from the basement membrane) in the tubules
• at 8dpRA, the average number of Stra8+ pre-leptotene spermatocytes is significantly reduced in synchronized testes, whereas the number of Stra8+ spermatogonia (located adjacent to the basement membrane) is normal
• staining with SYCP3 (a marker used to identify different types of spermatocytes in meiotic prophase I) confirmed the presence of fewer leptotene spermatocytes at 8dpRA
• at 10 days post-partum (dpp), unsynchronized testes show an increased proportion of Stra8+ differentiating spermatogonia/pre-leptotene spermatocytes with intense gammaH2AX foci
• at 48dpRA, spermatogonia differentiation is disrupted in synchronized adult testes, leading to formation of fewer pre-leptotene spermatocytes
• at 48dpRA, expression of markers for differentiating spermatogonia (Stra8, Dazl, Sohlh2) is markedly reduced in synchronized adult testes
• at 10 dpp, unsynchronized testes show an increased proportion of Stra8+ differentiating spermatogonia/pre-leptotene spermatocytes with intense gammaH2AX foci
• significant loss of differentiating spermatogonia, spermatocytes and extensive loss of spermatids are observed in the tubules at 4 months of age
• average cauda epididymal sperm number is only 3% of that in control males
• at 8dpRA, synchronized testes show a significantly higher % of TUNEL+ tubules than control testes; similar results are obtained by staining the same sections with cleaved, activated caspase 3
• at 8dpRA, the proportion of proliferating Stra8+ pre-leptotene spermatocytes incorporating BrdU in synchronized testes is ~50% lower than in control testes
• however, no difference is noted in the proportion of proliferating Stra8+ A1 spermatogonia at 8dpRA
• at 10 dpp, unsynchronized testes show an increased proportion of Stra8+ differentiating spermatogonia/pre-leptotene spermatocytes with intense gammaH2AX foci, along with recruitment of pCHK2 to these gammaH2AX foci, indicating activation of the DNA damage response pathway
• at 8dpRA, double staining for Tra98 (a germ cell marker) and cleaved caspase 3 showed a significant increase in male germ cell apoptosis in synchronized testes relative to control testes (~68% versus 46%, respectively)
• no significant colocalization of p53 with gammaH2AX foci is observed, suggesting p53-independent apoptosis
• adult males have significantly smaller testes than control males
• average weight of adult testes is only 30% of control males
• at 8dpRA, the proportion of proliferating Stra8+ pre-leptotene spermatocytes is reduced by ~50% while expression of early meiotic markers (Spo11, Smc1b, Sycp1, Sycp3) is significantly decreased in synchronized testes, indicating impaired entry into meiosis
• at 4 months of age, males show significant loss of differentiating spermatogonia, spermatocytes and extensive loss of spermatids in the seminiferous tubules, indicating a developmental arrest early in spermatogenesis
• few sperm are present in the caput epididymis
• males fail to sire any litters during a 4-week fertility assay
• however, the frequency of vaginal plugging is normal

cellular
• at 48 days post retinoic acid injection (48dpRA), synchronized adult testes contain fewer Stra8+ pre-leptotene spermatocytes (located away from the basement membrane of the tubules), indicating a defect in spermatogonial differentiation
• at 8 days post retinoic acid injection (8dpRA), synchronized testes show clear loss of pre-leptotene/leptotene spermatocytes (germ cells located away from the basement membrane) in the tubules
• at 8dpRA, the average number of Stra8+ pre-leptotene spermatocytes is significantly reduced in synchronized testes, whereas the number of Stra8+ spermatogonia (located adjacent to the basement membrane) is normal
• staining with SYCP3 (a marker used to identify different types of spermatocytes in meiotic prophase I) confirmed the presence of fewer leptotene spermatocytes at 8dpRA
• at 10 days post-partum (dpp), unsynchronized testes show an increased proportion of Stra8+ differentiating spermatogonia/pre-leptotene spermatocytes with intense gammaH2AX foci
• at 48dpRA, spermatogonia differentiation is disrupted in synchronized adult testes, leading to formation of fewer pre-leptotene spermatocytes
• at 48dpRA, expression of markers for differentiating spermatogonia (Stra8, Dazl, Sohlh2) is markedly reduced in synchronized adult testes
• at 10 dpp, unsynchronized testes show an increased proportion of Stra8+ differentiating spermatogonia/pre-leptotene spermatocytes with intense gammaH2AX foci
• significant loss of differentiating spermatogonia, spermatocytes and extensive loss of spermatids are observed in the tubules at 4 months of age
• average cauda epididymal sperm number is only 3% of that in control males
• at 8dpRA, synchronized testes show a significantly higher % of TUNEL+ tubules than control testes; similar results are obtained by staining the same sections with cleaved, activated caspase 3
• at 8dpRA, the proportion of proliferating Stra8+ pre-leptotene spermatocytes incorporating BrdU in synchronized testes is ~50% lower than in control testes
• however, no difference is noted in the proportion of proliferating Stra8+ A1 spermatogonia at 8dpRA
• at 10 dpp, unsynchronized testes show an increased proportion of Stra8+ differentiating spermatogonia/pre-leptotene spermatocytes with intense gammaH2AX foci, along with recruitment of pCHK2 to these gammaH2AX foci, indicating activation of the DNA damage response pathway
• at 8dpRA, double staining for Tra98 (a germ cell marker) and cleaved caspase 3 showed a significant increase in male germ cell apoptosis in synchronized testes relative to control testes (~68% versus 46%, respectively)
• no significant colocalization of p53 with gammaH2AX foci is observed, suggesting p53-independent apoptosis

endocrine/exocrine glands
• at 8dpRA, synchronized testes show a significantly higher % of TUNEL+ tubules than control testes; similar results are obtained by staining the same sections with cleaved, activated caspase 3
• adult males have significantly smaller testes than control males
• average weight of adult testes is only 30% of control males




Genotype
MGI:5473319
cn28
Allelic
Composition
Rxfp2tm1Aia/Rxfp2tm1c(EUCOMM)Wtsi
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S7/SvEvBrd * C57BL/6N * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Rxfp2tm1Aia mutation (0 available); any Rxfp2 mutation (53 available)
Rxfp2tm1c(EUCOMM)Wtsi mutation (0 available); any Rxfp2 mutation (53 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• mice exhibit normal reproductive organ abnormalities




Genotype
MGI:6510020
cn29
Allelic
Composition
Spata19tm1Lij/Spata19tm1Lij
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S7/SvEvBrd * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Spata19tm1Lij mutation (0 available); any Spata19 mutation (10 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• all sperm show tail defects
• severe defects in the sheaths in the midpiece
• mitochondria are variable in size and have an irregular appearance
• males are infertile despite displaying normal mating behavior
• male infertility can be bypassed using intracytoplasmic sperm injection

cellular
• all sperm show tail defects
• severe defects in the sheaths in the midpiece
• mitochondria are variable in size and have an irregular appearance
• significant decrease in the relative activities of the mitochondrial respiratory chain complex IV and lower ATP levels in sperm samples




Genotype
MGI:5559510
cn30
Allelic
Composition
Ppp1cctm1Lex/Ppp1cctm1Lex
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S/SvEvBrd * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Ppp1cctm1Lex mutation (0 available); any Ppp1cc mutation (30 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• female mice exhibit normal fertility
• a few tubules contain spermatozoa in their lumen
• between the midpiece and principal pieces

cellular
• a few tubules contain spermatozoa in their lumen
• between the midpiece and principal pieces




Genotype
MGI:7281440
cn31
Allelic
Composition
Ppp4cem1Qys/Ppp4cem1Qys
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S/SvEv * C57BL/6 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Ppp4cem1Qys mutation (0 available); any Ppp4c mutation (40 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• tail bending (head-bent-back) is consistently observed in the midpiece of the sperm tail
• cytoplasmic remnants are attached to the midpiece
• however, mitochondrial sheath assembly is normal
• ~91% of spermatozoa in the testes and cauda epididymis exhibit abnormal tail bending (head-bent-back)
• bending sites are always observed in the midpiece of the tail, not in the head or principal piece
• TEM analysis showed that the tail bent at the midpiece is held together by tissues resembling remnants of cytoplasm
• at 2 months of age, the total number of sperm in the cauda epididymes is decreased by ~50% relative to that in controls males
• TUNEL staining showed significant spermatocyte cell loss in the seminiferous epithelium
• however, spermatogonia cell survival is normal
• TUNEL staining showed a significant increase in the number of apoptotic cells in the testes relative to control males
• TUNEL+ signals are colocalized with the spermatocyte cell marker synaptonemal complex protein-3 (SYCP3), but not with the spermatogonia cell marker ZBTB16 (aka PLZF), indicating spermatocyte cell loss in the seminiferous epithelium
• at 2 months of age, the percentage of progressive sperm is markedly reduced
• at 2 months of age, the percentage of motile sperm is markedly reduced
• some seminiferous tubules appear disorganized with large vacuoles while others show normal tubule structure in histology
• at 2 months of age, testes size is significantly smaller than in control males
• weight of adult testes is significantly lower than in control males
• however, adult body weight is normal
• cytoplasmic remnants are abnormally retained on the bent midpiece, indicating a defect in cytoplasm removal from elongated spermatids which in turn leads to the failure of spermiogenesis
• however, many aspects of spermatogenesis, including acrosome formation, nuclear condensation and elongation, mitochondrial sheath arrangement and 9 + 2 flagellar structure assembly, are normal
• when mated with wild-type females, adult male mice do not produce any progeny despite normal vaginal plug formation
• male infertility can be overcome by intracytoplasmic injection-embryo transfer (ICSI-ET) but male offspring is sub-fertile
• although tail-bending spermatozoa are found in the vaginal plug after natural mating, no sperm are found in COCs of oviductal ampullae and no two pronuclei formation is observed
• in an IVF assay, the number of spermatozoa bound to the zona pellucida is significantly decreased and sperm fail to fertilize the eggs and develop to the 2-cell stage
• spermatozoa fail to fertilize eggs by natural mating or IVF
• the number of spermatozoa bound to the zona pellucida of metaphase II oocytes is significantly decreased after IVF

cellular
• tail bending (head-bent-back) is consistently observed in the midpiece of the sperm tail
• cytoplasmic remnants are attached to the midpiece
• however, mitochondrial sheath assembly is normal
• ~91% of spermatozoa in the testes and cauda epididymis exhibit abnormal tail bending (head-bent-back)
• bending sites are always observed in the midpiece of the tail, not in the head or principal piece
• TEM analysis showed that the tail bent at the midpiece is held together by tissues resembling remnants of cytoplasm
• at 2 months of age, the total number of sperm in the cauda epididymes is decreased by ~50% relative to that in controls males
• TUNEL staining showed significant spermatocyte cell loss in the seminiferous epithelium
• however, spermatogonia cell survival is normal
• TUNEL staining showed a significant increase in the number of apoptotic cells in the testes relative to control males
• TUNEL+ signals are colocalized with the spermatocyte cell marker synaptonemal complex protein-3 (SYCP3), but not with the spermatogonia cell marker ZBTB16 (aka PLZF), indicating spermatocyte cell loss in the seminiferous epithelium
• at 2 months of age, the percentage of progressive sperm is markedly reduced
• at 2 months of age, the percentage of motile sperm is markedly reduced

endocrine/exocrine glands
• some seminiferous tubules appear disorganized with large vacuoles while others show normal tubule structure in histology
• at 2 months of age, testes size is significantly smaller than in control males
• weight of adult testes is significantly lower than in control males
• however, adult body weight is normal

Mouse Models of Human Disease
DO ID OMIM ID(s) Ref(s)
oligoasthenoteratozoospermia DOID:0070311 J:309155




Genotype
MGI:6883592
cn32
Allelic
Composition
Atg5tm1Myok/Atg5tm1Myok
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: 129S/SvEv * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Atg5tm1Myok mutation (3 available); any Atg5 mutation (27 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• cauda epididymal sperm number is reduced by ~60% relative to controls
• cauda epididymal sperm show multiple abnormalities, including distorted and round heads, abnormal flagella, double heads, and sperm bundles
• 20-30% of cauda epididymal sperm display abnormal flagella
• some epididymal sperm show a short flagellum with a bulb at the tip of the tail
• some cauda epididymal sperm exhibit a discontinuous accessory structure in the midpiece
• some sperm show a large vesicle at the midpiece
• cross-sections of two or more axonemes are seen in one cell, some of which are surrounded with disconnected mitochondria
• mitochondria staining of cauda epididymal sperm revealed abnormal mitochondrial sheath formation: discontinuous and bundle forms of mitochondria signals are observed
• 23.12% sperm exhibit unsheathed flagella at the principal piece
• some epididymal sperm show a short flagellum with a bulb at the tip of the tail
• ~50% of cauda epididymal sperm display misshapen heads, including round, swollen or bent heads
• in some sperm, significant mitochondrial staining is present at the head region, suggesting that the middle pieces and tails are coiling around the heads
• cauda epididymal sperm exhibit abnormal acrosome formation: peanut-lectin (an acrosome marker) aggregates around the nucleus indicating abnormal acrosome localization
• TEM analysis revealed abnormally shaped acrosomes in developing spermatids
• distorted and round heads are observed
• cauda epididymal sperm exhibit an abnormally formed chromatin structure
• some sperm exhibit double heads
• some elongating spermatids fuse into giant cells
• only few sperm are motile but exhibit markedly reduced motility
• only a small % of epididymal sperm are motile versus >80% in controls
• adult testis-to-body weight ratio is slightly lower than that of control males
• testicular expression of autophagosome marker LC3A/B-II is significantly decreased while expression of autophagy receptor SQSTM1/p62 is significantly increased, indicating a reduction in testicular autophagy activity
• spermatogenesis defects are first observed in step 10-14 elongating spermatids
• abnormal nuclear shapes start appearing with mixing of step 9 and 12 spermatids in stage XII
• large residual bodies are sloughed into the lumen and found even in stages XI-XI
• step 16 spermatids show retention in the epithelium in stages IX-XII
• some elongating spermatids fuse into giant cells; multiple elongating spermatids are wrapped in one cell membrane, indicating failure of individualization
• sloughing of cytoplasm or cytoplasm with attached immature elongating spermatids into the lumen is observed
• however, no major defects are observed in mitosis or meiosis
• 33.29% of epididymal sperm aggregate into sperm bundles
• cauda epididymal sperm show loss of sperm individualization: two or more axonemes are frequently wrapped in one cell membrane
• in the seminiferous tubules, multiple elongating spermatids share a single cell membrane indicating failure of individualization
• step 16 spermatids show retention in the epithelium rather than being released by spermiation in stages IX-XII
• impaired release of sperm into the seminiferous tubule lumen is likely due to failure to remove intercellular bridge components, as a result of disrupted autophagy activity
• cauda epididymal lumen is filled with sloughed germ cells, large cytoplasmic bodies, and spermatozoa with disorganized heads and tails
• 3 of six 2-mo-old males did not sire any offspring during 1 month of mating with wild-type females
• 9 of 11 older males did not sire any pups through 3 months of breeding
• 3 of six 2-mo-old males sired only 5, 1, and 8 pups, respectively, totaling 14 pups during one month of mating with wild-type females
• one 3- to 5-month-old male sired 10 pups and one 5- to 7-month-old male sired one pup, respectively, through 3 months of breeding

cellular
• cauda epididymal sperm number is reduced by ~60% relative to controls
• cauda epididymal sperm show multiple abnormalities, including distorted and round heads, abnormal flagella, double heads, and sperm bundles
• 20-30% of cauda epididymal sperm display abnormal flagella
• some epididymal sperm show a short flagellum with a bulb at the tip of the tail
• some cauda epididymal sperm exhibit a discontinuous accessory structure in the midpiece
• some sperm show a large vesicle at the midpiece
• cross-sections of two or more axonemes are seen in one cell, some of which are surrounded with disconnected mitochondria
• mitochondria staining of cauda epididymal sperm revealed abnormal mitochondrial sheath formation: discontinuous and bundle forms of mitochondria signals are observed
• 23.12% sperm exhibit unsheathed flagella at the principal piece
• some epididymal sperm show a short flagellum with a bulb at the tip of the tail
• ~50% of cauda epididymal sperm display misshapen heads, including round, swollen or bent heads
• in some sperm, significant mitochondrial staining is present at the head region, suggesting that the middle pieces and tails are coiling around the heads
• cauda epididymal sperm exhibit abnormal acrosome formation: peanut-lectin (an acrosome marker) aggregates around the nucleus indicating abnormal acrosome localization
• TEM analysis revealed abnormally shaped acrosomes in developing spermatids
• distorted and round heads are observed
• cauda epididymal sperm exhibit an abnormally formed chromatin structure
• some sperm exhibit double heads
• some elongating spermatids fuse into giant cells
• testicular expression of autophagosome marker LC3A/B-II is significantly decreased while expression of autophagy receptor SQSTM1/p62 is significantly increased, indicating a reduction in testicular autophagy activity
• only few sperm are motile but exhibit markedly reduced motility
• only a small % of epididymal sperm are motile versus >80% in controls

homeostasis/metabolism
• testicular expression of autophagosome marker LC3A/B-II is significantly decreased while expression of autophagy receptor SQSTM1/p62 is significantly increased, indicating a reduction in testicular autophagy activity

endocrine/exocrine glands
• adult testis-to-body weight ratio is slightly lower than that of control males
• testicular expression of autophagosome marker LC3A/B-II is significantly decreased while expression of autophagy receptor SQSTM1/p62 is significantly increased, indicating a reduction in testicular autophagy activity




Genotype
MGI:5475537
cn33
Allelic
Composition
Rdh10tm1c(KOMP)Wtsi/Rdh10tm1c(KOMP)Wtsi
Tg(Amh-cre)8815Reb/0
Tg(RARE-Hspa1b/lacZ)12Jrt/0
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6 * C57BL/6N * CD-1 * FVB/N * SJL
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Rdh10tm1c(KOMP)Wtsi mutation (0 available); any Rdh10 mutation (20 available)
Tg(Amh-cre)8815Reb mutation (2 available)
Tg(RARE-Hspa1b/lacZ)12Jrt mutation (2 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system

endocrine/exocrine glands




Genotype
MGI:6864067
cn34
Allelic
Composition
Ift81tm1c(EUCOMM)Wtsi/Ift81tm1c(EUCOMM)Wtsi
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6 * C57BL/6N * FVB/NJ
Cell Lines EPD0382_4_F06
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Ift81tm1c(EUCOMM)Wtsi mutation (0 available); any Ift81 mutation (34 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• adult males exhibit no differences in testis-to-body weight ratio relative to control males
• sperm tail forms without an axonemal complex of microtubules in the core
• tail of elongating spermatids displays abnormal alignment of the outer dense fibers and mitochondria around the absent axoneme
• sperm mitochondrial sheath appears disorganized
• abnormal aggregates of fibrous sheath components are sometimes seen in areas bulging away from the core
• some sperm tails appear to be kinked
• all of the few epididymal sperm present exhibit short tails
• cauda epididymal sperm density is markedly reduced
• elongating spermatids exhibit abnormal head morphology
• in areas of nucleopod formation, the acrosome shows abnormal folding at the indented region of the nucleus
• however, acrosome biogenesis is normal
• in elongating spermatids with abnormal heads, nuclei are condensed but exhibit abnormal shapes, with extensions of nucleopods that are surrounded by the acrosome
• numerous multinucleated giant germ cells are found in the lumen of seminiferous tubules and epididymis
• only ~2% of epididymal sperm are motile but their motility, calculated as curvilinear velocity (VCL), is significantly reduced
• vast majority of epididymal sperm are immotile
• ultrastructural changes are observed in the seminiferous epithelium, including elongating spermatids with abnormal heads
• testicular expression levels of most IFT components (IFT20, IFT25, IFT27, IFT57, IFT74 and IFT88) are significantly decreased; AKAP4 (a processed form of the major component of the fibrous sheath) is also reduced
• cauda epididymis lumen is filled with large cytoplasmic bodies, sloughed spermatids, and rare sperm with abnormal heads and short or absent tails
• only limited acetylated tubulin staining is detected, indicating few sperm tails in the epididymal lumen; most clusters in the lumen are DAPI-negative, indicating residual bodies
• numerous multinucleated giant germ cells are seen in the lumen of seminiferous tubules and epididymis
• however, TUNEL staining showed no increase in apoptotic cells in seminiferous tubules
• evidence of failure of spermiation is observed with the heads of thin, highly condensed step 16 spermatids pulled deep into the seminiferous epithelium
• after 2 months of breeding with adult wild-type females of known fertility, none of the adult males tested sired any pups, indicating complete male sterility
• however, sexual behavior is normal and vaginal plugs are observed in the paired females

cellular
• sperm tail forms without an axonemal complex of microtubules in the core
• tail of elongating spermatids displays abnormal alignment of the outer dense fibers and mitochondria around the absent axoneme
• sperm mitochondrial sheath appears disorganized
• abnormal aggregates of fibrous sheath components are sometimes seen in areas bulging away from the core
• some sperm tails appear to be kinked
• all of the few epididymal sperm present exhibit short tails
• cauda epididymal sperm density is markedly reduced
• elongating spermatids exhibit abnormal head morphology
• in areas of nucleopod formation, the acrosome shows abnormal folding at the indented region of the nucleus
• however, acrosome biogenesis is normal
• in elongating spermatids with abnormal heads, nuclei are condensed but exhibit abnormal shapes, with extensions of nucleopods that are surrounded by the acrosome
• numerous multinucleated giant germ cells are found in the lumen of seminiferous tubules and epididymis
• only ~2% of epididymal sperm are motile but their motility, calculated as curvilinear velocity (VCL), is significantly reduced
• vast majority of epididymal sperm are immotile

endocrine/exocrine glands
• ultrastructural changes are observed in the seminiferous epithelium, including elongating spermatids with abnormal heads
• testicular expression levels of most IFT components (IFT20, IFT25, IFT27, IFT57, IFT74 and IFT88) are significantly decreased; AKAP4 (a processed form of the major component of the fibrous sheath) is also reduced




Genotype
MGI:5475535
cn35
Allelic
Composition
Rdh10tm1c(KOMP)Wtsi/Rdh10tm1c(KOMP)Wtsi
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6 * C57BL/6N * FVB/NJ * SJL
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Rdh10tm1c(KOMP)Wtsi mutation (0 available); any Rdh10 mutation (20 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• testes weight and morphology are indistinguishable from controls at 2 and 3 weeks of age




Genotype
MGI:5475536
cn36
Allelic
Composition
Rdh10tm1c(KOMP)Wtsi/Rdh10tm1c(KOMP)Wtsi
Tg(Amh-cre)8815Reb/0
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6 * C57BL/6N * FVB/N * SJL
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Rdh10tm1c(KOMP)Wtsi mutation (0 available); any Rdh10 mutation (20 available)
Tg(Amh-cre)8815Reb mutation (2 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• block of differentiation of Aal spermatogonia into A1 spermatogonia at 2 and 3 weeks of age
• most tubules are devoid of meiotic cells at 2 and 3 weeks of age, retaining only undifferentiated spermatogonia-like cells and Sertoli cells
• however, by 9 weeks of age, tubules contain the expected spermatogonia, spermatocytes, and spermatids
• over 85% of tubules are degenerated at 2 and 3 weeks of age
• at 2 and 3 weeks of age
• dramatically lower at 2 and 3 weeks of age compared to wild-type controls and homozygous mice carrying only one of the cre transgenes
• most tubules are devoid of meiotic cells at 2 and 3 weeks of age, retaining only undifferentiated spermatogonia-like cells and Sertoli cells
• treatment with retinoic acid enhances synchrony of spermatogenesis
• however, by 9 weeks of age, spermatogenesis is similar to controls
• in adults only 1 of 48 males shows spermatogenesis with an altered stage frequency
• fail to initiate meiosis at 2 and 3 weeks of age
• numerous degenerated germ cells are present in the testes at 2 and 3 weeks of age
• males under 7 weeks of age are infertile or subfertile
• however, mice over 9 weeks of age are fertile and produce litters similar in size to controls

endocrine/exocrine glands
• most tubules are devoid of meiotic cells at 2 and 3 weeks of age, retaining only undifferentiated spermatogonia-like cells and Sertoli cells
• however, by 9 weeks of age, tubules contain the expected spermatogonia, spermatocytes, and spermatids
• over 85% of tubules are degenerated at 2 and 3 weeks of age
• at 2 and 3 weeks of age
• dramatically lower at 2 and 3 weeks of age compared to wild-type controls and homozygous mice carrying only one of the cre transgenes

cellular
• block of differentiation of Aal spermatogonia into A1 spermatogonia at 2 and 3 weeks of age




Genotype
MGI:6864268
cn37
Allelic
Composition
Racgap1tm1Mahi/Racgap1tm1Mahi
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Racgap1tm1Mahi mutation (0 available); any Racgap1 mutation (38 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• massive reduction in PLZF-positive undifferentiated spermatogonia pool, starting from 6 dpp
• presence of several bi-nucleated or multi-nucleated spermatogonia at 6-10 dpp
• severe decrease in differentiated spermatogonia and primary spermatocytes, with massive reduction in PLZF-positive undifferentiated spermatogonia at 12 dpp
• depletion of germ cell lineage with presence of multinucleated germ cells in pre-pubertal testes at 6, 8, 10 and 12 dpp
• no spermatozoa are present in the epididymis or testes
• presence of several bi-nucleated or multi-nucleated spermatogonia at 6-10 dpp
• TEM confirmed presence of several syncytial spermatogonia at 10 dpp
• however, all nuclei are properly individualized, indicating normal karyokinesis
• marked reduction in Ki67-positive cells at 12 dpp, indicating a division defect of spermatogonial cells that results in multi-nucleated cells and further proliferation arrest
• at 45 dpp, ~91.5% of seminiferous tubules are abnormal with numerous large vacuoles and either (i) lack a germ cell layer, (ii) exhibit a single type of spermatocytes or spermatids, or (iii) contain only Sertoli cells and spermatogonia
• at 45 dpp, all testes show large Sertoli cell vacuolization
• at 45 dpp, many seminiferous tubules are markedly atrophic with a decreased number of spermatocytes, spermatids and spermatozoa
• drastic reduction in testis size, starting from 10 to 12 dpp
• at 45 dpp, mean testes weight is 21.1 mg relative to 68.3 mg in control males
• number of TUNEL-positive cells is significantly reduced in pre-pubertal testes at 10 and 12 dpp
• failure to form permanent intercellular bridges in the germ cell lineage induces a spermatogenesis arrest in the PLZF-positive population of undifferentiated spermatogonia
• all males are sterile
• however, sexual behavior is normal and vaginal plugs are observed in mated females

cellular
• massive reduction in PLZF-positive undifferentiated spermatogonia pool, starting from 6 dpp
• presence of several bi-nucleated or multi-nucleated spermatogonia at 6-10 dpp
• severe decrease in differentiated spermatogonia and primary spermatocytes, with massive reduction in PLZF-positive undifferentiated spermatogonia at 12 dpp
• depletion of germ cell lineage with presence of multinucleated germ cells in pre-pubertal testes at 6, 8, 10 and 12 dpp
• no spermatozoa are present in the epididymis or testes
• presence of several bi-nucleated or multi-nucleated spermatogonia at 6-10 dpp
• TEM confirmed presence of several syncytial spermatogonia at 10 dpp
• however, all nuclei are properly individualized, indicating normal karyokinesis
• marked reduction in Ki67-positive cells at 12 dpp, indicating a division defect of spermatogonial cells that results in multi-nucleated cells and further proliferation arrest

endocrine/exocrine glands
• at 45 dpp, ~91.5% of seminiferous tubules are abnormal with numerous large vacuoles and either (i) lack a germ cell layer, (ii) exhibit a single type of spermatocytes or spermatids, or (iii) contain only Sertoli cells and spermatogonia
• at 45 dpp, all testes show large Sertoli cell vacuolization
• at 45 dpp, many seminiferous tubules are markedly atrophic with a decreased number of spermatocytes, spermatids and spermatozoa
• drastic reduction in testis size, starting from 10 to 12 dpp
• at 45 dpp, mean testes weight is 21.1 mg relative to 68.3 mg in control males
• number of TUNEL-positive cells is significantly reduced in pre-pubertal testes at 10 and 12 dpp




Genotype
MGI:7287839
cn38
Allelic
Composition
Hnrnpktm1.1Cya/Hnrnpktm1.1Cya
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6 * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Hnrnpktm1.1Cya mutation (0 available); any Hnrnpk mutation (39 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• starting at P14
• spermatocytes undergo progressive depletion during meiotic prophase

growth/size/body
• starting at P14 in male mice

endocrine/exocrine glands
• starting at P14

cellular




Genotype
MGI:7266812
cn39
Allelic
Composition
Tardbptm1.1Pcw/Tardbptm1.1Pcw
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6 * FVB/NJ * SJL
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Tardbptm1.1Pcw mutation (1 available); any Tardbp mutation (60 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• most cauda epididymal sperm are morphologically abnormal
• ~55% of sperm tails are curved/bent
• roughly 50% of cauda sperm show deformed or detached heads
• ~55% of sperm show head and midpiece bent over the principal piece of the tail
• headless sperm are observed
• extensive male germ cell depletion is observed at P35
• germ cell depletion is progressively worse at 3, 7, and 21 months of age
• males exhibit 7.4 times less cauda epididymal sperm than control males
• TUNEL staining showed a significant increase in spermatocyte apoptosis
• vacuole formation is observed within the seminiferous epithelium at P35
• at advanced ages, the seminiferous epithelium only consists of Sertoli cells
• onset of seminiferous epithelium pathology with reduced cellularity is at P12
• at P35, the seminiferous tubule diameter appears narrower than in control males
• a 1.6-fold reduction in tubule cross-section diameter is seen at 3 months of age
• atrophy of the seminiferous tubules, germ cell loss, and vacuole formation are observed at P35
• vacuole formation is progressively worse at 3, 7, and 21 months of age
• testis size is severely decreased at P35
• a 2.9-fold reduction in testis weight is seen at 3 months of age
• diplotene-like spermatocytes display nonhomologous chromosome synapsis
• zygotene-like spermatocytes show nonhomologous chromosome synapsis, short chromosome length, and abnormal number of chromosomes
• pachytene-like spermatocytes show nonhomologous chromosome synapsis, with persistent gammaH2AX flares/foci on autosomes likely indicating unrepaired double-strand DNA breaks
• at P35, spermatogonia and a few meiotic cells can be seen but no round spermatids are detected
• at 3-4 months of age, flow cytometry data showed a significant decrease in 1N cells (round spermatids) and an increase in 2N (spermatogonia, Sertoli, peritubular, and Leydig cells) and 4N (primary spermatocytes) cells, indicating arrest in meiosis
• spermatocytes show failure to complete prophase I of meiosis with arrest at the mid-pachytene stage
• 3-month-old males are severely subfertile, producing a significantly lower number of litters than control males over a 6-month period (0.6 litters versus 6 litters, respectively)

cellular
• most cauda epididymal sperm are morphologically abnormal
• ~55% of sperm tails are curved/bent
• roughly 50% of cauda sperm show deformed or detached heads
• ~55% of sperm show head and midpiece bent over the principal piece of the tail
• headless sperm are observed
• extensive male germ cell depletion is observed at P35
• germ cell depletion is progressively worse at 3, 7, and 21 months of age
• males exhibit 7.4 times less cauda epididymal sperm than control males
• zygotene-like spermatocytes show short chromosome length and abnormal number of chromosomes
• some pachytene-like spermatocytes show abnormally long chromosomes
• zygotene-like spermatocytes show abnormal number of chromosomes likely due to abnormal synapsis between nonhomologous chromosomes
• TUNEL staining showed a significant increase in spermatocyte apoptosis
• pachytene-like spermatocytes with synapsis defects show gammaH2AX foci/flares on autosomes, indicating delayed DSB repair

endocrine/exocrine glands
• vacuole formation is observed within the seminiferous epithelium at P35
• at advanced ages, the seminiferous epithelium only consists of Sertoli cells
• onset of seminiferous epithelium pathology with reduced cellularity is at P12
• at P35, the seminiferous tubule diameter appears narrower than in control males
• a 1.6-fold reduction in tubule cross-section diameter is seen at 3 months of age
• atrophy of the seminiferous tubules, germ cell loss, and vacuole formation are observed at P35
• vacuole formation is progressively worse at 3, 7, and 21 months of age
• testis size is severely decreased at P35
• a 2.9-fold reduction in testis weight is seen at 3 months of age

homeostasis/metabolism
• pachytene-like spermatocytes with synapsis defects show gammaH2AX foci/flares on autosomes, indicating delayed DSB repair




Genotype
MGI:6830505
cn40
Allelic
Composition
Ewsr1tm1c(EUCOMM)Wtsi/Ewsr1tm1d(EUCOMM)Wtsi
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6J
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Ewsr1tm1c(EUCOMM)Wtsi mutation (1 available); any Ewsr1 mutation (92 available)
Ewsr1tm1d(EUCOMM)Wtsi mutation (0 available); any Ewsr1 mutation (92 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
cellular
• spermatocytes show decreased PRDM9-dependent H3K4 and H3K36 trimethylation: decreased H3K4me3 in leptonema and zygonema and slightly decreased H3K36me3 in leptonema and zygonema but not diplonema; H3K4me3 ChIP-seq from spermatocytes identified fewer than normal peaks, with approximately 38.7% of hotspot peaks missing, in particular those with low H3K4 trimethylation in controls; reduced levels of H3K36 trimethylation, particularly in intergenic region hotspots

reproductive system
• spermatocytes show decreased PRDM9-dependent H3K4 and H3K36 trimethylation: decreased H3K4me3 in leptonema and zygonema and slightly decreased H3K36me3 in leptonema and zygonema but not diplonema; H3K4me3 ChIP-seq from spermatocytes identified fewer than normal peaks, with approximately 38.7% of hotspot peaks missing, in particular those with low H3K4 trimethylation in controls; reduced levels of H3K36 trimethylation, particularly in intergenic region hotspots
• decreased testes weight in adults but not at 14 or 18 days of age
• In spermatocytes the number of double stranded breaks, indicated by DMC1 foci, is normal in early leptotene and zygotene but double stranded repair is delayed, PRDM-dependent hotspots differ somewhat from controls, and there are increased unsynapsed chromosomes
• meiosis is blocked at pachynema/diplonema, with normal proportions of leptotene and zygotene spermatocytes, no spermatocytes are found in seminiferous tubules beyond prophase 1, and there is increased apoptotic germ cells in testes

endocrine/exocrine glands
• decreased testes weight in adults but not at 14 or 18 days of age




Genotype
MGI:6457270
cn41
Allelic
Composition
Pramex1tm1c(KOMP)Wtsi/Y
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6J * C57BL/6N
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Pramex1tm1c(KOMP)Wtsi mutation (0 available); any Pramex1 mutation (8 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• mature males are fertile with a normal pregnancy rate and litter size; sperm motility is normal at both 4 and 12 months of age
• a decrease in the number of germ cells is first noted in a few seminiferous tubules at P14 and becomes obvious at P21
• by P35, Sertoli cellonly (SCO) tubules are observed in some of the affected tubules
• at P21, TRA98+ germ cells and SOX9+ Sertoli cells are present, whereas DDX4+ germ cells are completely or partially lost, suggesting that the lost germ cells in affected tubules are primarily spermatocytes
• number of TUNEL+ germ cells is significantly increased at P7, reaches a peak level at P14 and then drops at P21; by P35, number of TUNEL+ germ cells is reduced to basal levels seen in control mice
• at P14, TUNEL/DDX4 dual-staining suggests that apoptotic cells found in the lumen of seminiferous tubules are predominantly pachytene spermatocytes
• % of apoptotic TUNEL+ seminiferous tubules is significantly increased at P7 and P14, but not at P21 and P35
• at P35, Sertoli cell-only (SCO) tubules and abnormal lumen are observed; affected tubules are frequently located at the periphery of testes
• % of abnormal seminiferous tubules is significantly increased at P35 and P120 relative to controls, with 5-7% of tubules being affected
• at P120 and P365, most of the affected tubules exhibit an abnormal lumen
• at P120, the frequency of misplaced Sertoli cells away from the basal side of the tubule is significantly increased relative to controls
• however, the number of Sertoli cells is unaffected at P21, P35, and P120
• males show a significant (12%) reduction in tests size at P120
• testis weight and testis index (testis weight/body weight x 100) begin to diverge at P21; a gradual reduction in testis weight is observed from P21 to P56 when maximum testis weight is reached
• at P120, testis weight and testis index are, respectively, 12.35% and 8.20% lower than in control mice
• males show germ cell loss in the affected seminiferous tubules during the first and subsequent rounds of spermatogenesis
• epididymal sperm count is significantly reduced by 13% at 4 months, but not at 12 months of age
• at P21, TRA98+ germ cells and SOX9+ Sertoli cells are present, whereas DDX4+ germ cells are completely or partially lost, suggesting that the lost germ cells in abnormal tubules are primarily spermatocytes
• germ cell arrest at spermatogonia stage for other affected tubules in mature mice

endocrine/exocrine glands
• % of apoptotic TUNEL+ seminiferous tubules is significantly increased at P7 and P14, but not at P21 and P35
• at P35, Sertoli cell-only (SCO) tubules and abnormal lumen are observed; affected tubules are frequently located at the periphery of testes
• % of abnormal seminiferous tubules is significantly increased at P35 and P120 relative to controls, with 5-7% of tubules being affected
• at P120 and P365, most of the affected tubules exhibit an abnormal lumen
• at P120, the frequency of misplaced Sertoli cells away from the basal side of the tubule is significantly increased relative to controls
• however, the number of Sertoli cells is unaffected at P21, P35, and P120
• males show a significant (12%) reduction in tests size at P120
• testis weight and testis index (testis weight/body weight x 100) begin to diverge at P21; a gradual reduction in testis weight is observed from P21 to P56 when maximum testis weight is reached
• at P120, testis weight and testis index are, respectively, 12.35% and 8.20% lower than in control mice

cellular
• at P21, TRA98+ germ cells and SOX9+ Sertoli cells are present, whereas DDX4+ germ cells are completely or partially lost, suggesting that the lost germ cells in abnormal tubules are primarily spermatocytes
• a decrease in the number of germ cells is first noted in a few seminiferous tubules at P14 and becomes obvious at P21
• by P35, Sertoli cellonly (SCO) tubules are observed in some of the affected tubules
• at P21, TRA98+ germ cells and SOX9+ Sertoli cells are present, whereas DDX4+ germ cells are completely or partially lost, suggesting that the lost germ cells in affected tubules are primarily spermatocytes
• epididymal sperm count is significantly reduced by 13% at 4 months, but not at 12 months of age
• number of TUNEL+ germ cells is significantly increased at P7, reaches a peak level at P14 and then drops at P21; by P35, number of TUNEL+ germ cells is reduced to basal levels seen in control mice
• at P14, TUNEL/DDX4 dual-staining suggests that apoptotic cells found in the lumen of seminiferous tubules are predominantly pachytene spermatocytes
• % of apoptotic TUNEL+ seminiferous tubules is significantly increased at P7 and P14, but not at P21 and P35




Genotype
MGI:6887371
cn42
Allelic
Composition
Slc9a8tm1c(KOMP)Wtsi/Slc9a8tm1c(KOMP)Wtsi
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6J * C57BL/6N * FVB/NJ
Cell Lines EPD0033_5_A07
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Slc9a8tm1c(KOMP)Wtsi mutation (0 available); any Slc9a8 mutation (108 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• males show no significant changes in the morphology and diameter of seminiferous tubules relative to control males
• ~5% of spermatozoa are NHE8-positive and show intact acrosomes and elongated nuclei in some tubules; this is attributed to the known ~95% deletion efficiency of Stra8-iCre mice
• spermatozoa show abnormal mitochondrial distribution, similar to Slc9a8tm1d(KOMP)Wtsi homozygotes
• MitoTracker staining revealed loss of the mitochondrial sheath, similar to Slc9a8tm1d(KOMP)Wtsi homozygotes
• in mature spermatozoa, DNA is condensed but sperm heads are not elongated and lack acrosomal caps
• acrosome biogenesis is impaired, similar to Slc9a8tm1d(KOMP)Wtsi homozygotes
• although electron-dense vesicles accumulate between the Golgi and the nuclear envelope, no acrosomal granules or acrosomal caps are found, similar to Slc9a8tm1d(KOMP)Wtsi homozygotes
• round-headed spermatozoa are observed, similar to Slc9a8tm1d(KOMP)Wtsi homozygotes
• round-headed sperm lack acrosomes, similar to Slc9a8tm1d(KOMP)Wtsi homozygotes
• males are infertile, similar to Slc9a8tm1d(KOMP)Wtsi homozygotes

cellular
• spermatozoa show abnormal mitochondrial distribution, similar to Slc9a8tm1d(KOMP)Wtsi homozygotes
• MitoTracker staining revealed loss of the mitochondrial sheath, similar to Slc9a8tm1d(KOMP)Wtsi homozygotes
• in mature spermatozoa, DNA is condensed but sperm heads are not elongated and lack acrosomal caps
• acrosome biogenesis is impaired, similar to Slc9a8tm1d(KOMP)Wtsi homozygotes
• although electron-dense vesicles accumulate between the Golgi and the nuclear envelope, no acrosomal granules or acrosomal caps are found, similar to Slc9a8tm1d(KOMP)Wtsi homozygotes
• round-headed spermatozoa are observed, similar to Slc9a8tm1d(KOMP)Wtsi homozygotes
• round-headed sperm lack acrosomes, similar to Slc9a8tm1d(KOMP)Wtsi homozygotes




Genotype
MGI:7327651
cn43
Allelic
Composition
Mfn2em1Cya/Mfn2em1Cya
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6J * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Mfn2em1Cya mutation (0 available); any Mfn2 mutation (26 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• spermatocytes exhibit aggregation of mitochondria with disrupted respiratory chain compared with control cells
• folded or broken tails
• reduced diameter
• abnormal progression from the zygotene to pachytene stages and impaired crossing over

endocrine/exocrine glands
• reduced diameter

cellular
• spermatocytes exhibit aggregation of mitochondria with disrupted respiratory chain compared with control cells
• folded or broken tails




Genotype
MGI:5702983
cn44
Allelic
Composition
Ranbp9tm1Wyan/Ranbp9tm1Wyan
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6J * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Ranbp9tm1Wyan mutation (0 available); any Ranbp9 mutation (40 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• epididymal sperm shows a variety of morphological abnormalities in the flagellum including coiled or headless flagellum
• epididymal sperm shows a variety of morphological abnormalities in the sperm head like crooked, round, or bent heads
• seminiferous epithelium is thinner in 3 month old testes and contains numerous vacuoles, indicative of germ cell depletion
• presence of numerous degenerating/degenerated spermatids, or even spermatocytes in the epididymis; this enhanced depletion of spermatocytes and spermatids and the production of deformed spermatozoa are seen only in mice older than 2 months and no differences are seen during the first wave of spermatogenesis
• proportional increase of meiotic cells (i.e.spermatocytes) and decrease in haploid cells (spermatids) despite lowered total cell number in both populations, indicating active depletion of spermatocytes and spermatids
• sperm counts are lower in 12 week old males
• about 2-fold increase in apoptotic cells, mainly spermatocytes and early round spermatids, in the testes
• seminiferous tubules at around stage VI exhibit round spermatids with an increase in unrepaired DNA double-strand breaks
• total motility and progressive motility of sperm are reduced in 12 week old males
• seminiferous epithelium is thinner in 3 month old testes and contains numerous vacuoles
• 12 week old males exhibit reduced testis size
• 12 week old males exhibit reduced testis weight
• caput and cauda epididymides are smaller
• males exhibit reduced fertility in 5-month long fertility tests, with some males completely infertile and others close to normal fertility

endocrine/exocrine glands
• seminiferous epithelium is thinner in 3 month old testes and contains numerous vacuoles
• 12 week old males exhibit reduced testis size
• 12 week old males exhibit reduced testis weight

cellular
• epididymal sperm shows a variety of morphological abnormalities in the flagellum including coiled or headless flagellum
• epididymal sperm shows a variety of morphological abnormalities in the sperm head like crooked, round, or bent heads
• seminiferous epithelium is thinner in 3 month old testes and contains numerous vacuoles, indicative of germ cell depletion
• presence of numerous degenerating/degenerated spermatids, or even spermatocytes in the epididymis; this enhanced depletion of spermatocytes and spermatids and the production of deformed spermatozoa are seen only in mice older than 2 months and no differences are seen during the first wave of spermatogenesis
• proportional increase of meiotic cells (i.e.spermatocytes) and decrease in haploid cells (spermatids) despite lowered total cell number in both populations, indicating active depletion of spermatocytes and spermatids
• sperm counts are lower in 12 week old males
• about 2-fold increase in apoptotic cells, mainly spermatocytes and early round spermatids, in the testes
• seminiferous tubules at around stage VI exhibit round spermatids with an increase in unrepaired DNA double-strand breaks
• total motility and progressive motility of sperm are reduced in 12 week old males




Genotype
MGI:7356612
cn45
Allelic
Composition
Rbm46em3Rpc/Rbm46em3Rpc
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6J * FVB/NJ * SJL/J
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Rbm46em3Rpc mutation (0 available); any Rbm46 mutation (25 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• seminiferous epithelia of testes contain only somatic Sertoli cells and a few spermatogonia

endocrine/exocrine glands




Genotype
MGI:7444373
cn46
Allelic
Composition
Cep250tm1a(EUCOMM)Wtsi/Cep250tm1a(EUCOMM)Wtsi
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6N
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Cep250tm1a(EUCOMM)Wtsi mutation (2 available); any Cep250 mutation (98 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
cellular
• reduction of germ cells was observed in mutant testes at 8 weeks

endocrine/exocrine glands
• fewer seminiferous tubules are present
• at 8 and 15 weeks of age

reproductive system
• reduction of germ cells was observed in mutant testes at 8 weeks
• fewer seminiferous tubules are present
• at 8 and 15 weeks of age




Genotype
MGI:6156877
cn47
Allelic
Composition
Tdrd5tm1c(EUCOMM)Hmgu/Tdrd5tm1c(EUCOMM)Hmgu
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6N * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Tdrd5tm1c(EUCOMM)Hmgu mutation (0 available); any Tdrd5 mutation (40 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
endocrine/exocrine glands
• average 50% reduction
• average 50% of wild-type weight

reproductive system
• average 50% reduction
• average 50% of wild-type weight
• at round spermatid stage; no elongating spermatids or spermatozoa formed




Genotype
MGI:6274241
cn48
Allelic
Composition
Katnal2tm1c(EUCOMM)Wtsi/Katnal2tm1c(EUCOMM)Wtsi
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6N * FVB/NJ
Cell Lines EPD0656_4_A03
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Katnal2tm1c(EUCOMM)Wtsi mutation (0 available); any Katnal2 mutation (201 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype

Spermatogenic defects in Katnal2tm1a(EUCOMM)Wtsi/Katnal2tm1a(EUCOMM)Wtsi Tg(Stra8-icre)1Reb/0 mice

reproductive system
• males exhibit a sperm tail phenotype identical to that seen in Katnal2m1Anu homozygotes
• elongating spermatids frequently exhibit abnormal nuclear morphology
• males exhibit a manchette phenotype identical to that seen in Katnal2m1Anu homozygotes
• multinucleated symplasts are frequently observed in the seminiferous epithelium
• multinucleated symplasts are frequently observed in the seminiferous epithelium
• total epididymal sperm content is reduced by 94.1% relative to control mice; however, testis weight and daily sperm output in the testes are normal, indicating a massive failure of spermiation
• retained elongated spermatids are often observed in stage IX tubules, unlike in control mice
• however, in contrast to Katnal2m1Anu homozygotes, tubulobulbar complex and residual body formation is similar to that observed in control mice

cellular
• males exhibit a sperm tail phenotype identical to that seen in Katnal2m1Anu homozygotes
• elongating spermatids frequently exhibit abnormal nuclear morphology
• males exhibit a manchette phenotype identical to that seen in Katnal2m1Anu homozygotes
• multinucleated symplasts are frequently observed in the seminiferous epithelium

endocrine/exocrine glands
• multinucleated symplasts are frequently observed in the seminiferous epithelium




Genotype
MGI:6857985
cn49
Allelic
Composition
Ift140tm1c(KOMP)Wtsi/Ift140tm1c(KOMP)Wtsi
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6N * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Ift140tm1c(KOMP)Wtsi mutation (0 available); any Ift140 mutation (71 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• males show normal testis size with no significant differences in testis weight and testis/body weight ratio relative to control males
• sperm count is significantly reduced; only few sperm are found in the lumen of cauda epididymis
• SEM analysis revealed epididymal sperm with amorphous heads, short/swollen flagella, uneven thickness of flagella, and other distorted shapes
• cauda epididymal sperm from 3- to 4-mo-old males display short and/or bent tails
• vacuoles are present at the base of stunted flagella
• misplaced ODFs are observed
• TEM analysis revealed that the few epididymal and testicular sperm recovered show disorganized axoneme microtubule array
• some sperm lack a 9 + 2 core axoneme structure
• some sperm display a swollen tail tip
• misplaced mitochondria components are observed
• misplaced fibrous sheath components are observed
• amorphous sperm heads are observed
• some sperm show disorganized chromatin structure
• in round spermatids, IFT20 protein does not appear to attach as tightly to the nuclear membrane as in control spermatids
• IFT27 protein appears to form a large vesicle, not observed in control round spermatids
• although IFT88 protein is still present in the cytoplasm of spermatocytes and round spermatids, it is absent from the manchette of elongating spermatids and shows abnormal accumulation in the cells at later stages
• a small % of sperm exhibit low beating amplitude with limited forward movement; curvilinear velocity is largely reduced
• less than 10% of sperm remain motile; however, their motility is significantly weaker than that of control sperm
• >90% of sperm display no mobility
• step 16 spermatids are phagocytosed along the basement membrane after stage VIII
• abnormal shapes of the developing spermatid heads appear in stage X
• an excessive accumulation of cytoplasm is observed in elongating spermatids beginning in stage XII
• epididymides contain round bodies of cytoplasm derived from the sloughing of the cytoplasmic lobes and residual bodies
• failure of spermiation in stage VIII leading to a reduction of sperm numbers in the cauda epididymis
• only few sperm are detected in the lumen of seminiferous tubules
• when mated with adult wild-type females for at least 2 months, all 6-wk-old or adult males tested fail to produce offspring, indicating complete male sterility
• however, males show normal sexual behavior and vaginal plug formation
• in an in vitro fertilization assay, only 5 of 84 C57BL/6J oocytes incubated with mutant sperm progressed to the 2-cell stage; three of these appeared markedly abnormal and none progressed to the 4-cell stage

cellular
• sperm count is significantly reduced; only few sperm are found in the lumen of cauda epididymis
• SEM analysis revealed epididymal sperm with amorphous heads, short/swollen flagella, uneven thickness of flagella, and other distorted shapes
• cauda epididymal sperm from 3- to 4-mo-old males display short and/or bent tails
• vacuoles are present at the base of stunted flagella
• misplaced ODFs are observed
• TEM analysis revealed that the few epididymal and testicular sperm recovered show disorganized axoneme microtubule array
• some sperm lack a 9 + 2 core axoneme structure
• some sperm display a swollen tail tip
• misplaced mitochondria components are observed
• misplaced fibrous sheath components are observed
• amorphous sperm heads are observed
• some sperm show disorganized chromatin structure
• in round spermatids, IFT20 protein does not appear to attach as tightly to the nuclear membrane as in control spermatids
• IFT27 protein appears to form a large vesicle, not observed in control round spermatids
• although IFT88 protein is still present in the cytoplasm of spermatocytes and round spermatids, it is absent from the manchette of elongating spermatids and shows abnormal accumulation in the cells at later stages
• a small % of sperm exhibit low beating amplitude with limited forward movement; curvilinear velocity is largely reduced
• less than 10% of sperm remain motile; however, their motility is significantly weaker than that of control sperm
• >90% of sperm display no mobility




Genotype
MGI:6369095
cn50
Allelic
Composition
Crisp2tm1c(KOMP)Mbp/Crisp2tm1c(KOMP)Mbp
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6N * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Crisp2tm1c(KOMP)Mbp mutation (0 available); any Crisp2 mutation (12 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• female fertility is similar to controls
• significant reduction in the percentage of sperm displaying rapid forms of progressive motility
• show a 28.7% reduction in the number of sperm showing any form of motility
• males are sub-fertile
• however, testes weight and sperm production and histology are similar to controls
• decrease in the percentage of sperm that undergo progesterone-evoked acrosome reaction

cellular
• significant reduction in the percentage of sperm displaying rapid forms of progressive motility
• show a 28.7% reduction in the number of sperm showing any form of motility




Genotype
MGI:6690603
cn51
Allelic
Composition
Phf7tm1c(EUCOMM)Hmgu/Phf7tm1c(EUCOMM)Hmgu
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6N * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Phf7tm1c(EUCOMM)Hmgu mutation (0 available); any Phf7 mutation (27 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• globular and larger head with nuclei
• less condensed chromatin; defective histone to protamine exchange during spermiogenesis
• during in vitro fertilization

cellular
• globular and larger head with nuclei
• less condensed chromatin; defective histone to protamine exchange during spermiogenesis




Genotype
MGI:6838391
cn52
Allelic
Composition
Cenpvtm1c(EUCOMM)Hmgu/Cenpvtm1c(EUCOMM)Hmgu
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: C57BL/6N * FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Cenpvtm1c(EUCOMM)Hmgu mutation (0 available); any Cenpv mutation (60 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• decrease in polar body extrusion




Genotype
MGI:5904161
cn53
Allelic
Composition
Tg(Stra8-icre)1Reb/0
Zscan21tm1.1Jiwa/Zscan21tm1.1Jiwa
Genetic
Background
involves: FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Tg(Stra8-icre)1Reb mutation (2 available)
Zscan21tm1.1Jiwa mutation (0 available); any Zscan21 mutation (43 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• testes show no round spermatids
• testes show few spermatocytes, without round spermatids
• testis weight of adult males is less than 40% of heterozygotes
• unsynapsed or partially synapsed chromosomes are seen in spermatocytes indicating failure of chromosomal synapsis
• spermatocytes do not progress to meiotic prophase I
• males are infertile, with 6-8 week old males failing to produce offspring after natural mating with wild-type females for more than 4 months

cellular
• testes show no round spermatids
• testes show few spermatocytes, without round spermatids
• failure of double-strand DNA break repair in spermatocytes as indicated by reduced MLH1 foci and RAD51 foci

endocrine/exocrine glands
• testis weight of adult males is less than 40% of heterozygotes

homeostasis/metabolism
• failure of double-strand DNA break repair in spermatocytes as indicated by reduced MLH1 foci and RAD51 foci




Genotype
MGI:5763275
cn54
Allelic
Composition
Tg(RNU6-RNAi:Rhox3)2Wilk/0
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Tg(RNU6-RNAi:Rhox3)2Wilk mutation (0 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
• abnormal of elongated spermatids
• few sperm in the cauda epididymides (greater than 100 fold reduction)
• of pachytene spermatocyte in seminiferous epithelial cycle stage IV
• of spermatocytes undergoing meiotic metaphase in stage XII
• of abnormal elongated spermatids
• decreased diameter
• however, seminal vesicle weight is normal
• defective meiotic prophase

cellular
• abnormal of elongated spermatids
• few sperm in the cauda epididymides (greater than 100 fold reduction)
• of pachytene spermatocyte in seminiferous epithelial cycle stage IV
• of spermatocytes undergoing meiotic metaphase in stage XII
• of abnormal elongated spermatids

endocrine/exocrine glands
• decreased diameter
• however, seminal vesicle weight is normal




Genotype
MGI:7507026
cn55
Allelic
Composition
Cxxc1tm2.1Skk/Cxxc1tm2.1Skk
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: FVB/NJ
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Cxxc1tm2.1Skk mutation (0 available); any Cxxc1 mutation (28 available)
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
reproductive system
N
• at 11 weeks of age, seminal vesicles, prostate, vas deferens and epididymis appear normal
• number of spermatogonial cells is modestly increased
• marked depletion of germ cells starting at 3 weeks of age
• significant increase of TUNEL+ male germ cells in the seminiferous tubules at 3 and 4 weeks, but not at 2 weeks, of age
• at 6 weeks of age, most seminiferous tubules contain only spermatogonia, few spermatocytes and spermatids, and Sertoli cells
• significant reduction in testis weight/body weight (g) at 6 and 11 weeks of age
• testis hypoplasia at 11 weeks of age
• spermatogonia enter the first stage of the meiotic process but fail to form synapsed homologous chromosomes during pachynema
• relative number of normal synapsed chromosomes is significantly reduced
• although spermatogonia and spermatocytes are present at P18, only few or no spermatids are detected in the seminiferous tubules at 6 weeks of age
• no mature sperm are detected in the lumen of the epididymis at 6 weeks of age
• at 6 weeks of age, immunofluorescence with PNA (which labels the acrosomal cap of spermatids and spermatozoa) showed a small dot-like pattern, unlike the crescent-like pattern seen in control spermatids
• flow cytometric analysis showed that one chromatid per chromosome (1C)-containing germ cells (round spermatids) are markedly reduced (13.43% vs. 4.20% in controls), whereas 4C (meiocytes in prophase I) and 2C [spermatogonia and secondary spermatocytes (MII)] germ cells are not grossly altered
• pachynema, diplonema, and MII are significantly decreased
• Cenpa staining suggests that pachynemas showing incomplete centromere pairing are eliminated as their numbers at diplotene are reduced
• failure in prophase I of the first meiosis; complete meiotic arrest is due to reduced expression of meiotic genes and reduced homologous recombination process
• male mice fail to produce pups when mated with wild-type female mice for over 6 months

cellular
• at 6 weeks of age, immunofluorescence with PNA (which labels the acrosomal cap of spermatids and spermatozoa) showed a small dot-like pattern, unlike the crescent-like pattern seen in control spermatids
• number of spermatogonial cells is modestly increased
• marked depletion of germ cells starting at 3 weeks of age
• no mature sperm are detected in the lumen of the epididymis at 6 weeks of age
• significant increase of TUNEL+ male germ cells in the seminiferous tubules at 3 and 4 weeks, but not at 2 weeks, of age
• H3K4me3 levels at transcription start sites (TSSs) and CpG islands (CGIs) are diminished in spermatocytes

endocrine/exocrine glands
• at 6 weeks of age, most seminiferous tubules contain only spermatogonia, few spermatocytes and spermatids, and Sertoli cells
• significant reduction in testis weight/body weight (g) at 6 and 11 weeks of age
• testis hypoplasia at 11 weeks of age




Genotype
MGI:3850155
tg56
Allelic
Composition
Tg(Stra8-icre)1Reb/0
Genetic
Background
involves: FVB
Find Mice Using the International Mouse Strain Resource (IMSR)
Mouse lines carrying:
Tg(Stra8-icre)1Reb mutation (2 available)
phenotype observed in females
phenotype observed in males
N normal phenotype
normal phenotype
• mice are viable and fertile; no phenotypic details are provided





Contributing Projects:
Mouse Genome Database (MGD), Gene Expression Database (GXD), Mouse Models of Human Cancer database (MMHCdb) (formerly Mouse Tumor Biology (MTB)), Gene Ontology (GO)
Citing These Resources
Funding Information
Warranty Disclaimer, Privacy Notice, Licensing, & Copyright
Send questions and comments to User Support.
last database update
04/23/2024
MGI 6.23
The Jackson Laboratory