Download Impact of genetic engineering on the understanding of

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Oncogenomics wikipedia , lookup

RNA-Seq wikipedia , lookup

Gene nomenclature wikipedia , lookup

Gene wikipedia , lookup

Genomic imprinting wikipedia , lookup

X-inactivation wikipedia , lookup

Gene expression programming wikipedia , lookup

Epigenetics of neurodegenerative diseases wikipedia , lookup

Epigenetics in stem-cell differentiation wikipedia , lookup

Gene therapy wikipedia , lookup

Epigenetics of human development wikipedia , lookup

Epigenetics in learning and memory wikipedia , lookup

Point mutation wikipedia , lookup

Genetic engineering wikipedia , lookup

Gene expression profiling wikipedia , lookup

Therapeutic gene modulation wikipedia , lookup

Nutriepigenomics wikipedia , lookup

Genome (book) wikipedia , lookup

Gene therapy of the human retina wikipedia , lookup

Microevolution wikipedia , lookup

Polycomb Group Proteins and Cancer wikipedia , lookup

Artificial gene synthesis wikipedia , lookup

Vectors in gene therapy wikipedia , lookup

Site-specific recombinase technology wikipedia , lookup

Designer baby wikipedia , lookup

History of genetic engineering wikipedia , lookup

Mir-92 microRNA precursor family wikipedia , lookup

NEDD9 wikipedia , lookup

Transcript
Human Reproduction Update, Vol.7, No.2 pp. 191±210, 2001
Impact of genetic engineering on the understanding of
spermatogenesis
Denise Escalier
INSERM U25, HoÃpital Necker, Paris Cedex 15, France. Email:[email protected]
Address for correspondence: INSERM U25, HoÃpital Necker, 149 Rue de SeÁvres, F-75743 Paris Cedex 15, France. Tel: 33 1 44 49
53 67; Fax: 33 1 43 06 23 88; E-mail: [email protected]
To date, about 100 genes have been found, by genetic engineering, to be implicated in spermatogenesis. Primordial
germ cells, spermatogonia, spermatocytes I and elongating spermatids are particularly sensitive. Transgenic and
knockout mice permit an approach to be made to the question of genetic factors involved in DNA damage repair,
thermal injury, sperm chromatin compaction and sex-speci®c recombination. Knockout mice reveal unexpected
functional redundancies of testis-speci®c genes. This review considers how functional divergences can exist among
homologous genes from different species, and to what extent the phenotypes of knockout mice can be similar to those
from spontaneous mutations. Additional anomalies in reproductive function have frequently been found in these
mice, as were found factors leading to tumour susceptibility and/or various diseases. Finally, knockout mice remind
us that, in nearly all cases, hemizygous individuals retain a fertility and a wild-type sperm phenotype, although half
of the spermatozoa share a genetic defect. The ®ndings strongly emphasize the importance of understanding
epidemiology in male infertility, to identify hereditary forms of impaired spermatogenesis, and to create DNA and
pathological germ cell banks.
Key words: Germ cell/meiosis/spermatozoa/spermatogenesis/testis
TABLE OF CONTENTS
Genetic engineering strategies
Introduction
Genetic engineering strategies
Spermatogenesis revisited by means of knockout (KO) mice
Other data relative to reproduction
Diseases and male infertility
An animal model with some restraints
Conclusions
References
Data on mouse spermatogenesis have been obtained by several
genetic engineering strategies, such as null mutations in the
genome (so-called knock-out), gene overexpression, exogenous
gene expression and gene misexpression (Table I). Multiple
mutants consist either in the inactivation of several genes or gene
expression by transgenesis in a null mutant for a functional
rescue. Various information has been obtained from multiple
mutants. The method allowed mimicry of human deletions of the
Y chromosome RBM and DAZ genes (Vogel et al., 1999),
induction of the rescue of male fertility in Kit receptor-de®cient
mice by loss of p53 (Jordan et al., 1999), and the survival of
Brca1-de®cient mice by p53 deletion (Cressman et al., 1999).
Double mutants for p53 and the zinc-®nger transcription factor
Egr4 have allowed the demonstration that germ cell apoptosis
occurring in Egr4-de®cient male mice was p53-independent
(Tourtellotte et al., 1999). Egr4-Egr1 double mutants have
permitted the elucidation of the level of redundancy between
these Egr family members in regulating luteinizing hormone
production in male mice (Tourtellotte et al., 2000). Double
mutants suggest that Bmp8b and Bmp4 function as heterodimers
and homodimers in primordial germ cell speci®cation in the
mouse (Ying et al., 2000). The similar meiotic phenotypes of
Msh5±/± mice and of double mutant Msh4±/±/Msh5±/± have
Introduction
Many mutant mice obtained by genetic engineering exhibit
spermatogenesis anomalies (for reviews see Simoni, 1994;
Grootegoed et al., 1998; Okabe et al., 1998; Lamb, 1999). In a
previous review, the various germ cell phenotypes obtained by
genetic engineering were classi®ed with regard to the spermatogenesis step affected. This previous review also considered the
impact of germ cell genetic alterations on Sertoli cell behaviour
and the in¯uence of genetic background on the extent of
spermatogenesis failure (Escalier, 1999a). The present review
presents subsequently published data on the subject, and an
attempt is made to enlighten on the gains brought by genetic
engineering to the knowledge of spermatogenesis and the biology
of reproduction.
Ó European Society of Human Reproduction and Embryology
191
D.Escalier
Table I. Overview of the genes found to be involved in spermatogenesis by genetic engineeringa
Gene
Spermatogenesis anomaly
Reference(s)
Recombination, caretaker genes (safeguard genomic integrity)
Mlh1
DNA mismatch repair
Msh5
DNA mismatch repair
Msh4
DNA mismatch repair
Pms2
DNA mismatch repair
Dmc1
recombinase
Scp3
synaptonemal complex protein 3
FAC
Fanconi anaemia C gene
Fanca
Fanconi anaemia A gene
Baker, S. et al., 1996; Edelmann et al., 1996
Edelmann et al., 1999
Kneitz et al., 2000
Baker et al., 1995
Pittman et al., 1998; Yoshida et al., 1998
Yuan et al., 2000
Whitney et al., 1996
Cheng et al., 2000
Cell cycle
Ccna1
Ccnd2
Cdk4
INK4d
P27Kip1
cyclin A1
cyclin D2
cyclin-dependent kinase
cyclin D-dependent holoenzyme
cyclin-dependent kinase inhibitor
Liu et al., 1998
Sicinski et al., 1996
Rane et al., 1999
Zindy et al., 2000
Teixeira et al., 2000
Lawson et al., 1999
Il-2 (ecto)
PDGF-A
Follistatin (over)
ActRIIA
bone morphogenetic proteins
(intercellular signalling proteins)
bone morphogenetic proteins
bone morphogenetic proteins
inhibin a
epidermal growth factor
vascular endothelial
insulin-like
neurotrophic factor
stem cell growth factor receptor
Kit receptor tyrosine kinase
interleukin-2
platelet-derived growth factor
activin-binding
activin type IIA receptor
Zhao et al., 1998; Ying et al., 2000
Zhao et al., 1996
Matzuk et al., 1992; Pierson et al., 2000
Wong et al., 2000
Korpelainen et al., 1998
Baker, J. et al., 1996
Meng et al., 2000
Blume-Jensen et al., 2000
Kissel et al., 2000
Ohta et al., 1990
Gnessi et al., 2000
Guo et al., 1998
Matzuk et al.; 1995
Regulatory transcription
Egr4
A-myb
c-fos
c-myc (exo; over)
Jun D
Tarbp2
Crem
Lhx9
Sprm-1
mOvo1
Nhlh2
HSF1 (active form)
zinc ®nger, down-regulation
substrate for cdk
response to mitogens
activator
activator protein 1 complex
activator
activator
homeobox
Pou-homeodomain
zinc ®nger, possible regulator
basic helix±loop±helix
heat shock transcription factor 1
Tourtellotte et al., 1999
Toscani et al., 1997
Johnson et al., 1992
Suzuki et al., 1996; Kodaira et al., 1996
TheÂpot et al., 2000
Zhong et al., 1999
Blendy et al., 1996; Nantel et al., 1996
Birk et al., 2000
Pearse et al., 1997
Dai et al., 1998
Good et al., 1997
Nakai et al., 2000
DNA, RNA-binding proteins
Cenpb
TIAR
TSL
Mvh
N-ras
DNA-binding (centromeric)
RNA-binding
RNA-binding
RNA helicase
GDP/GTP binding
Hudson et al., 1998
Beck et al., 1998
Kuroda et al., 2000
Tanaka et al., 2000
Mangues et al., 1990
Signalling intermediate
Hsp70-2
Telomerase
Nek1
Pp1cg
MAT (over)
Dhh
IRS4
chaperone (protein folding, assembly)
holoenzyme
NIMA-related kinase
phosphatase catalytic subunit
matrix metalloproteinase
Desert hedgehog
insulin receptor substrates
Dix et al., 1997; Zhu et al., 1997
Rudolph et al., 1999; Lee, H.-W. et al., 1998
Upadhya et al., 2000
Varmuza et al., 1999; Jurisicova et al., 1999
Rudolph-Owen et al., 1998
Bitgood et al., 1996
Fantin et al., 2000
Growth factors
TGFb
Bmp4
Bmp8a
Bmp8b
EGF (exo)
VEGF (over)
IGF1
GDNF (+/±; over)
Kit/SCF-R
192
Genetic engineering and spermatogenesis
Gene
Spermatogenesis anomaly
Hormones
FSH-R
ER-a
ER-a+b
Dax1 (Ahch) (over)
ABP (over)
SRC-1
ACE
Rxrb
PLR
Insl3
Cyp19
Apoptosis
Bcl-2
Bcl-w
Bax
Apaf
FSH receptor
oestrogen receptor
orphan receptor
androgen-binding protein
steroid receptor coactivator-1
angiotensin-converting enzyme
steroid receptor
prolactin, pituitary
insulin-like hormone superfamily
aromatase cytochrome P450
(converts androgens to estrogens)
Kinases
c-mos (mis)
v-mos (over)
c-abl
c-ros R
Tyro-3
Csnk2a2
Camk4
Histones, protamines
H3.3A (hypomorphic)histone
Tnp1
Tnp2
Protamines (mis)
Other membrane-related factors
Osp/claudin-11
Gja1
Nectin-2
Basigin
Fertilin
Cyritestin
b1 integrin
Slc12a2
Oncostatin
Other enzymes
HR6B
HSL
Proacrosin
Proacrosin (exo)
PC4
GT
ODC (over; mis)
Dierich et al., 1998; Krishnamurthy et al., 2000; Abel
et al., 2000
Hess et al., 1997; Couse and Korach, 1999
Couse et al., 1999
Yu et al., 1998
Joseph et al., 1997; Selva et al., 2000
Xu et al., 1998
Hagaman et al., 1998; Ramaraj et al., 1998
Kastner et al., 1996
Gof®n et al., 1998; Steger et al., 1998
Zimmermann et al., 1999; Nef and Parada, 1999
Fisher et al., 1998; Robertson et al., 1999
pro-survival family
pro-survival family
apoptosis promoter
apoptosis activating factor
Furuchi et al., 1996; Knudson et al., 1997
Ross et al., 1998; Print et al., 1998
Knudson et al., 1995; Rucker et al., 2000
Honarpour et al., 2000
transcription activator
Matsui et al., 2000; Rotter et al., 1993; Shao et al.,
2000
Holmberg et al., 1998; Yamasaki et al.,1996
Tumour suppressor
p53
E2F-1 (exo)
Reference(s)
serine threonine kinase
serine threonine kinase
tyrosine kinase
tyrosine kinase receptor
tyrosine kinase receptor
casein kinase II a¢
Ca2+/calmodulin-dependent protein
kinase IV (serine/threonine kinase)
Higgy et al., 1995
Rosenberg et al., 1995
Kharbanda et al., 1998
Yeung et al., 1999, 2000
Lu et al., 1999
Xu et al., 1999
Wu et al., 2000
transition protein 1
transition protein 2
Couldrey et al., 1999
Yu et al., 2000
Yu et al., 2000
Lee et al., 1995; Maleszewski et al., 1998
transmembrane protein
connexin 43
Ig gene superfamilly
Ig gene superfamily, transmembrane
ADAM family (transmembrane protein)
ADAM family (membrane-anchored)
cell surface receptor
Na+-K+-2Cl±cotransporter NKCC1
haematopoietin cytokine
Gow et al., 1999
Juneja et al., 1999
Bouchard et al., 2000
Igakura et al., 1998
Cho et al., 2000
Shamsadin et al., 1999
Anderson et al., 1999
Pace et al., 2000
Malik et al., 1995
ubiquitin-conjugating enzyme
hormone-sensitive lipase
serine proteinase
Roest et al., 1996
Osuga et al., 2000
Baba et al., 1994; Adham et al., 1997
O'Brien et al., 1996
Mbikay et al., 1997
Lu and Shur, 1997
Halmekyto et al., 1991
serine protease, proprotein convertase
serine protease, galactosyltransferase
ornithine decarboxylase
Table I continued overleaf
193
D.Escalier
Gene
Spermatogenesis anomaly
Reference(s)
Others
Calmegin
IFN-b
BrcA1
THEG (insertion)
E-MAP-15
chaperone (folding control)
interferon-b
nuclear phosphoprotein
testicular haploid expressed gene
epithelial microtubule-associated protein
Ikawa et al., 1997
Iwakura et al., 1988
Cressman et al., 1999
Yanaka et al., 2000
Komada et al., 2000
a
Most were knockout mice, except: over = overexpression of the gene; mis = misexpression, either premature or delayed; exo = exogenous expression of a gene
from another species; ecto = ectopic.)
indicated that Msh5 might be epistatic to Msh4 (Kneitz et al.,
2000). Even triple mutants have been produced to study the
function of Sertoli cell receptors with tyrosine kinase activity
(Tyro3, Axl and Mer) and their simultaneous inactivation had led
to azoospermia (Lu et al., 1999).
Alternatives are to create conditional genome alterations by
controlling their onset, frequency, spatial location and tissue/cell
type speci®city. To avoid potential transcriptional interference,
point mutation by the Cre-lox P system is largely used in other
®elds. Of particular interest is the control of transgene expression
by endogenous regulatory sequence of the gene of interest (socalled knock-in). The latter enables the study of gene complementation, such as a possible rescue of a disease by a member of
the same gene family. Gene-trap mutagenesis in embryonic stem
cells (ES) is a powerful forward genetic approach in mice as it
allows the preselection or prescreening of the gene trap of
interest. This method has been used to identify retinoic acidresponsive genes during spermatogenesis, and simultaneously to
mutate them. The trapped gene was E-MAP-115 (encoding an
epithelial microtubule-associated protein of 115 kDa) and was
identi®ed as a retinoic-inducible gene (Komada et al., 2000).
Germ cell transplantation has been used in combination with
genetically modi®ed mice. Oestrogen receptor a-null mutant
males (ERa±/±) had been transplanted to testes of recipient wildtype mice expressing the receptor (ERa) but depleted of germ
cells. The recipients served as surrogate fathers for the infertile
ERa±/± males. Data showed that male germ cells do not require
ERa for development or to function in fertilization, suggesting
that male ERa±/± mice were infertile due to disruption of
oestrogen action within somatic cells of the male reproductive
system (Mahato et al., 2000). Toxic transgenesis for eliminating
germ cells may have practical application in making recipients for
spermatogonial stem cell transplantation. Expression of diphtheria
toxin A-chain was directed to the male germ line by fusion to the
rat histone H1t gene. This resulted in the total elimination of male
germ cells in two lines of mice. This experiment has demonstrated
the complete tissue speci®city of the H1t promoter (Bartell et al.,
2000).
In another ®eld, a living marker of germ cells has been
generated using a transgenic strain of mice in which green
¯uorescent protein (GFP) is driven by part of the promoter of the
Oct4 gene (Anderson et al., 1999). This facility has enabled
visualization of living primordial germ cells (PGC) and their
active migration at the time they exit the primitive streak, their
migration into the allantois, and then directly into the endoderm to
194
be embedded in the hind-gut epithelium (Anderson et al., 2000).
In addition, this method has allowed germ-cell puri®cation by
¯ow cytometry to be used to prepare cDNA and identify both
cell±cell and cell±matrix adhesion molecules expressed by germ
cells during and after migration. Using this method, functional
studies have shown that E-cadherin is required for compaction of
the germ cells into the genital ridges (reviewed in Wylie, 2000). A
mouse strain expressing enhanced GFP (EGFP) under chicken
beta-actin promoter showed that beta-actin is highly expressed in
spermatogonia and elongating spermatids, and that its expression
is regulated by FSH (Ventela et al., 2000).
Finally, the generation of transgenic animals has been made by
gene transfer into male germ line stem cells using a retroviral
vector. At least one in 300 stem cells could be infected. The
expression of the retrovirally delivered reporter lacZ transgene
persisted in the testis for more than 6 months. This study provides
a method to address the potential of human somatic cell therapy
DNA constructs to enter a patient's germ line, but also to
introduce accidentally permanent genetic changes by gene
therapy (Nagano et al., 2000).
Spermatogenesis revisited by means of knockout mice
To date, about 100 genes expressed in the male reproductive
system have been studied by genetic engineering (Table I). Many
genes found to be involved in spermatogenesis following genetic
engineering have already been described and listed in reviews
(Okabe et al., 1998; Escalier, 1999a; Lamb, 1999; Layman and
McDonough, 2000); those published later are presented in
Table II.
Spermatogenesis, a very sensitive function to genetic damages
Null mutation is a powerful method to reveal genes that can be
involved in failure of spermatogenesis since, in most cases, the
®nding of the involvement of a gene in spermatogenesis was
fortuitous and unexpected. Three examples can be given. The
casein kinase IIa¢ catalytic subunit (CkIIa¢ was known to be
expressed in brain. Mice with inactivation of the Csnk2a2 gene
(encoding CkIIa¢) were sterile due to severe anomalies of
spermiogenesis, while brain functions were apparently unaffected
(Xu et al., 1999). The zinc-®nger transcription factor Egr4 was
known to be expressed at high levels in the brain and not in other
tissues. Null mutants for Egr4 appeared neurologically normal,
but males were infertile due to an arrest of the early stages of
meiosis for most of spermatocytes and to premature germ cell
Genetic engineering and spermatogenesis
Table II. Spermatogenesis failure following genetic engineering in the mouse. (For data prior to February 1999, see Escalier, 1999a.)
Gene
±/±
Gja1 (Cx43)
BMP4±/±
b1 integrin±/±
Kit/SCF-R±/±a
INK4d±/±
Fanca±/±
Apaf-1±/±
GDNF+/±
(over)
Mvh±/±
Scp3±/±
Msh5±/±
Msh4±/±
TLS±/±
Tyro±/± Axl±/± Mer±/±
EGF (exo)
HSF1 (active form)
Insl3±/±
Osp/claudin-11±/±
PDGF-A±/±
Pp1cg±/±
Tnp 1 (TP1)±/±
FSHR±/±
Tarbp2±/±
E-MAP-115±/±
EGR4±/±
Jun D±/±
THEG (insertion)
cyp 19±/± (ArKO)
Csnk2a2±/±
Camk4±/±
Slc12a2±/± (NKCC1)
nectin-2±/±
c-ros±/±
Nek1(kat mutations)
HSL±/±
Cyritestin
Fertilin b±/±
Spermatogenesis anomaly
Reference(s)
primordial germ cell de®ciency
absence of germ cells
absence of gonad colonization by germ cells
early spermatogonial stem cell block
testis atrophy, germ cell apoptosis
testis atrophy, germ cell loss
spermatogonia degeneration
some spermatogonia depletion
spermatogonia accumulation and degeneration
zygotene step arrest, apoptosis
zygotene arrest
late zygotene arrest
zygotene arrest, chromosome
condensation and pairing failure
unpaired, mispaired spermatocytes
spermatocytes, spermatid degeneration
partial pachytene arrest, no spermatozoa
late pachytene step arrest and apoptosis
spermatocytes I arrest
spermatocyte lysis
spermatocyte arrest, absence of adult
Leydig cells
round spermatid arrested and polyploid
round spermatid depletion
spermatid chromatin condensation anomaly,
very poor sperm motility
hypospermatogenesis, cytoplasmic droplet,
reduced sperm chromatin compaction
abnormal sperm nuclear morphogenesis
manchette and nuclear shape abnormalities
¯agellar defects
sperm head misshapen;
¯agellum misassembled
spermatid arrest, head misshapen,
¯agellum misassembled
spermatid arrest, acrosomal abnormalities
(4.5 months±1 year)
sperm nuclear elongation defect
malformed and detached acrosome
abnormally heads and tail improper
attachment
nuclear shape and acrosomal anomalies
sperm nuclear and cytoskeletal anomalies
sperm tail angulation; unable to enter
the oviduct
testicular hypoplasia
oligospermia, Sertoli injuries
zona pellucida binding inability
reduced sperm-egg fusion
Juneja et al., 1999
Lawson et al., 1999
Anderson et al., 1999
Kissel et al., 2000
Zindy et al., 2000
Cheng et al., 2000
Honarpour et al., 2000
Meng et al., 2000
Tanaka et al., 2000
Yuan et al., 2000
Edelmann et al., 1999
Kneitz et al., 2000
Kuroda et al., 2000
Lu et al., 1999
Wong et al., 2000
Nakai et al., 2000
Nef and Parada, 1999
Gow et al., 1999
Gnessi et al., 2000
Varmuza et al., 1999
Jurisicova et al., 1999
Yu et al., 2000
Krishnamurthy et al., 2000
Zhong et al., 1999
Komada et al., 2000
Tourtellotte et al., 1999
Thepot et al., 2000
Yanaka et al, 2000
Fisher et al., 1998; Robertson et al.,
1999
Xu et al., 1999
Wu et al., 2000
Pace et al., 2000
Bouchard et al., 2000
Yeung et al., 2000
Upadhya et al., 2000
Osuga et al., 2000
Shamsadin et al., 1999
Cho et al., 1998, 2000
exo = exogenous expression of a gene from another species.
a
Point mutation.
death (Tourtellotte et al., 1999). Inactivation of the apoptotic
protease-activating factor-1 gene (Apaf-1) induced degeneration
of spermatogonia and, inversely, an excess of neuronal progenitor
cells (Honarpour et al., 2000).
In other cases, widespread disturbances were expected due to
the ubiquitous expression of the gene, but it was the testis that was
primarily affected. This was the case for the cyclin D-dependent
holoenzyme, INK4d, suspected to be a tumour suppressor but
whose Ink4d gene inactivation led to testicular atrophy and germ
cell apoptosis (Zindy et al., 2000). Similarly, the inactivation of
Slc12a2 (encoding the Na+-K+-2Cl± cotransporter NKCC1) did
not lead to kidney or blood pressure disturbances, but to failure of
spermatogenesis and inner-ear defects (Pace et al., 2000). Nectin2 is a component of adherens junctions, the expression of which is
ubiquitous. However, nectin-2±/± mice had no overt defects other
than infertility in males. Nectin-2 was found expressed in the
195
D.Escalier
testes only during the later stages of spermatogenesis and the
structural defects observed in spermatozoa of nectin-2±/± mice
suggested a role of the protein in sperm cytoskeletal organization
(Bouchard et al., 2000). The kit receptor tyrosine kinase was
known to have a role in proliferation, survival, adhesion, secretion
and differentiation. Point mutation in this gene had shown that it
is critical in spermatogenesis at premeiotic stages, and that other
factors compensate for haematopoiesis, melanogenesis and
primordial germ cell development (Kissel et al., 2000). Such
examples are numerous (see in Escalier, 1999a), and show that
many genes common to germ cells and somatic cells are critical
for spermatogenesis. This could also be the case for oogenesis,
since Cd9-de®cient mice exhibited only female sterility, although
Cd9 is known to participate in cell migration and adhesion (Kaji
et al., 2000). Finally, genetic engineering reveals that some genes
thought to be essential for female reproduction are more critical to
spermatogenesis. Dax1 was considered as an ovarian-determining
gene, but Dax1-deleted mice showed testicular epithelium
degeneration and normal ovarian development (Yu et al., 1998).
Conversely, genetic engineering has revealed that some gene
products thought to be determinants for spermatogenesis or
fertilization have, in fact, a less important function. This was the
case following proacrosin gene inactivation, as this did not lead
to sterility or sperm phenotype anomalies but only to a delay in
fertilization (Baba et al., 1994; Adham et al., 1997). Also,
inactivation of the genes of the transition proteins (Tp) involved in
the replacement of histones during sperm chromatin compaction
did not lead to sterility of Tp2±/± mice or impaired only partially
the male fertility of Tp1±/± mice (Yu et al., 2000) (see also Gene
redundancy). Xist is implicated in female X-inactivation and
thought to be involved in X inactivation during meiosis. However,
Xist-de®cient mice did not exhibit testis anomalies and were fully
fertile, demonstrating that Xist is not implicated in the reversible
XY inactivation in spermatocytes (Marahrens et al., 1997).
Mesothelin (a glycoprotein that is highly expressed in mesothelial
cells) is expressed in many tissues, including the testis.
Unexpectedly, null mutant mice for this gene developed normally
and produced offspring normally (Bera and Pastan, 2000). A
widely proposed model suggested that fertilin promotes spermegg fusion. In mice lacking the fertilin b-subunit, fertilin a is also
absent from mature spermatozoa. Nevertheless, sperm lacking
both fertilin subunits can fuse with eggs at 50% of the wild-type
rate. Data suggested that other gamete surface molecules act to
promote membrane fusion, and that fertilin's role is in
spermatozoon±egg plasma membrane adhesion (Cho et al., 2000).
Very sensitive steps: germ cell proliferation and survival and
meiotic prophase I
Primordial germ cell and spermatogonia
Knockout mice are also models used to study several factors
involved in the migration (Anderson et al., 2000) and maintenance of germ cells. Bmp4 null mutant embryos contain no PGC
and lack an allantois, showing that BMP4 is a cell signalling
factor required for the formation of the germline during
gastrulation. Beta-galactosidase activity in Bmp4 (lacZneo)
embryos revealed that before gastrulation, Bmp4 is expressed
®rst in the extraembryonic ectoderm and later in the extraem-
196
bryonic mesoderm. Bmp4 extraembryonic ectoderm expression
regulates the formation of allantois and PGC precursors and the
size of the founding population of PGC (Lawson et al., 1999).
Bmp8b is found to be expressed in the extraembryonic ectoderm
in pregastrula and gastrula stage mouse embryos. The absence of
PGC in 43% of Bmp8b null mutants showed that Bmp8b is
required for PGC generation (Ying et al., 2000). Besides, integrin
b1±/± PGC have shown that b1 integrin is required for gonad
colonization by germ cells (Anderson et al., 1999).
Knockout mice have shown that Tiar (Beck et al., 1998) and
p53 (Matsui et al., 2000) are involved in regulation by apoptosis
of maintenance of fetal germ cells. Tiar is a RNA-binding protein
whose role might be in the expression of a survival factor or
survival factor receptor that is essential for PGC development
(Beck et al., 1998). Both body growth and the number of PGC can
be affected by inactivation of Tiar (Beck et al., 1998) and Zfx
genes (Luoh et al., 1997). The growth factor gene GDNF (Meng
et al., 2000) and the TR gene encoding the telomerase
holoenzyme TR (Lee, H.-W. et al., 1998) play a role in
spermatogonia proliferation. GDNF is involved in DNA synthesis
in spermatogonia, and is expressed by Sertoli cells, while the
GDNF receptor and the Ret receptor tyrosine kinase are expressed
in testicular germ cells. GDNF interacts with membrane receptors
which mediate stimulation of the Ret receptor tyrosine kinase
(Viglietto et al., 2000). Dazla is essential for the survival of
spermatogonia (Ruggiu et al., 1997). Germ cell proliferation and
initiation of spermatogenesis are under the control of the
transforming growth factor gene BMP8b (Zhao et al., 1996),
while the mouse homologue of Drosophila Vasa (Mvh) operates
on the proliferation and differentiation of male germ cells (Tanaka
et al., 2000). Bcl-2 family members (Knudson and Korsmeyer,
1997) are involved in control of the number of spermatogonia (see
Escalier, 1999a). The localization of Bcl-x(l), the long transcript
of Bcl-x, suggests that it may regulate germ cell density, possibly
in cooperation with the apoptotic inducer Bax (reviewed in
Beumer et al., 2000). Mice containing two copies of a
hypomorphic allele of Bcl-x lacked spermatogonia and were
sterile, the primordial germ cells being depleted by day 15.5
embryos. The loss of Bcl-x function in the hypomorph was
corrected by deletion of both copies of the bax gene, showing that
the balance of Bcl-x and Bax control PGC survival and apoptosis
(Rucker et al., 2000).
Primary spermatocytes
Primary spermatocytes are very sensitive to genetic manipulation,
particularly when genes are involved in recombination. In most
cases this leads to meiotic arrest that reveals the existence of
meiotic checkpoints for DNA damage in mammals, particularly in
primary spermatocytes at the pachytene step (reviewed in
Escalier, 1999b). Other meiotic prophase I stages can be affected
by a null mutation, as found in null mutant mice with an arrest at
the leptotene step for Atm (Barlow et al., 1998), at the zygotene
step for Dmc1 (Pittman et al., 1998; Yoshida et al., 1998), Scp3
(Yuan et al., 2000), Msh5 (Edelmann et al., 1999), Msh4 (Kneitz
et al., 2000) and Mvh (Tanaka et al., 2000), and at the diplotene
step for cyclin A1 (Liu et al., 1998). Inactivation of DNA
mismatch repair genes, thought to be involved in recombination,
can lead to various spermatogenesis impairments. Mlh1-de®cient
mice showed meiotic arrest (Edelmann et al., 1996), while Pms2-
Genetic engineering and spermatogenesis
de®cient mice could produce spermatids, although abnormal
(Baker et al., 1995).
Spermatocytes arrested in the meiotic prophase were targets for
apoptotic events leading to their elimination and seminiferous
tubule depletion. At advanced stages, Sertoli cells showed
vacuolation or sloughing, demonstrating the importance of
somatic and germinal cell interactions for their survival (review
in Escalier, 1999a). The in¯uence of Sertoli cells on germ cell
survival has been shown by inactivation of three Sertoli receptor
genes of the Tyro3 family (Tyro 3, Axl, Mer). These Sertoli cell
receptors are normally expressed during postnatal development,
and their ligands (related to sex hormone-binding globulin) are
expressed by Leydig cells before sexual maturity. Mice lacking
any single receptor, or any combination of two receptors, were
fertile while male triple mutants were sterile. They exhibited germ
cell death which did not result from the death of Sertoli cells. Data
showed that Tyro 3 family receptors and their ligands might act as
essential regulators of Sertoli-cell function. The combined
activation of the receptors in Sertoli cells may be required for
the production of germ-cell trophins and thus to the role of Sertoli
cells as nurturers of developing germ cells (Lu et al., 1999). The
in¯uence of Sertoli cells on spermiogenesis is shown in Rxrb
(retinoid 3 receptor) mutant mice. Rxr is normally expressed in
Sertoli cells and, in Rxrb null mice, Sertoli cells accumulated
unsaturated triglycerides while spermatid release did not occur.
The epididymis contained few spermatozoa with abnormal
acrosomes and tails. In old mutant males, progressive degeneration of the germinal epithelium occurred, ending with acellular
lipid-®lled tubules (Kastner et al., 1996).
Another main gain of genetic engineering is knowledge of the
importance in germ cells of tumour suppressor genes, such as p53,
and of genes involved in apoptosis induction, such as Bax, or
involved in protection against apoptosis, such as members of the
bcl2 family (Escalier, 1999a; Print and Loveland, 2000). Of
interest is the ®nding that p53 promotes apoptosis of fetal
testicular cells only after day 16.5 postcoitum, but not before
(Matsui et al., 2000) (see also section, Diseases and male
infertility). How the Bcl-2 gene and its pro-survival relatives
prevent activation of the cytoplasmic proteases (called caspases)
that mediate apoptosis is unknown. Bcl-w is an important prosurvival protein that participates in the regulation of apoptosis by
binding pro-apoptotic factors Bax and Bak (Yan et al., 2000). The
apoptotic protease-activating factor-1 (Apaf-1) is a key trigger of
apoptotic events through cytochrome c-mediated apoptosis. The
complex composed of Apaf-1, dATP and cytochrome c activates a
series of caspases, leading to apoptotic cell death. Degeneration of
spermatogonia resulting in the virtual absence of spermatozoa in
surviving Apaf-1 null mutants suggested that alternative apoptotic
pathways work in conjunction with and parallel to Apaf-1 and can
modify its effect on programmed cell death in testis (Honarpour et
al., 2000).
Pachytene spermatocyte: a target to thermal injury response
Another aspect of data obtained by genetic engineering is related
to the vulnerable nature of spermatogenesis to thermal insult.
Heat shock induces activation of heat shock transcription factor
(HSF), which regulates expression of heat shock genes, the major
products of which act as molecular chaperones by facilitating
protein folding and assembly (Hartl, 1996). HSF1 is a major heat
stress-responsive factor expressed ubiquitously to protect cells.
Mice expressing the active form of HSF1 (Nakai et al., 2000)
were infertile as a result of blockage of spermatogenesis at the
spermatocyte I stage. Spermatocyte apoptosis appeared by 3
weeks. At 4 weeks, 25±30% of the seminiferous tubules contained
clusters of apoptotic cells compared with 30±35% after mild heat
exposure (43°C for 15 min). A much lower cyclin A1 level than
that in wild-type mice was found (cyclin A1 normally increases in
late pachytene and reaches a peak in the diplotene stage). The
transgene was found to be highly expressed in spermatogenic
cells, but not in Sertoli cells or interstitial Leydig cells. HSF1
would be a major trigger for the induction of apoptosis of germ
cells. Late pachytene spermatocytes were revealed to be target
cells for HSF1-induced cell death. It was suggested that HSF1 is a
good candidate for acceleration of apoptosis of germ cells because
it can sense thermal stress directly as well as indirectly. In isolated
pachytene spermatocytes, HSF1 was activated by a lower
temperature threshold than somatic cells in the testis. HSF1
active form expression by transgenesis did not affect cell growth,
differentiation or deathÐan important ®nding considering the
therapeutic use of heat shock response. Another important
repercussion of these data should be in the ®eld of cryptorchidism,
because experimental cryptorchidism induces apoptosis of almost
the same subset of cells (Nakai et al., 2000).
It can be noted that the function of molecular chaperone
assumed by heat shock proteins may be used for other functions
than thermal injury during spermatogenesis. This is well
illustrated by Hsp70-2-de®cient mice, showing that HSP70-2 is
required for assembly of a functional CDC2/cyclin B1 complex in
pachytene spermatocytes, and thus that HSP70-2 plays a role in
synaptonemal complex desynapsis which necessitates phosphorylation of the synaptonemal protein-1 (SCP1) by CDC2 (Zhu et
al., 1997; for review, see Escalier,1999a).
Unexpected sex-speci®c factors
The use of knockout mice allows a list to be compiled of genes
involved in meiotic recombination and chromosome pairing. As
expected, many genes involved in recombination are common to
both sexes, such as Dmc1 (Pittman et al., 1998; Yoshida et al.,
1998), Msh5 (Edelmann et al., 1999) and Atm (Barlow et al.,
1996; Xu et al., 1996). Moreover, some of the genes are also
involved in somatic recombination, such as Mlh1 and Atm. It is
noteworthy that there are sex-speci®c factors for apparently
similar events. The products of the DNA mismatch repair genes
Mlh1 and Pms2 are known to form heterodimers. Inactivation of
Pms2 has led to meiotic arrest in the male only, although Pms2
might be involved in DNA mismatch repair in a variety of tissues
(Baker et al., 1995). In contrast, inactivation of Mlh1 (Baker, et
al., 1996; Edelmann et al., 1996) and Msh4 (Kneitz et al., 2000)
has led to meiotic arrest in both sexes (Edelmann et al., 1996).
Chromosome pairing is also related to sex-speci®c factors. SCP3
is a major component of the synaptonemal complexes, and Scp3
inactivation disrupted only male meiosis (Yuan et al., 2000). The
same phenomenon is found for the Xlr family whose members are
expressed in lineages undergoing recombination (Escalier et al.,
1999). Xmr is the testicular member of this family that is
expressed in spermatocytes during the meiotic recombination
(Calenda et al., 1994). Unexpectedly, it is the lymphoid Xlr
member, and not Xmr, that it is expressed during the female
197
D.Escalier
meiotic recombination (unpublished data). This fact supported
data suggesting that Xmr could be involved in male Xchromosome inactivation in mammals (Escalier and Garchon,
2000).
A similar divergence in germ cell processes between sexes is
also observed at earlier stages, as seen following inactivation of
the cKit receptor/stem cell factor (kit/SCF-R) gene leading to only
male infertility (Blume-Jensen et al., 2000). By contrast, point
mutation in the binding site for phosphatidylinositol 3'-kinase in
the kit gene reduced female fertility due to development of
ovarian cysts and ovarian tubular hyperplasia, while spermatogenesis was arrested at the premeiotic stages, leading to male
sterility (Kissel et al., 2000). Also a targeted mutation of the
mouse Vasa homologue gene (Mvh) affected only male
reproduction, although Mvh expression was ®rst detected in
primordial germ cells after colonization of the genital ridges
(Tanaka et al., 2000) and in germ cells undergoing gametogenic
processes in both the male and female (Toyooka et al., 2000). On
the contrary, both sexes were affected in other genes involved in
spermatogonia maintenance such as Tiar, Tr (telomerase) and
Dazla (see Escalier, 1999a).
Functional analysis of genes involved in sperm chromatin
compaction
Many germ cell differentiation events can also be shown by
mouse models. An example is histone replacement by transition
proteins (TP) and protamines during spermiogenesis (Baarends et
al., 1999; Zhong et al., 1999). It has been shown that the doublestranded RNA binding protein PRBP (encoded by the gene
Tarbp2) is required for correct translational activation of the
mRNAs encoding the protamines. Disruption of the Tarbp2 gene
has led to sterility and oligospermia. A failure to synthesize the
protamines resulted in delayed replacement of TP. Spermatids
showed abnormal nuclear morphogenesis, developmental arrest
and degeneration. PRBP is suspected to be a chaperone in the
assembly of speci®c, translationally regulated ribonucleoproteins
(Zhong et al., 1999).
Pre-existing protamine-1 (Prm1) mRNAs are stored for several
days, and their temporal translation or messenger RNA stability
are under the control of regulatory elements located in the 3¢
untranslated region (3¢UTR) of mRNAs. Prm1 mRNA is stored in
spermatids as a cytoplasmic ribonucleoprotein particles that
contain Y box proteins. Y box family members are nucleic acid
binding proteins that can bind to the Prm1 3'UTR in vitro. Data
suggest that Y box proteins could function as translational
repressors or could protect mRNAs from degradation during their
extended period of storage (reviewed in Braun, 2000).
Premature translation of Prm1 mRNA caused precocious
condensation of spermatid nuclear DNA and abnormal head
morphogenesis. Round spermatids were arrested and intercellular
bridges were relaxed, leading to multinucleated cells. The
incomplete processing of protamine-2 protein suggested that its
processing is coupled with protamine-2 codeposition with
protamine-1 (Lee et al., 1995).
The sequential deposition of sperm basic nuclear proteins on
chromatin was disrupted in Camk4±/± mice which had sperm
counts less than 4% of those of wild-type males and abnormal
spermatozoa with failure to develop hook-shaped heads and
improper attachment of the ¯agellum. Camk4 is the Ca2+/
198
calmodulin-dependent protein kinase IV implicated in transcriptional regulation and associated with chromatin and nuclear
matrix. Step-15 spermatids in Camk4±/± mice showed loss of
protamine-2 and prolonged retention of TP2. However, transcription of Prm2 was unaffected, suggesting that protamine-2 was
prematurely degraded. Protamine-2 was phosphorylated by
Camk4 in vitro, indicating a connection between Camk4
signalling and the exchange of basic nuclear proteins in male
germ cells. The results suggested that successful interaction of
mature protamine-2 with chromatin is required for displacement
of TP2 (Wu et al., 2000).
In vitro, transition protein 1 (TP1) reduces the interaction of
DNA with the nucleosome core, while transition protein 2 (TP2)
compacts the DNA in nucleosomal cores. Mice lacking the major
TP1 have been obtained following targeted deletion of the Tnp1
gene. Only 60% of Tnp1-null males were infertile, and the fertile
Tnp1±/± mice showed a decrease in the number of litters produced.
Sperm production was normal in Tnp1±/± mice, but only 23% of
the spermatozoa showed any movement, and most of those did not
show forward progression. Abnormal rod-shaped chromatin
condensation units appeared in the nuclei of condensing
spermatids at step 12, but all the chromatin condensed from step
14. Sperm heads with a blunted or bent tip were seen in 16% of
epididymal spermatozoa, possibly generated by the abnormal
chromatin condensation that could reduce the rigidity of the ®ne
apex of the spermatozoon. In Tnp1±/± mice, TP2Ðindividually or
in concert with the precursor of protamine-2 (pP2)Ðwere able to
initiate chromatin condensation, and these proteins completely
replaced histone without involvement of TP1. Data suggested that
the signals that normally activate TP1 translation can, in the
absence of TP1, act on TP2 and P2 mRNA and that TP1 is
functionally related to TP2 and the protamines (Yu et al., 2000).
Moreover, most Tnp2±/± male mice had normal numbers of
spermatozoa and normal fertility, although the litter size was
reduced and some spermatozoa presented blunt-tipped heads (Yu
et al., 2000). In Tnp2 mutants, protamine-2 precursors were found
in epididymal spermatozoa, and no changes were detected in TP1
concentrations (Yu et al., 2000).
Tnp1 contains a cAMP-responsive element (CRE) that serves
as a binding site for the CRE modulator CREM. CREM is
involved in the regulation of Tnp1 gene expression, and human
CREM protein is synthesized in steps 1±3 round spermatids. This
could explain why a reduction of Crem expression and a lack of
both CREM and TP1 were found in human arrested spermatids at
step 3 (Steger et al., 1999), but it is unclear whether this anomaly
is secondary or not to the arrest. Mice deleted in Crem presented a
spermatogenesis arrest at the round spermatid step (reviewed in
De Cesare et al., 1999). Recent data have shown that CREM
requires a tissue-speci®c coactivator, ACT (Activator of CREM in
Testis) to elicit its regulatory function in the testis (Sassone-Corsi,
2000). Disruption of mHR6B normally involved in histone 2A and
2B ubiquitination for their degradation did not lead to blockage of
the synthesis of TP, but to an anomaly of their distribution leading
to abnormally shaped sperm heads (Roest et al., 1996).
The reduction in size of the testis, seminal vesicles and
epididymis found in follicle-stimulating hormone receptor knockout (FORKO) mice that lack the FSH receptor gene (FSHR) could
be due to a decrease in the level of testosterone. The 23% increase
in propidium iodide stainability of elongated spermatids and the
Genetic engineering and spermatogenesis
increased sperm head size in FORKO mice suggested a
disturbance in the normal replacement of histones by protamines
during spermiogenesis, leading to poor condensation of spermatid
nuclei. The decreased motility was associated with a bent tail, and
80% of the ¯agella had retention of the cytoplasmic droplet
(Krishnamurthy et al., 2000).
A disturbance of protamine deposition in mice expressing the
galline protamine led to heterogeneous chromatin and instability
of the head-¯agellar attachment (Maleszewski et al., 1998) (for
the examples in this paragraph, see Escalier, 1999a).
Taken together, these data show that disturbances of expression
timing of either TP or protamines lead to spermatid arrest. It is
noteworthy that anomalies of the spermatid chromatin compaction result in ¯agellar anomalies, showing the importance of
nuclear integrity on the ¯agellar anchorage. That the ¯agellar
anomalies result from defects in the shape of the implantation
fossa did not seem to be the explanation in all cases, suggesting
that disturbances of the chromosome territories and/or the
nucleoskeleton might also be involved.
Unexpected ¯agellar morphogenetic factors
Knockout mice help to detect factors involved in the organization
and reorganization of the cytoskeleton during spermiogenesis.
Nectin-2 is a component of cell±cell anchoring junctions
(adherens junctions) and is linked to F-actin through the actin
®lament-binding protein l-afadin. Anchoring junctions connect
the cytoskeletal elements of neighbouring cells, and are also
believed to regulate cell shape and differentiation through
signalling pathways. Nectin-null mutant males produced morphologically aberrant spermatozoa with defects in nuclear and
cytoskeletal morphology and in mitochondrial localization.
Sperm nuclei were irregularly shaped, with translucent vesicles
and indentations. The formation of a tightly packed helical sheath
of mitochondria was impaired, and mitochondria were frequently
present alongside the nucleus. The outer dense ®bres were
jumbled and extended into the head. While F-actin was found to
be prominent in the middle piece in wild-type spermatozoa, in
nectin±/± mice, F-actin was mainly present in the head and poorly
present in the middle piece. It was suggested that nectin-2 on the
middle piece is required for proper localization of F-actin,
through interaction with l-afadin. Nectin-2 might bring the lafadin±F-actin complex to the middle piece and mediate structural
changes of actin. These changes may be important in nuclear
shaping, mitochondrial relocation and dense ®bre organization
during spermiogenesis (Bouchard et al., 2000).
JunD contributes to the AP-1 transcription factor complex
whose function is complex, including tumour suppression. JunDde®cient mice presented anomalies of both spermatogenesis and
mating behaviour, suggesting that functions of JunD dimers in the
pituitary and the testis are essential to maintain normal testis
function. A speci®c block in spermiogenesis was observed in
homozygous JunD±/±, and an absence of ¯agella in the lumen of
the seminiferous tubules correlated with a marked decrease in the
number of late spermatid heads located near the lumen. There
were two classes of sperm phenotypes, either asthenoteratozoospermia or asthenozoospermia alone. In the ®rst case, the sperm
head showed an abnormal hammer-like shape, and misassemblage
of the microtubules and of the periaxonemal structures of the
¯agellum. When the ¯agellum merged out of the cell cytoplasm,
the periaxonemal cytoplasm bulged out, forming irregular
cytoplasmic masses (TheÂpot et al., 2000). Therefore, the absence
of JunD led to sperm ¯agellar growth impairment and, as in other
tissues, JunD is thought to be a growth inhibitor.
Ki/ki (kisimo) mice had disruption of THEG (testicular haploid
expressed gene) by insertional mutation. THEG is expressed in
round and elongated spermatids, and needs Sertoli cells for its
continued expression in spermatids (Nayernia et al., 1999). Ki/ki
mice had virtually no spermatozoa, and the spermatids exhibited
misshapen nuclei. At the ¯agellar level, microtubules and coarse
®bres were arranged in a whirl. THEG interacts in vitro with
chaperin containing TCP-1 (t-complex poly peptide Ie) (CCTe),
which is known to be required for the correct folding or assembly
of cytoskeletal proteins. Data have suggested that abnormal or
absent ¯agella in ki/ki mice may be due to impairment of the
assembly of cytoskeletal proteins such as the tubulins (Yanaka et
al., 2000).
Homozygous c-ros knockout mice are sterile, and the
epididymal spermatozoa exhibit bent tails. After mating, 46±
86% of knockout spermatozoa in the uterus were bent at the
cytoplasmic droplet even when motile, and motile spermatozoa
showed a reduction in their straightline and curvilinear velocities.
No spermatozoa were recovered from the oviduct or observed
within the uterotubal junction. The infertility of c-ros knockout
male mice can be explained by the inability of the spermatozoa to
enter the oviduct, as a result of their bent tails and their
compromised ¯agellar vigour within the uterus (Yeung et al.,
2000).
Discovery of other genes involved in spermiogenesis
Null mutants for the zinc-®nger transcription factor Egr4
presented a maturation arrest with 70% of spermatocytes undergoing apoptosis, and these results suggested that spermatocytes
required Egr4 to transgress the early to mid-pachytene stage of
meiotic prophase. The maturation arrest was incomplete, and
some Egr4-de®cient animals exhibited spermatozoa with heads
that were either separated entirely or bent sharply back on the
¯agella. In addition, the ¯agella were often fragmented, sharply
kinked, or had tightly coiled distal ends. Widespread alterations in
testicular gene expression were found, although no de®nitive
target genes were identi®ed to account for these anomalies. Crem
and A-myb were markedly up-regulated in Egr4-null testis, as
were several genes that participate in steroidogenesis or steroid
signalling. These alterations possibly re¯ected compensatory
responses to ineffective pachytene spermatocyte maturation and
inadequate levels of spermatogenesis (Tourtellotte et al., 1999).
Null mice in the Sla12a2 gene (normally expressing the Na++
K -2Cl± co-transporter NKCC1) had delayed formation of the
lumen of the seminiferous tubules. By 1.5 months of age, marked
abnormalities in spermatogenesis were observed and cellular loss
increased with advancing age. Few spermatids were present, but
defects were particularly striking when spermatids gradually
acquired the polarized shape and structural features of spermatozoa. Spermatids showed anomalies of the cap-phase acrosomal
vesicle and of the nuclear shape. Acrosomal defects included
abnormal placement of the acrosomal granule and vesicular
structures resembling two acrosomes at opposite poles of one
nucleus. Normally, the seminiferous tubule ¯uid contains 10-fold
higher concentrations of K+, and 10-fold lower concentrations of
199
D.Escalier
Table III. Various other disorders of the reproductive functions after gene inactivation
Gene
Anomalies
Reference(s)
mOvo1
IGF1
FSHR
urogenital defects
vestigial ducts, seminal vesicles and prostate
smaller epididymis and seminal vesicles;
reduced testicular weight; follicular
development arrest; vagina imperforate,
atrophic uterus
efferent ductule ¯uid reabsorption failure
smaller epididymis
epididymal degeneration
cryptorchidism; deregulation oestrus cycle
failure initial epididymis segment
lack of genital ridges and gonads
failure of sperm-egg fusion
premature arrest of female fertility
block in folliculogenesis (early antral stage)
impaired fertility; reduced primary follicles
ovulation of pre-meiotic oocytes
lack of embryo implantation
seminal vesicles and prostate anomalies
embryo implantation defects
ovarian teratomas
ovary sex reversal
uterus disruption (epithelium, glands)
atretic follicles; external genitalia, uteri,
mammary gland underdevelopment
defective decidualization
absence of an allantois
reduced placenta;
de®cit in mounting capacity
breast underdevelopment; impaired nursing
developmental impairment
embryonic lethality
embryonic lethality
defects in mating behaviour
defects in mating behaviour
Dai et al., 1998
Baker, J. et al., 1996
Krishnamurthy et al., 2000;
Abel et al., 2000
ER-a
E-MAP-115
Bmp8b
Insl3
c-ros
Lhx9
Cd9
Fanca
Inhibin a (over)
Bcl-x (hypomorphic allele)
PLR
basigin
Mos
ab ER
centromere protein B
cyp19
interleukin-11
BMP4
c-fos
A-myb
cyclin B1
Rad 51
BLM
Jun D
EGF (exo)
Hess et al., 1997
Komada et al., 2000
Zhao et al., 1998
Zimmermann et al., 1999; Nef and Parada, 1999
Yeung et al., 1999
Birk et al., 2000
Kaji et al., 2000
Cheng et al., 2000
Pierson et al., 2000
Rucker et al., 2000
Gof®n et al., 1998
Steger et al., 1998
Igakura et al., 1998
Hirao and Eppig, 1997
Couse et al., 1999
Fowler et al., 2000
Fisher et al., 1998
Robb et al., 1998
Lawson et al., 1999
Johnson et al., 1992
Baum et al., 1994
Toscani et al., 1997
Brandeis et al., 1998
Tsuzuki et al., 1996
Chester et al., 1998
Thepot et al., 2000
Wong et al., 2000
ER = oestrogen receptor; exo = exogenous expression of a gene from another species.
Na+ than does blood plasma, and is thought to play a vital role in
normal germ cell development and Sertoli cell function. It was
suggested that K+ can be pumped into the Sertoli cell from the
interstitial ¯uid by an active process, probably involving a Na+,
K+ translocating ATPase, and that NKCC1 transports K+ at the
basolateral surface of the Sertoli cells (Pace et al., 2000).
Protein kinase casein kinase II (Ck2) is a cyclic-AMP and
calcium-independent serine-threonine kinase. Ck2 has been
implicated in DNA replication, regulation of transcription,
translation and control metabolism. Ck2 has been found to be
associated with the nuclear matrix, and also with chromatin itself.
Mice de®cient for Csnk2a2 encoding the casein kinase IIa¢
catalytic subunit (CkIIa¢) showed anomalies of spermatid nuclear
morphogenesis with nuclear and acrosomal shaping anomalies.
Elongating spermatids developed abnormalities of the anterior
head, and the nuclear and acrosomal morphology became
increasingly deformed, with many acrosomes becoming detached
from the nucleus or exhibiting an excrescence. In the epididymis,
sperm heads were round or ovoid. It was suggested that CkIIa¢
may play a role in regulating the development of the nuclear
200
architecture, and that CsnK2a2 could be a candidate globozoospermia gene. This study was the ®rst case in which a unique role
for one of the isoforms of Ck2 was demonstrated (Xu et al.,
1999).
Aromatase enzyme cyp19 converts C19 steroids (androgens) to
C18 steroids (oestrogens), and is active in Sertoli cells and Leydig
cells. Aromatase is present in spermatocytes and spermatids.
ArKO mice with targeted disruption of cyp19 were initially
fertile, but developed Leydig hyperplasia/hypertrophy and
progressively disrupted spermatogenesis, despite no decrease in
gonadotrophins or androgens. Female ArKO mice also showed a
progressive phenotype. Spermatogenesis disruption was evident
from 18 weeks of age and affected all 1-year-old ArKO mice.
Spermiogenesis was arrested at early stages in ArKO mice. Round
spermatids did not complete elongation and spermiation, and
degenerating and multinucleated round spermatids were seen in
tubules exhibiting spermiogenic arrest. Abnormal acrosomes
often were observed in which uneven spreading over the nuclear
envelope was apparent, and more than one acrosomal granule was
frequently visible. The phenotype was different from ERKO mice
Genetic engineering and spermatogenesis
Table IV. Genes involved in spermatogenesis and tumour susceptibility
Gene de®ciency
Tumour susceptibility
Reference(s)
N-ras
P53
mammary and salivary glands tumours
lymphoma, sarcoma, adenoma
Leydig cell tumour, gonadoblastoma
chromosome loss/duplication, interstitial deletion
human Bloom syndrome
thymic lymphomas
increased incidence of malignancies
various tumours
ovarian and testicular tumours
ovarian teratomas
lymphomas; intestinal adenomas;
intestinal adenomas; adenocarcinoma
lymphomas; sarcomas
telomerase teratocarcinomas (germ cells)
lymphomas, carcinomas
known to be involved in human
liposarcomas and leukaemias
testicular tumours
Mangues et al., 1990
Donehower et al., 1992
BLM
Atm
FAC
E2F-1
Cyclin D2
Mos
Mlh1
Pms2
Tls (RNA-binding)
GDNF (over)
Shao et al., 2000
German, 1993
Barlow et al., 1996; Xu et al., 1996
Whitney et al., 1996
Yamasaki et al., 1996
Sicinski et al., 1996
Hirao and Eppig, 1997
Prolla et al., 1998
Prolla et al., 1998
Rudolph et al., 1999
Kuroda et al., 2000
Meng et al., 2000
over = overexpression.
that lack ERa and exhibit disruption of spermatogenesis caused
by abnormal ¯uid reabsorption. These ®ndings suggest that
oestrogen plays a hitherto unsuspected role in spermatid
differentiation and spermatogenesis (Robertson et al., 1999).
Use of the gene trap screen has allowed the isolation of
ROSA63 mutant mice exhibiting a phenotype similar to those in
vitamin A-de®cient animals and retinoic acid receptor a (RARa)
knockout mice. ROSA63 mice shared a mutation in the gene
encoding an epithelial, microtubule-associated protein of 115 kDa
(E-MAP-115), that is highly expressed in testis and kidney and
whose expression is induced by retinoic acid. E-MAP-115 is a
MAP implicated in stabilizing and reorganizing microtubules
(MT), and is found in the perinuclear subdomain of MT in
cultured cell lines of epithelial origin. In ROSA63 mutant mice,
the testes and epididymis were smaller. As early as the ®rst wave
of spermatogenesis, all condensed spermatids demonstrated
abnormal shape, underwent progressive degeneration, and were
phagocytosed by Sertoli cells. Spermatids were ®rst lost by 3
months of age, followed by loss of spermatocytes and
spermatogonia by 12 months. The nuclear shape was slightly
abnormal at steps 8 and 9, and became more abnormal in mid-step
9±11. About 80% of spermatid sections showed anomalies of the
microtubular manchette (absent, reduced, ectopic). Regions of the
spermatid nucleus without manchette were rounded, and regions
of the nucleus impacted by ectopic manchette were indentedÐa
phenotype that could be compared with azh mutants. By contrast,
the ¯agella appeared normal. Fewer MT bundles were seen in
Sertoli cells, and these MT bundles were thinner and poorly
developed. These data suggested that impaired MT functions in
the Sertoli cells in the absence of E-MAP-115 led to depletion of
germ cells in the mutant. E-MAP-115 could also be required in
the Sertoli cells for secreting proteins necessary for survival and
differentiation of germ cells (Komada et al., 2000).
Impairment of spermiogenesis beginning at the round spermatid stage and production of polyploid spermatids was seen after
disruption of the protein phosphatase catalytic subunit Pp1cg
(Varmuza et al., 1999). Moreover, spermatids presented high rates
of DNA fragmentation, as are found in spermatids from men with
non-obstructive azoospermia (Jurisicova et al., 1999).
What about the blood±testicular barrier?
In this ®eld, the ®ndings are surprising. The inactivation of the
Osp/claudin-11 gene, a component of the Sertoli cell-occluding
junctions, led to Sertoli cell release. It was expected that this
disruption of the blood±testicular barrier could lead to an
autoimmunity process, but neither blood cells nor in¯ammation
were found in testes of these mice (Gow et al., 1999).
Misexpression of the human interleukin-2 gene in mice has led
to lymphocyte in®ltration in cerebellar tissues and skin, but not in
testes (although these testes were affected, as seen by the
depletion of later stages of spermatogenesis; Ohta et al., 1990).
Other data relative to reproduction
Some knockout mice presented anomalies of different components of the male reproductive system (Table III; see also
Nishimori and Matzuk, 1996). Mice de®cient for the growth
factor Bmp8b showed germ cells and epididymal epithelium
degenerations (Zhao et al., 1998). Mice with a targeted mutation
of the insulin-like growth factor gene Igf1 had reduced
spermatogenesis at 18% of the normal level and vestigial ducts,
seminal vesicles and prostate (Baker, J. et al., 1996). In mice
de®cient for the nuclear ERa, the testes were atrophic and the
efferent ductules showed defective ¯uid reabsorption, leading to
sperm dilution and infertility (Hess et al., 1997). Insl3, a member
of the insulin-like hormone superfamily, was seen to be expressed
201
D.Escalier
in Leydig cells and postnatal ovary. Inactivation of Insl3 led to
bilateral cryptorchidism (Zimmermann et al., 1999) and deregulation of the oestrus cycle with subsequent female infertility (Nef
and Parada, 1999). Homozygous c-ros knockout male mice
presented an epididymal anomaly and angulation of the sperm
¯agella due to an impaired volume regulation in the epididymis
(Yeung et al., 1999).
Other studies not directly related to spermatogenesis, but
nevertheless concerning reproduction, are worth mentioning.
Transgenic mice overexpressing Dax1 have contributed to
knowledge on the factors involved in sex determination (Hanley
et al., 2000). Dax1 is responsible for dosage-sensitive sex
reversal. XY mice carrying extra copies of mouse DAX1 as a
transgene show delayed testis development when the transgene is
expressed at high levels. Complete sex reversal occurs when the
transgene is tested against weak alleles of the sex-determining Ychromosome gene Sry (Swain et al., 1998). Mice lacking the LIM
homeobox gene Lhx9 failed to develop genital ridges and gonads
failed to form. Genetically male mice were phenotypically female
due to an absence of anti-MuÈllerian hormone and testosterone. It
was suggested that some human gonadal agenesis can be related
to Lhx9 mutations (Birk et al., 2000). Cd9, a member of the
transmembrane-4 superfamily, interacts with integrin proteins and
possibly participates in cell migration and adhesion. Cd9 is
present on egg microvilli and, in its absence, females were
infertile due to a failure of sperm-egg fusion while sperm-egg
binding was normal (Kaji et al., 2000). A null mutation in
Basigin, an immunoglobulin superfamily gene member, affected
embryo implantation and male meiosis (Igakura et al., 1998).
Mice with a targeted disruption of the prolactin gene shared male
and female reproduction anomalies. Half of the homozygous
males were infertile or showed reduced fertility (Gof®n et al.,
1998), and the growth of seminal vesicles and ventral prostate was
affected (Steger et al., 1998). Homozygous females were infertile
as a result of multiple reproductive abnormalities, including
ovulation of premeiotic oocytes, reduced pre-implantation oocyte
development, lack of embryo implantation, and absence of
pseudopregnancy (Gof®n et al., 1998). Mos-de®cient mice
developed ovarian teratomas that possibly were derived from
parthenogenetically activated oocytes undergoing early embryonic development within the ovaries (Hirao and Eppig, 1997). Null
mice for the centromere protein B gene exhibited testis weight
reduction and disrupted luminal and glandular epithelium of the
uterus (Fowler et al., 2000). Mice with a null mutation in the
interleukin-11 receptor-a chain gene had defective decidualization, leading to female infertility (Robb et al., 1998). Null A-myb
female mice had underdevelopment of the breast epithelial
compartment following pregnancy and were unable to nurse their
newborn pups, while males exhibited arrest of spermatogenesis
(Toscani et al., 1997). The ®nding that mice de®cient for the
Fanca gene (Fanconi anaemia complementation group A) did not
develop anaemia, but sterility is of particular interest. Male mice
exhibited hypospermatogenesis and rarely produced offspring,
while female mice produced smaller litters and ceased to breed
between 10 and 21 weeks of age. Therefore, at the gonad level,
these mice can be compared with Fanconi patients, who often
display hypogonadism, reduced sperm count and premature
menopause (Cheng et al., 2000).
202
Diseases and male infertility
Knowledge of the genes involved in spermatogenesis and cancer
DNA mismatch repair genes appear to be responsible for most
cases of hereditary non-polyposis colorectal cancer cells (Arnheim
and Shibata, 1997). Use of knockout mice has led to the
identi®cation of which genes are involved in the maintenance of
the genome integrity of both somatic and meiotic cells (Table IV)
and which protect somatic cells only. This was the case for the
DNA mismatch repair gene Msh2 expressed in spermatogonia and
spermatocytes (Richardson et al., 2000) whose inactivation
induced susceptibility to lymphoid tumours and fully conserved
fertility (De Wind et al., 1995; Reitmair et al., 1995). Inversely,
inactivation of the DNA mismatch repair gene Msh5 led only to
sterility in both sexes (Edelmann et al., 1999). Therefore tissuespeci®c genes, genes common to actively proliferating cells, and
genes common to cells undergoing recombination can be
distinguished. This leads to the identi®cation of genes involved
in both cancer predisposition and sterility (Table IV). An
outstanding model was the Atm gene due to its involvement in
human ataxia telangiectasia (Hawley and Friend, 1996).
Mechanisms of tumorigenesis can be analysed by means of
knockout and transgenic mice. Inhibin-de®cient mice developed
gonadal stromal tumours, showing that inhibin is a negative
regulator of gonadal stromal cell proliferation and a tumoursuppressor gene with gonadal speci®city (Matzuk et al., 1992). In
cell lines, loss of function of the p53 tumour suppressor protein is
associated with increased genetic instability including aneuploidy,
gene ampli®cation, and point mutation. In null mutants for p53, a
signi®cant proportion of ®broblasts and T lymphocytes showed
chromosome loss/duplication and interstitial deletion. Increased
interstitial deletion was seen also in p53 heterozygous mice. It is
suggested that this increased genetic variation starts at the
initiation stage of tumorigenesis when functional p53 is absent
(Shao et al., 2000).
Transgenesis also allows the discovery of genes that might
have a role in the formation of testicular tumours (see review in
Matzuk et al., 1996) where GDNF, a neurotrophic factor whose
receptor is expressed in spermatogonia, is overexpressed. The
transgenic spermatogonia began to spread into the interstitium
and formed non-metastatic tumours after one year of age. Among
12 old mice, 10 had bilateral and two had unilateral tumours
(Meng et al., 2000). GDNF-family ligands may act as paracrine
factors in spermatogenesis, and this circuit may be active in germ
cell tumours (Viglietto et al., 2000).
Various diseases associated with male sterility
Male infertility can be associated with other diseases in
knockout mice (Table V). This association is not surprising
given the numerous laboratory mice with spontaneous mutations leading to male sterility and associated with a panel of
diseases.
Association of male sterility with reduced body growth is
frequently encountered when transcriptional factors are involved
(not listed). Deletion of the transcription factor gene Nhlh2
resulted in disruption of the hypothalamic±pituitary axis, leading
to obesity and male sterility due to germ cell loss (Good et al.,
1997). Inactivation of cyclin-dependent kinase 4 has led to a
Genetic engineering and spermatogenesis
Table V. Somatic tissues disorders found in null mutant mice with altered spermatogenesis
Gene
Anomalies
Reference(s)
c-fos
Bax
Atm
c-Abl
FAC
E2F-1
Nhlh2
mOvo1
Ahch (Dax1)
histone 3.3A
connexin 43
Osp/claudin-11
osteopetrosis, lymphopoenia
lymphoid hyperplasia
immunode®ciency
lymphopoenia
Fanconi anaemia
exocrine gland dysplasia
obesity
kidney, hair and urogenital defects
adrenal hypoplasia congenita
neuromuscular de®cit
heart abnormality
tight junction defects (CNS);
conduction velocity defects
insulin-de®cient diabetes
adipocyte hypertrophy
inner ear defects
polycystic kidney disease
facial dysmorphism, anaemia,
cystic choroid plexus
exencephaly, cranioschesis
lung emphysema; skin and hair defects;
defective oligodendrogenesis
bone maturation
Johnson et al., 1992
Knudson et al., 1995
Xu et al., 1996; Barlow et al., 1996
Kharbanda et al., 1998
Whitney et al., 1996
Yamasaki et al., 1996
Good et al., 1997
Dai et al., 1998
Yu et al., 1998
Couldrey et al., 1999
Juneja et al., 1999
Gow et al., 1999
cyclin-dependent kinase 4
hormone-sensitive lipase
Slc12a2a
NIMA-related kinase
Apaf-1
PDGF-A
cyp 19
a
Na+-K+-2Cl± cotransporter.
Rane et al., 1999
Osuga et al., 2000
Pace et al., 2000
Upadhya et al., 2000
Honarpour et al., 2000
Gnessi et al., 2000
Robertson et al., 1999
CNS = central nervous system.
model of insulin-de®cient diabetes and, consequently, to associated male sterility (Rane et al., 1999). Disruption of the
hormone-sensitive lipase gene led to oligospermia and male
sterility and the adipocytes were 2- to 5-fold enlarged (Osuga et
al., 2000). Mutation of the histone 3.3A gene resulted in postnatal
death or neuromuscular de®cit, reduced copulatory activity, and
poor reproduction rate (Couldrey et al., 1999). Mice lacking the
transcription factor E2F-1 were fertile, though with testicular
atrophy; in addition, they presented exocrine gland dysplasia, and
a broad and unusual spectrum of tumours (Yamasaki et al., 1996).
Loss of the Ahch (Dax1) gene led to male sterility, and mutation
of Ahch in human caused adrenal hypoplasia congenita (Yu et al.,
1998). Male and female germ cell de®ciency and heart
abnormalities were found in mice lacking connexin 43, a
component of intercellular membrane channels of gap junctions
(Juneja et al., 1999). Animals de®cient for the platelet-derived
growth factor PDGF-A develop lung emphysema, defective
oligodendrogenesis, and skin and hair defects. Most die within
a few days, but some individuals survive as long as 6 weeks.
Mutant prepubertal testis contained mature fetal Leydig cells,
while adult Leydig cells were absent in older ±/± testis, leading to
spermatocyte arrest (Gnessi et al., 2000).
In some cases, knockout mice with somatic disorders may be
compared with human diseases. Homozygous mutants for the
Nek1 (a NIMA-related kinase) gene showed polycystic kidney
disease (PKD) similar to the human autosomal-dominant PKD. In
addition, they showed pleiotropic anomalies such as male
sterility, facial dysmorphism, dwar®ng, anaemia and cystic
choroid plexus (Upadhya et al., 2000). Mice de®cient for the
mOvo1 gene showed kidney, hair and urogenital defects,
hypogonadism and reduced sperm production. The phenotype of
mOvo±/± mice can be compared with the human Bardet±Bield
syndrome (Dai et al., 1998).
An animal model with some restraints
The challenges of translating knockout phenotypes into gene
function has been already discussed for somatic cells: `often a
deletion of a gene that has accepted functions from biochemical
and cell biology experiments results in an inconsistent phenotype.
Frequently this takes the form of no or mild phenotypes and
evokes the response that a redundantly functioning gene exists.
Equally frequently there is a dramatic phenotype that challenges
the investigators' skills to translate it into gene function and draws
them into unknown realms' (Ihle, 2000).
Transgenic insertions and embryonic stem cell knockouts
imply some inherent limitations, and their use as animal
models for the understanding of spermatogenesis is still under
discussion. Considerations include variations in the phenotype
on different genetic backgrounds, confounding effects due to
the promoter used, position±effect variegation, and insertion of
tandem multi-copy arrays (reviewed in Escalier, 1999a). In
addition to these technical limitations, some other parameters
constitute restraints for animal models of male reproduction.
Gene widespread expression
When a gene is involved both in vital processes and spermatogenesis, null mutants may not bring information on its role in
203
D.Escalier
Table VI. Expression of the DAZ-like genes in various species
Gene
Species
Male
DAZ
human
DAZL1
human
spermatogonia
spermatocyte I
PGC
yes
germ cells (embryo)
SPGY
cynDAZLA
DAZL
Dazla
human
monkey
marmoset
mouse
Dazl
rat
Xdazl
Xenopus
Boule
Drosophila
zDazl
Dazl
zebra®sh
C. elegans
yes
yes
spermatogonia
B spermatogonia
spermatocyte I
PGC
spermatogonia
prospermatogonia
spermatocyte I ?
spermatogonia?
spermatocytes I
spermatids
PGC (migration)
PGC + germ cell
spermatogonia
spermatocyte I
(meiotic divisions)
yes
no
Female
PGC, oocyte
oocyte
oocyte I
oogonia, oocytes,
granulosa (embryo)
no
no
yes
Reference(s)
Menke et al., 1997
Lee, J.H. et al., 1998
Seligman and Page, 1998
Dorfman et al., 1999
Tsai et al., 2000
Brekhman et al., 2000
Shan et al., 1996
Carani et al., 1997
Ruggiu et al., 2000
Niederberger et al., 1997
Seligman and Page, 1998
Reijo et al., 1996
oocyte I ?
Ruggiu et al., 1997
Rocchietti-March et al., 2000
PGC (migration)
PGC+ germ cells
no
Houston and King, 2000
Mita and Yamashita, 2000
Cheng et al., 1998
yes
oocyte I
Eberhart et al., 1996
Maegawa et al., 1999
Karashima et al., 2000
PGC = primordial germ cell.
spermatogenesis. It is the case when the use of gene knockouts
leads to lethality of embryos, of fetus or of newborns such as for
cyclin B1 (Brandeis et al., 1998), the recombinase Rad51
(Tsuzuki et al., 1996), the phospholipase cPLA2 (Bonventre et
al., 1997) and the Brca2 genes (Sharan et al., 1997). It is also the
case for the helicase gene Blm (mutated in Bloom's syndrome),
the disruption of which has led to embryonic death (Chester et al.,
1998). Nevertheless, it is known that men with Bloom's syndrome
are sterile with abnormally small testes and no mature
spermatozoa (German, 1993).
Gene redundancy
Besides indicating candidate genes for infertilities, null mutations
demonstrate that some genes expressed in germ cells can be
altered without consequence on spermatogenesis and fertilization
capacity, although the genes were known to be implicated in
spermatogenesis. In most cases this fact is due to the diversity of
members of a protein family which can provide a functional
overlap and complementation. This is the case for cyclin B1 that
compensates cyclin B2 in testis (Brandeis et al., 1998). The same
phenomenon is observed for histones (Rabini et al., 2000; see
discussion in Couldrey et al., 1999), even for the testis-speci®c
linker histone H1t (Lin et al., 2000). H1t is expressed exclusively
in pachytene spermatocytes of the testis. H1t-de®cient mice were
fertile and reproduced as wild-type mice. The process of
spermatogenesis and the testicular morphology remained
unchanged. H1.1, H1.2 and H1.4 histone gene expression were
enhanced in these mice (Drabent et al., 2000), possibly due
204
to compensatory processes. Compensatory factors are also
suspected for the transforming growth factor-alpha (Levine
et al., 2000).
Anomalies of neuromuscular activity due to a hypomorphic
mutation of histone H3.3A may contribute to reduced fertility in
mutant mice by compromising their copulatory performance.
However, the spermatozoa appeared normal, suggesting a
functional redundancy with the H3.3B gene (Couldrey et al.,
1999). The presence of redundant mechanisms in the stimulatory
control of the gonads by the pituitary is suggested by growth
hormone receptor knockout mice. The conservation of sexual
maturation and fertility in these mice indicate that their ability of
producing prolactin and thyroid-stimulating hormone partially
compensates the absence of growth hormone signalling (reviewed
in Bartke et al., 1999).
Inversely, a gene found to have a redundant function for tissues
other than testis can be essential for spermatogenesis. This is the
case for Bcl-w, a pro-survival member of the Bcl-2 family (Print
et al., 1998) and for the zinc ®nger transcription factor Egr4
(Tourtellotte et al., 1999). The growth factors Bmp8a and Bmp8b
seem to be compensated by other Bmps for the maintenance of
female reproduction, while they are essential for male reproduction (Zhao et al., 1998).
Null mutants by genetic engineering versus spontaneous mutants
Spontaneous mutations can be point mutations, leading to various
extents of the disease or disturbances restricted to a tissue. The
example of the LMNA gene that encodes lamin A/C, a major
Genetic engineering and spermatogenesis
constituent of the nuclear matrix, is very demonstrative. In the
human, different diseases are found following the localization of a
mutation in the gene, including muscular dystrophy, cardiopathy
or lipodystrophy (Shackleton et al., 2000). A mutation in the FSH
receptor can lead to female infertility, while another inactivating
mutation of the same gene has been found in a fertile woman
(Tapanainen et al., 1998).
Knockout mice with a point mutation demonstrate that a
particular phenotype can be obtained by this method. Kit/SCFReceptor is encoded by the mouse dominant white-spotting (W)
locus. The loss-of-function W mutation affects all signalling
pathways from the receptor, leading to failure of haematopoiesis,
melanogenesis and infertility. Knockout mice with a different
point mutation in the receptor exhibited only male sterility
(Blume-Jensen et al., 2000). Drosophila mutants show how
mutations for a gene involved in spermatogenesis and with
various penetration lead to different germ cell phenotypes (see
Escalier, 1999b). Therefore, it is noteworthy that diseases
observed in knockout mice can differ from human diseases
following alterations of the same gene.
Gene homologies, but functional differences between species
Genetic engineering in mammals is the most effective means of
controlling the function of a gene. Nevertheless, great differences
in the differentiation of male germ cells exist among mammals,
including those affecting the recombination steps. For example,
the nuclear changes at the pachytene stage differ between the
human and the mouse, and even between the mouse and the rat.
This can be seen at the level of the granular component of the
nucleolus (re¯ecting the level of rRNA storage), which is
maximal in the mouse and the rat but not in the human, and
this nucleolar compartment is mixed with the XY chromosomes
in the mouse but not in the rat (reviewed in Solari, 1974).
Many genes involved in mammalian spermatogenesis have
been identi®ed ®rst by homology with those known in yeast,
particularly genes related to recombination (recombinases, DNA
mismatch repair genes) and in Drosophila. Nevertheless, a
sequence homology between species does not necessary imply a
similar function. For example, the ovo gene is required for
survival and differentiation of Drosophila female germ cells, but
is required for sperm development in the mouse (Dai et al., 1998)
(other examples are given in Escalier, 1999b). The mouse insulinlike hormone 3 (also designated Leydig insulin-like or relaxin-like
factor) is expressed in pre- and postnatal Leydig cells of the testis
and in postnatal theca cells of the ovary, and is essential for
establishment of the sexual dimorphic position of the gonads
(Adham et al., 2000). Indeed, inactivation of the Insl3 gene leads
to cryptorchidism in the mouse (Nef and Parada, 1999;
Zimmermann et al., 1999). In the human, a common polymorphism is found in the Insl3 C-peptide region which is not related
with idiopathic cryptorchidism (Koskimies et al., 2000; Krausz et
al., 2000).
The reasons for these functional divergences might be: (i)
restriction of homology to only one domain (as found for
members of a super-family) while the other domains have a
distinct function; (ii) conditioning of the gene function by
interaction with other factors; and (iii) differences between
species in expression timing and/or gene product cell localization
that can be associated to distinct functions. However, other
mechanisms can be involved.
The gene product can have a function on different factors, regulating
their translation or expression, or they can regulate the stability of
mRNAs
This can be the case for the RNA-binding DAZL (Cheng et al.,
1998; Houston and King, 2000), RBM (Venables et al., 2000) and
DAX-1 (Lalli et al., 2000). RBM interacts with members of the SR
family of splicing factors, and may be a splicing regulator which
operates as a germ cell-speci®c cofactor for more ubiquitously
expressed pre-mRNA splicing activators (Elliott et al., 2000). The
mouse Dazl1 is associated with actively translating ribosomes
(Tsui et al., 2000b), and can interact through DAZ repeats with
DAZAP1, a RNA-binding protein (Tsui et al., 2000a). Table VI
shows that Dazla-like genes are expressed at various stages of
germ cell differentiation, depending on the species. Moreover,
their expression concerns either only the male or the female, or
both. It is noteworthy that the functionality of the Daz gene on the
human Y chromosome remains to be demonstrated (Shan et al.,
1996), while two Y-chromosomal genes of the AZFa region,
USP9Y (Sun et al., 1999) and DBY (Foresta et al., 2000), seem to
be implicated in human azoospermia. In contrast to other AZFa
genes, DBY produces a short transcript which is only expressed in
the testis, suggesting that DBY might be the major AZFa
candidate to testiculopathies (Foresta et al., 2000).
The homology can be the result of co-option of a gene during
evolution for a new function
This could be the case of the mouse germ cell-less gene (mgcl-1)
which is homologous to the Drosophila germ cell-less (Gcl).
Functional homology was demonstrated, since ectopic expression
of mgcl-1 in Drosophila partially rescues the gcl-null phenotype.
Nevertheless, unlike the case in Drosophila, mgcl-1 is not
expressed at the time the primordial germ cells appear, but in
adult male germ cells (Leatherman et al., 2000). Other mouse
genes homologous to Drosophila germ plasm components
involved in the formation of pole cells are highly expressed in
mouse spermatocytes. There are the vasa-like family members
PL10, P68 and Mvh. Thus, it appears that some mechanisms used
in pole cell formation in Drosophila have been co-opted as a
group for a later developmental process in mouse spermatogenesis (Leatherman et al., 2000).
Genetic status of spermatozoa
Intracytoplasmic sperm injection (ICSI) could promote the transgenerational transmission of genetic defects, causing gametogenic
failure. Among infertile couples treated with ICSI, 6.4% were
shown to have a fertility problem with a de®nite genetic basis, and
males displayed a distinct pattern of familial aggregation. The
infertile couples had fewer siblings than did fertile controls,
suggesting suboptimal fertility in the infertile couples' parents.
Therefore, male factor infertility should be considered a
potentially heritable condition, and the recurrence risk for
infertility in the offspring of couples treated with ICSI might be
substantial (Meschede et al., 2000a).
A noteworthy ®nding in knockout mice was that only
homozygous mice were sterile, supporting the notion that
mRNAs and proteins pass through bridges between male germ
205
D.Escalier
cells (see Escalier, 1999a). Therefore, sperm cells from heterozygous mice presented a normal phenotype, although half of these
spermatozoa shared a genetic defect (Escalier, 1999b). This might
be the case for human males heterozygous for the helicase BLM
involved in recessive autosomal Bloom's syndrome whose
mutation leads to immunode®ciency, cancer predisposition and
sterility (German, 1993). Also, spontaneous mutants present male
sterility in homozygotes whereas heterozygotes are fertile, as in
the case of rat hypodactyly (Krenova et al., 1999).
By contrast, the expression of a transgene can cause dominant
male sterility at the hemizygous status because of syncitial
spermatids. This is found in mice carrying a protamine-1
transgene lacking its normal 3¢ untranslated region, and leads to
premature translation of protamine-1 and round spermatid arrest
(Lee et al., 1995). Finally, a hemizygous status can lead to Sertoli
cell-only syndrome in older GDNF +/± mice, re¯ecting haploinsuf®ciency for this gene product. GDNF is expressed in Sertoli
cells, and is essential for undifferentiated spermatogonia (stem
cells) (Meng et al., 2000).
Conclusions
Although there are some limitations inherent to the experimental
procedures used, genetic engineering has promoted several major
advances in our understanding of mammalian spermatogenesis.
The techniques are, however, in their early days, and the
emergence of these methodsÐin conjunction with ICSIÐhas
generated new interest in the study of spermatogenesis. Moreover,
bene®ts have also been derived from advances in cell biology.
The ®ndings of mouse genetic engineering studies should be
the basis to seek the genes responsible for human male infertility.
Male germ cell phenotypes for both infertile men and mouse
models should be identi®ed using a rigorous de®nition, the main
factor for success being the wide cooperation of laboratories.
There is a clear need to create a constitutional DNA `bank' after
collecting blood samples from subjects belonging to families with
hereditary forms of impaired spermatogenesis, and to create
similar banks of pathological germ cells.
In humans, although infertile patients show a slight higher
prevalence of potentially heritable, non-reproductive diseases
(Meschede et al., 2000b), this situation is not studied to any great
extent. However, the frequent association of male sterility with
diseases and/or tumour predisposition in infertile mice must be
mentioned, as this can occur as a result of both activation or
overexpression of genes. These pleiotropic phenotypes strongly
emphasize the importance of understanding the epidemiology of
male infertility with regard to the development of medically
assisted human reproduction and gene therapy.
Note added at proof
A recent study reveals that Insl 3 is involved in some cases of
cryptorchidism in human (Tomboe et al., 2000).
References
Abel, M.H., Wootton, A.N., Wilkins, N. et al. (2000) The effect of null
mutation in the follicle-stimulating hormone receptor gene on mouse
reproduction. Endocrinology, 141, 1795±1803.
206
Adham, I.M., Nayernia, K., and Engel, W. (1997) Spermatozoa lacking
acrosin protein show delayed fertilization. Mol. Reprod. Dev., 46, 370±
376.
Adham, I.M., Emmen, J.M.A. and Engel, W. (2000) The role of the testicular
factor INSL3 in establishing the gonad position. Mol. Cell. Endocrinol.,
160, 11±16.
Anderson, R., Fassler, R., Georges-Labouesse, E. et al. (1999) Mouse
primordial germ cells lacking beta1 integrins enter the germline but fail to
migrate normally to the gonads. Development, 126, 1655±1664.
Anderson, R., Copeland, T.K., Sholer, H. et al. (2000) The onset of germ cell
migration in the mouse embryo. Mech. Dev., 91, 61±68.
Arnheim, N. and Shibata, D. (1997) DNA mismatch repair in mammals: role
in disease and meiosis. Curr. Opin. Genet. Dev., 7, 364±370.
Baarends, W.N., Hoogerbrugge, J.W., Roest, H.P. et al. (1999) Histone
ubiquitination and chromatin remodeling in mouse spermatogenesis. Dev.
Biol., 207, 322±333.
Baba, T., Azuma, S., Kashiwabara, S.I. et al. (1994) Sperm from mice carrying
a targeted mutation of the acrosin gene can penetrate the oocyte zona
pellucida and effect fertilization. J. Biol. Chem., 269, 31845±31849.
Baker, J., Hardy, M.P., Zhou, J. et al. (1996) Effects of an Igf1 gene mutation
on mouse reproduction. Mol. Endocrinol., 10, 903±918.
Baker, S.M., Bronner, C.E., Zhang, L. et al. (1995) Male defective in the DNA
mismatch repair gene PMS2 exhibit abnormal chromosome synapsis in
meiosis. Cell, 82, 309±319.
Baker, S.M., Plug, A.W., Prolla, T.A. et al. (1996) Involvement of mouse
Mlh1 in DNA mismatch repair and meiotic crossing over. Nature Genet.,
13, 336±342.
Barlow, C., Hirotsune, S., Paylor, R. et al. (1996) Atm-de®cient mice: a
paradigm of ataxia telangiectasia. Cell, 86, 159±171.
Barlow, C., Liyanage, M., Moens, P.B. et al. (1998) Atm de®ciency results in
severe meiotic disruption as early as leptonema of prophase I.
Development, 125, 4007±4017.
Bartell, J.G., Fantz, D.A., Davis, T. et al. (2000) Elimination of male germ
cells in transgenic mice by the diphtheria toxin A chain gene directed by
the histone H1t promoter. Biol. Reprod., 63, 409±416.
Bartke, A., Chandrashekar, V., Turyn, D. et al. (1999) Effects of growth
hormone overexpression and growth hormone resistance on
neuroendocrine and reproductive functions in transgenic and knock-out
mice. Proc. Soc. Exp. Biol. Med., 222, 113±123.
Baum, M.J., Brown, J.J., Kica, E. et al. (1994) Effect of a null mutation of the
c-fos proto-oncogene on sexual behavior of male mice. Biol. Reprod., 50,
1040±1048.
Beck, A.R.P., Miller, I.J., Anderson, P. et al. (1998) RNA-binding protein
TIAR is essential for primordial germ cell development. Proc. Natl Acad.
Sci. USA, 95, 2331±2336.
Bera, T.K. and Pastan, I. (2000) Mesothelin is not required for normal mouse
development or reproduction. Mol. Cell. Biol., 20, 2902±2906.
Beumer, T.L., Roepers-Gajadien, H. L., Gademan, I.S. et al. (2000) Apoptosis
regulation in the testis: involvement of bcl-2 family members. Mol.
Reprod. Dev., 56, 353±359.
Birk, O.S., Casiano, D.E., Wassif, C.A. et al. (2000) The LIM homeobox gene
Lhx9 is essential for mouse gonad formation. Nature, 403, 909±913.
Bitgood, M.J., Shen, L. and McMahon, A.P. (1996) Sertoli cell signaling by
Desert hedgehog regulates the male germline. Curr. Biol., 6, 298±304.
Blendy, J.A., Kaestner, K.H., Weinbauer, G.F. et al. (1996) Severe impairment
of spermatogenesis in mice lacking the CREM gene. Nature, 380, 162±
165.
Blume-Jensen, P., Jiang, G., Hyman, R. et al. (2000) Kit/stem cell factor
receptor-induced activation of phosphatidylinositol 3'-kinase is essential
for male fertility. Nature Genet., 24, 157±162.
Bonventre, J.V., Huang, Z., Taheri, M.R. et al. (1997) Reduced fertility and
postischaemic brain injury in mice de®cient in cytosolic phospholipase
A2. Nature, 390, 622±625.
Bouchard, M.J., Dong, Y., McDermott, B.M., Jr et al. (2000) Defects in
nuclear and cytoskeletal morphology and mitochondrial localization in
spermatozoa of mice lacking nectin-2, a component of cell-cell adherens
junctions. Mol. Cell. Biol., 20, 2865±2873.
Brandeis, M., Rosewell, I., Carrington, M. et al. (1998) Cyclin B2-null mice
develop normally and are fertile whereas cyclin B1-null mice die in utero.
Proc. Natl Acad. Sci. USA, 95, 4344±4349.
Braun, R.E. (2000) Temporal control of protein synthesis during
spermatogenesis. Int. J. Androl., 23 (Suppl. 2), 92±94.
Brekhman, V., Itskovitz-Eldor, J., Yodko, E. et al. (2000) The DAZL1 gene is
expressed in human male and female embryonic gonads before meiosis.
Mol. Hum. Reprod., 6, 465±468.
Genetic engineering and spermatogenesis
Calenda, A., Allenet, B., Escalier, D. et al. (1994) The meiosis-speci®c Xmr
gene product is homologous to the lymphocyte Xlr protein and is a
component of the XY body. EMBO J., 13, 100±109.
Carani, C., Gromoll, J., Brinkworth, M.H. et al. (1997) CynDAZLA: a
cynomolgus monkey homologue of the human autosomal DAZ gene. Mol.
Hum. Reprod., 3, 479±483.
Cheng, M.H., Maines, J.Z. and Wasserman, S.A. (1998) Biphasic subcellular
localization of the DAZL-related protein boule in Drosophila
spermatogenesis. Dev. Biol., 204, 567±576.
Cheng, N.C., van de Vrugt, H.J., van der Valk, M.A. et al. (2000) Mice with a
targeted disruption of the Fanconi anemia homolog Fanca. Hum. Mol.
Genet., 9, 1805±1811.
Chester, N., Kuo, F., Kozak, C. et al. (1998) Stage-speci®c apoptosis,
developmental delay, and embryonic lethality in mice homozygous for a
targeted disruption in the murine Bloom's syndrome gene. Genes Dev.,
12, 3382±3393.
Cho, C., Bunch, D.O., Faure, J.-E. et al. (1998) Fertilization defects in sperm
from mice lacking fertilin b. Science, 281, 1857±1859.
Cho, C., Ge, H., Branciforte, D. et al. (2000) Analysis of mouse fertilin in
wild-type and fertilin b±/± sperm: evidence for C-terminal modi®cation, a/
b dimerization, and lack of essential role of fertilin a in sperm-egg fusion.
Dev. Biol., 222, 289±295.
Couldrey, C., Carlton, M.B.L., Nolan, P.M. et al. (1999) A retroviral gene trap
insertion into the histone 3.3A gene causes partial neonatal lethality,
stunted growth, neuromuscular de®cits and male sub-fertility in transgenic
mice. Hum. Mol. Genet., 8, 2489±2495.
Couse, J.F. and Korach, K.S. (1999) Estrogen receptor null mice: what have
we learned and where do they lead us? Endocrinol. Rev., 20, 358±417.
Couse, J.F., Hewitt S.C., Bunch, D.O. et al. (1999) Postnatal sex reversal of
the ovaries in mice lacking estrogen receptors a and b. Science, 286,
2328±2331.
Cressman, V.L., Backlund, D.C., Avrutskaya, A.V. et al. (1999) Growth
retardation, DNA repair defects, and lack of spermatogenesis in BRCA1de®cient mice. Mol. Cell. Biol., 19, 7061±7075.
Dai, X., Schonbaum C., Degenstein, L. et al. (1998) The ovo gene required for
cuticle formation and oogenesis in ¯ies is involved in hair formation and
spermatogenesis in mice. Genes Dev., 12, 3452±3463.
De Cesare, D., Fimia, G.M. and Sassone-Corsi, P. (1999) Signaling routes to
CREM and CREB: plasticity in transcriptional activation. Trends
Biochem. Sci., 24, 281±285.
De Wind, N., Dekker, M., Berns, A. et al. (1995) Inactivation of the mouse
Msh2 gene results in mismatch repair de®ciency, methylation tolerance,
hyperrecombination and predisposition to cancer. Cell, 82, 321±330.
Dierich, A., Sairam, M.R., Monaco, L. et al. (1998) Impaired folliclestimulating hormone (FSH) signaling in vivo. Targeted disruption of the
FSH receptor leads to aberrant gametogenesis and hormonal imbalance.
Proc. Natl Acad. Sci. USA, 95, 13612±13617.
Dix, D.J., Allen, J.W., Collins, B.W. et al. (1997) HSP70-2 is required for
desynapsis of synaptonemal complexes during meiotic prophase in
juvenile and adult mouse spermatocytes. Development, 124, 4595±4603.
Donehower, L.A., Harvey, M., Slage, B.L. et al. (1992) Mice de®cient for p53
are developmentally normal but susceptible to spontaneous tumors.
Nature, 356, 215±221.
Dorfman, D.M., Genest, D.R. and Reijo Pera, R.A. (1999) Human DAZL1
encodes a candidate fertility factor in women that localizes to the prenatal
and postnatal germ cells. Hum. Reprod., 14, 2531±2536.
Drabent, B., Saftig, P., Bode, C. et al. (2000) Spermatogenesis proceeds
normally in mice without linker histone H1t. Histochem. Cell Biol., 113,
433±442.
Eberhart, C.G., Maines, J.Z. and Wasserman, S.A. (1996) Meiotic cell cycle
requirement for a ¯y homologue of human Deleted in Azoospermia.
Nature, 381, 783±785.
Edelmann, W., Cohen, P.E., Kane, M. et al. (1996) Meiotic pachytene arrest in
Mlh1-de®cient mice. Cell, 85, 1125±1134.
Edelmann, W., Cohen, P.E., Kneitz, B. et al. (1999) Mammalian Muts
homologue 5 is required for chromosome pairing in meiosis. Nature
Genet., 21, 123±127.
Elliott, D.J., Bourgeois, C.F., Klink, A. et al. (2000) A mammalian germ cellspeci®c RNA-binding protein interacts with ubiquitously expressed
proteins involved in splice site selection. Proc. Natl Acad. Sci. USA, 97,
5717±5722.
Escalier, D. (1999a) Mammalian spermatogenesis investigated by genetic
engineering. Histol. Histopathol., 14, 945±958.
Escalier, D. (1999b) What are the germ cell phenotypes from infertile men
telling us about spermatogenesis? Histol. Histopathol., 14, 959±971.
Escalier, D. and Garchon, H.-J. (2000) XMR is associated with the asynapsed
segments of sex chromosomes in the XY body of mouse primary
spermatocytes. Chromosoma, 109, 259±265.
Escalier, D., Allenet, B., Badrichani, A. et al. (1999) High level expression of
the Xlr nuclear protein in immature thymocytes and colocalization with
the matrix-associated region-binding SATB1 protein. J. Immunol., 162,
292±298.
Fantin, V.R., Wang, Q., Lienhard, G.E. et al. (2000) Mice lacking insulin
receptor substrate 4 exhibit mild defects in growth, reproduction and
glucose homeostasis. Am. J. Physiol., 278, E127±E133.
Fisher, C.R., Graves K.H., Parlow A.F. et al. (1998) Characterization of mice
de®cient in aromatase (ArKO) because of targeted disruption of the cyp19
gene. Proc. Natl Acad. Sci. USA, 95, 6965±6970.
Foresta, C., Ferlin, A. and Moro, E. (2000) Deletion and expression analysis of
AZFa genes on the Y chromosome revealed a major role for DBY in male
fertility. Hum. Mol. Genet., 9, 1161±1169.
Fowler, K.J., Hudson, D.F., Salamonsen, L.A. et al. (2000) Uterine
dysfunction and genetic modi®ers in centromere protein-de®cient mice.
Genome Res., 10, 30±41.
Furuchi, T., Masuko, K., Nishimune, Y. et al. (1996) Inhibition of testicular
germ cell apoptosis and differentiation in mice misexpressing Bcl-2 in
spermatogonia. Development, 122, 1703±1709.
German, J. (1993) Bloom syndrome: a mendelian prototype of somatic
mutational disease. Medicine (Baltimore), 71, 393±406.
Gnessi,L., Basciani, S., Mariani, S. et al. (2000) Leydig cell loss and
spermatogenic arrest in PDGF-A-de®cient mice. J. Cell Biol., 149, 1019±
1025.
Gof®n, V., Bouchard, B., Ormandy, C.J. et al. (1998) Prolactin: a hormone at
the crossroads of neuroimmunoendocrinology. Ann. N. Y. Acad. Sci., 840,
498±509.
Good, D.J., Porter, F.D., Mahon, K.A. et al. (1997) Hypogonadism and obesity
in mice with targeted deletion of the Nhlh2 gene. Nature Genet., 15, 397±
401.
Gow, A., Southwood, C.M., Li, J.S. et al. (1999) CNS myelin and Sertoli cell
tight junction strands are absent in Osp/claudin-11 null mice. Cell, 99,
649±659.
Grootegoed, J.A., Baarends, W.M., Roest, H.P. et al. (1998) Knockout mouse
model and gametogenic failure. Mol. Cell. Endocrinol., 145, 161±166.
Guo, Q., Kumar, T.R., Hadsell, L.A. et al. (1998) Overexpression of mouse
follistatin causes reproductive defects in transgenic mice. Mol.
Endocrinol., 12, 96±106.
Hagaman, J.R., Moyer, J.S., Bachman, E.S. et al. (1998) Angiotensinconverting enzyme and male fertility. Proc. Natl Acad. Sci. USA, 95,
2552±2557.
Halmekyto, M., Hyttinen, J.M., Sinervirta, R. et al. (1991) Transgenic mice
aberrantly expressing human ornithine decarboxylase gene. J. Biol.
Chem., 266, 19746±19751.
Hanley, N.A., Hagan, D.M., Clement-Jones, M. et al. (2000) SRY, SOX9 and
DAX1 expression patterns during human sex determination and gonadal
development. Mech. Dev., 91, 403±407.
Hartl, F.U. (1996) Molecular chaperones in cellular protein folding. Nature,
381, 571±580.
Hawley, R.S. and Friend, S.H. (1996) Strange bedfellows in even stranger
places: the role of ATM in meiotic cells, lymphocytes, tumors and its
functional links to p53. Genes Dev., 10, 2383±2388.
Hess, R.A., Bunick, D., Lee, K.-H. et al. (1997) A role for oestrogens in the
male reproductive system. Nature, 390, 509±512.
Higgy, N.A., Zackson, S.L. and Van der Hoorn, F.A. (1995) Cell interactions
in testis development: overexpression of c-mos in spermatocytes leads to
increased germ cell proliferation. Dev. Genet., 16, 190±200.
Hirao, Y. and Eppig, J.J. (1997) Parthenogenetic development of Mosde®cient mouse oocytes. Mol. Reprod. Dev., 48, 391±396.
Holmberg, C., Helin, K., Sehested, M. et al. (1998) E2F-1-induced p-53independent apoptosis in transgenic mice. Oncogene, 17, 143±155.
Honarpour, N., Richardson, D.C., Hammer, R.E. et al. (2000) Adult Apaf-1de®cient mice exhibit male infertility. Dev. Biol., 218, 248±258.
Houston, D.W. and King, M.L. (2000) A critical role for Xdazl, a germ plasmlocalized RNA, in the differentiation of primordial germ cells in Xenopus.
Development, 127, 447±456.
Hudson, D.F., Fowler, K.J., Earle, E. et al. (1998) Centromere protein B null
mice are mitotically and meiotically normal but have lower body and
testis weights. J. Cell Biol., 141, 309±319.
Igakura, T., Kadomatsu, K., Kaname, T. et al. (1998) A null mutation in
Basigin, an immunoglobulin superfamily member, indicates its important
207
D.Escalier
roles in peri-implantation development and spermatogenesis. Dev. Biol.,
194, 152±165.
Ihle, J.N. (2000) The challenges of translating knockout phenotypes into gene
function. Cell, 102, 131±134.
Ikawa, M., Wada, I., Kominami, K. et al. (1997) The putative chaperone
calmegin is required for sperm fertility. Nature, 387, 607±611.
Iwakura, Y., Asano, M., Nishimune, Y. et al. (1988) Male sterility of
transgenic mice carrying exogenous mouse interferon-beta gene under the
control of the metallothionein enhancer-promoter. EMBO J., 7, 3757±
3762.
Johnson, R.S., Spiegelman, B.M. and Papaioannou, V. (1992) Pleiotropic
effects of a null mutation in the c-fos proto-oncogene. Cell, 71, 577±586.
Jordan, S.A., Speed, R.M., Bernex, F. et al. (1999) De®ciency of Trp53
rescues the male fertility defects of Kit(W-v) mice but has no effect on the
survival of melanocytes and mast cells. Dev. Biol., 215, 78±90.
Joseph, D.R., Obrien, D.A., Sullivan, P.M. et al. (1997) Overexpression of
androgen-binding protein/sex hormone-binding globulin in male
transgenic mice: tissue distribution and phenotypic disorders. Biol.
Reprod., 56, 21±32.
Juneja, S.C., Barr, K.J., Enders, G.C. et al. (1999) Defects in the germ line and
gonads of mice lacking connexin 43. Biol. Reprod., 60, 1263±1270.
Jurisicova, A., Lopes, S., Meriano, J. et al. (1999) DNA damage in round
spermatids of mice with a targeted disruption of the Pp1c gamma gene
and in testicular biopsies of patients with non-obstructive azoospermia.
Mol. Hum. Reprod., 5, 323±330.
Kaji, K., Oda, S., Shikano, T. et al. (2000) The gamete fusion process is
defective in eggs of Cd9-de®cient mice. Nature Genet., 24, 279±282.
Karashima, T., Sugimoto, A. and Yamamoto, M. (2000) Caenorhabditis
elegans homologue of the human azoospermia factor DAZ is required for
oogenesis but not for spermatogenesis. Development, 127, 1069±1079.
Kastner, P., Mark, M., Leid, M. et al. (1996) Abnormal spermatogenesis in
RXR beta mutant mice. Genes Dev., 10, 80±92.
Kharbanda, S., Pandey, P., Morris, P.L. et al. (1998) Functional role for the cAbl tyrosine kinase in meiosis I. Oncogene, 16, 1773±1777.
Kissel, H., Timokhina, I., Hardy, M.P. et al. (2000) Point mutation in kit
receptor tyrosine kinase reveals essential roles for Kit signaling in
spermatogenesis and oogenesis without affecting other Kit responses.
EMBO J., 19, 1312±1326.
Kneitz, B., Cohen, P.E., Avdievich, E. et al. (2000) Muts homolog 4
localization to meiotic chromosomes is required for chromosome pairing
during meiosis in male and female mice. Genes Dev., 14, 1085±1097.
Knudson, C.M. and Korsmeyer, S.J. (1997) Bcl-2 and Bax function
independently to regulate cell death. Nature Genet., 16, 358±363.
Knudson, C.M., Tung, K.S.K., Tourtelotte, W.G. et al. (1995) Bax-de®cient
mice with lymphoid hyperplasia and male germ cell death. Science, 270,
96±99.
Kodaira, K., Takahashi, R.-I., Hirabayashi, M. et al. (1996) Overexpression of
c-myc induces apoptosis at the prophase of meiosis of rat primary
spermatocytes. Mol. Reprod. Dev., 45, 403±410.
Komada, M., McLean, D.J., Griswold, M.D. et al. (2000) E-MAP-115,
encoding a microtubule-associated protein, is a retinoic acid-inducible
gene required for spermatogenesis. Genes Dev., 14, 1332±1342.
Korpelainen, E.I., Karkkainen, M.J., Tenhunen, A. et al. (1998)
Overexpression of VEGF in testis and epididymis causes infertility in
transgenic mice: evidence for nonendothelial targets for VEGF. J. Cell
Biol., 143, 1705±1712.
Koskimies, P., Virtanen, H., Lindstrom, M. et al. (2000) A common
polymorphism in the human relaxin-like factor (RLF) gene: no
relationship with cryptorchidism. Pediatr. Res., 47, 538±541.
Krausz, C., Quintana-Murci, L., Fellous, M. et al. (2000) Absence of
mutations involving the INSL3 gene in human idiopathic cryptorchidism.
Mol. Hum. Reprod., 6, 298±302.
Krenova, D., Jirsova, Z., Bila, V. et al. (1999) Genetics of rat hypodactyly. J.
Exp. Anim. Sci. 41, 47±50.
Krishnamurthy, H., Danilovich, N., Morales, C.R. et al. (2000) Qualitative and
quantitative decline in spermatogenesis of the follicle-stimulating
hormone receptor knockout (FORKO) mouse. Biol. Reprod., 62, 1146±
1159.
Kuroda, M., Sok, J., Webb, L. et al. (2000) Male sterility and enhanced
radiation sensitivity in TSL±/± mice. EMBO J., 19, 453±462.
Lalli, E., Ohe, K., Hindelang, C. et al. (2000) Orphan receptor DAX-1 is a
shuttling RNA binding protein associated with polyribosomes via mRNA.
Mol. Cell. Biol., 20, 4910±4921.
Lamb, D.J. (1999) Debate: is ICSI a genetic time bomb ? Yes. J. Androl., 20,
23±32.
208
Lawson, K., Dunn, N., Roelen, B. et al. (1999) BMP4 is required for the
generation of primordial germ cells in the mouse embryo. Genes Dev., 13,
424±436.
Layman, L.C. and McDonough, P.G. (2000) Mutations of follicle stimulating
hormone-beta and its receptor in human and mouse: genotype/phenotype.
Mol. Cell. Endocrinol., 161, 9±17.
Leatherman, J.L., Kaestner, K.H. and Jongens, T.A. (2000) Identi®cation of a
mouse germ cell-less homologue with conserved activity in Drosophila.
Mech. Dev., 92, 145±153.
Lee, H.-W., Blasco, M.A., Gottlieb, G.J. et al. (1998) Essential role of mouse
telomerase in highly proliferative organs. Nature, 392, 569±574.
Lee, J.H., Lee, D.R., Yoon, S.J. et al. (1998) Expression of DAZ (deleted in
azoospermia), DAZL1 (DAZ-like) and protamine-2 in testis and its
application for diagnosis of spermatogenesis in non-obstructive
azoospermia. Mol. Hum. Reprod., 4, 827±834.
Lee, K., Haugen, H.S., Clegg, C.H. et al. (1995) Premature translation of
protamine-1 mRNA causes precocious nuclear condensation and arrests
spermatid differentiation in mice. Proc. Natl Acad. Sci. USA, 92, 12451±
12455.
Levine, E., Cupp, A.S., Miyashiro, L. et al. (2000) Role of transforming
growth factor-alpha and the epidermal growth factor receptor in
embryonic rat testis development. Biol. Reprod., 62, 477±490.
Lin, Q.C., Sirotkin, A. and Skoultchi, A.I., (2000) Normal spermatogenesis in
mice lacking the testis-speci®c linker histone H1t. Mol. Cell. Biol., 20,
2122±2128.
Liu, D., Matzuk, M.M., Sung, W.K. et al. (1998) Cyclin A1 is required for
meiosis in the male mouse. Nature Genet., 20, 377±380.
Lu, Q.X. and Shur, B.D. (1997) Sperm from beta-1,4-galactosyltransferasenull mice are refractory to ZP3-induced acrosome reactions and penetrate
the zona pellucida poorly. Development, 124, 4121±4131.
Lu, Q., Gore, M., Zhang, Q. et al. (1999) Tyro-3 family receptors are essential
regulators of mammalian spermatogenesis. Nature, 398, 723±728.
Luoh, S.-W., Bain, P.A., Polakiewicz, R.D. et al. (1997) Zfx mutation results
in small animal size and reduced germ cell number in male and female
mice. Development, 124, 2275±2284.
Maegawa, S., Yasuda, K. and Inoue, K. (1999) Maternal mRNA localization
of zebra®sh DAZ-like gene. Mech. Dev., 81, 223±226.
Mahato, D., Goulding, E.H., Korach, K.S. et al. (2000) Spermatogenic cells do
not require estrogen receptor-alpha for development or function.
Endocrinology, 141, 1273±1276.
Maleszewski, M., Kuretake, S., Evenson, D. et al. (1998) Behaviour of
transgenic mouse spermatozoa with galline protamine. Biol. Reprod., 58,
8±14.
Malik, N., Haugen, S., Modrell, B. et al. (1995) Developmental abnormalities
in mice transgenic for bovine oncostatin. Mol. Cell. Biol., 15, 2349±2358.
Mangues, R., Seidman, I., Pellicier, A. et al. (1990) Tumorigenesis and male
sterility in transgenic mice expressing a MMTV/N-ras oncogene.
Oncogene, 5, 1491±1497.
Marahrens, Y., Panning, B., Dausman, J. et al. (1997) Xist-de®cient mice are
defective in dosage compensation but not spermatogenesis. Genes Dev.,
11, 156±166.
Matsui, Y., Nagano, R. and Obinata, M. (2000) Apoptosis of fetal testicular
cells is regulated by both p53-dependent and independent mechanisms.
Mol. Reprod. Dev., 54, 399±405.
Matzuk, M.M., Finegold, M.J., Su, J.G. et al. (1992) Alpha-inhibin is a tumorsuppressor gene with gonadal speci®city in mice. Nature, 360, 313±319.
Matzuk, M.M., Kumar, T.R. and Bradley, A. (1995) Different phenotypes for
mice de®cient in either activins or activin receptor type II. Nature, 374,
356±360.
Matzuk, M.M., Kumar, T.R., Shou, W. et al. (1996) Transgenic models to
study the roles of inhibins and activins in reproduction, oncogenesis and
development. Recent Prog. Horm. Res., 41, 123±154.
Mbikay, M., Tadros, H., Ishida, N. et al. (1997) Impaired fertility in mice
de®cient for the testicular germ-cell protease PC4. Proc. Natl Acad. Sci.
USA, 94, 6842±6846.
Meng, X., Lindahl, M., HyvoÈnen, M.E. et al. (2000) Regulation of cell fate
decision of undifferentiated spermatogonia by GDNF. Science, 287,
1489±1493.
Menke, D.B., Mutter, G.L. and Page, D.C. (1997) Expression of DAZ, an
azoospermia factor candidate, in human spermatogonia. Am. J. Hum.
Genet., 60, 237±241.
Meschede, D., Lemcke, B., Behre, H.M. et al. (2000a) Clustering of male
infertility in the families of couples treated with intracytoplasmic sperm
injection. Hum. Reprod., 15, 1604±1608.
Genetic engineering and spermatogenesis
Meschede, D., Lemcke, B., Behre, H.M. et al. (2000b) Non-reproductive
heritable disorders in infertile couples and their ®rst degree relatives.
Hum. Reprod., 15, 1609±1612.
Mita, K. and Yamashita, M. (2000) Expression of Xenopus Daz-like protein
during gametogenesis and embryogenesis. Mech. Dev., 94, 251±255.
Nagano, M., Shinohara, T., Avarbock, M.R. et al. (2000) Retrovirus-mediated
gene delivery into male germ line stem cells. FEBS Lett., 475, 7±10.
Nakai, A., Suzuki, M. and Tanabe, M. (2000) Arrest of spermatogenesis in
mice expressing an active heat shock transcription factor 1. EMBO J., 19,
1545±1554.
Nantel, F., Monaco, L., Foulkes, N.S. et al. (1996) Spermiogenesis de®ciency
and germ-cell apoptosis in CREM-mutant mice. Nature, 380, 159±162.
Nayernia, K., von Mering, M.H.P., Kraszucka, K. et al. (1999) A novel
testicular haploid expressed gene (THEG) involved in mouse spermatidSertoli cell interaction. Biol. Reprod. 60, 1488±1495.
Nef, S. and Parada, L.F. (1999) Cryptorchidism in mice mutant for Insl3.
Nature Genet., 22, 295±299.
Niederberger, C., Agulnik, A.I., Cho, Y. et al. (1997) In situ hybridization
shows that Dazla expression in mouse testis is restricted to premeiotic
stages IV-VI of spermatogenesis. Mamm. Genome, 8, 277±278.
Nishimori, K. and Matzuk M.M. (1996) Transgenic mice in the analysis of
reproductive development and function. Rev. Reprod., 1, 203±212.
O'Brien, D.A., Welch, J.E., Goulding, E.H. et al. (1996) Boar proacrosin
expressed in spermatids of transgenic mice does not reach the acrosome
and disrupts spermatogenesis. Mol. Reprod. Dev., 43, 236±247.
Ohta, M., Mitomi, T., Kimura, M. et al. (1990) Anomalies in transgenic mice
carrying the human interleukin-2 gene. Tokai J. Exp. Clin. Med., 15, 307±
315.
Okabe, M., Ikawa, M. and Ashkenas, J. (1998) Male infertility and the
genetics of spermatogenesis. Am. J. Hum. Genet., 62, 1274±1281.
Osuga, J., Ishibashi, S., and Oka, T. et al. (2000) Targeted disruption of
hormone-sensitive lipase results in male sterility and adipocyte
hypertrophy, but not in obesity. Proc. Natl Acad. Sci. USA, 97, 787±792.
Pace, A.J., Lee, E., Athirakul, K. et al. (2000) Failure of spermatogenesis in
mouse lines de®cient in the Na+-K+-2Cl± cotransporter. J. Clin. Invest.,
105, 441±450.
Pearse, R.V., Drolet, D.W., Kalla, K.A. et al. (1997) Reduced fertility in mice
de®cient for the POU protein sperm-1. Proc. Natl Acad. Sci. USA, 94,
7555±7560.
Pierson, T.M., Wang, Y.L., DeMayo, F.J. et al. (2000) Regulable expression
of inhibin A in wild-type and inhibin alpha null mice. Mol. Endocrinol.,
14, 1075±1085.
Pittman, D.L., Cobb, J., Schimenti, K.J. et al. (1998) Meiotic prophase arrest
with failure of chromosome synapsis in mice de®cient for Dmc1, a
germline-speci®c RecA homolog. Mol. Cell, 1, 697±705.
Print, C.G. and Loveland, K.L. (2000) Germ cell suicide: new insights into
apoptosis during spermatogenesis. BioEssays, 22, 423±430.
Print, C.G., Loveland, K.L., Gibson, L. et al. (1998) Apoptosis regulator bcl-w
is essential for spermatogenesis but not appears otherwise redundant.
Proc. Natl Acad. Sci. USA, 95, 12424±12431.
Prolla, T.A., Baker, S.M., Harris, A.C. et al. (1998) Tumor susceptibility and
spontaneous mutation in mice de®cient in Mlh1, Pms1 and Pms2 DNA
mismatch repair. Nature Genet., 18, 276±279.
Rabini, S., Franke, K., Saftig, P. et al. (2000) Spermatogenesis in mice is not
affected by histone H1.1 de®ciency. Exp. Cell Res., 255, 114±124.
Ramaraj, P., Kessler, S.P., Colmenares, C. et al. (1998) Selective restoration of
male fertility in mice lacking angiotensin-converting enzymes by spermspeci®c expression of the testicular isozyme. J. Clin. Invest., 102, 371±
378.
Rane, S.G., Dubus, P., Mettus, R.V. et al. (1999) Loss of Cdk4 expression
causes insulin-de®cient diabetes and Cdk4 activation results in b-islet cell
hyperplasia. Nature Genet., 22, 44±52.
Reijo, R., Seligman, J., Dinulos, M.B. et al. (1996) Mouse autosomal homolog
of DAZ, a candidate male sterility gene in human, is expressed in male
germ cells before and after puberty. Genomics, 35, 346±352.
Reitmair, A.H., Schmits, R., Ewel, A. et al. (1995) MSH2-de®cient mice are
viable and susceptible to lymphoid tumours. Nature Genet., 11, 64±70.
Richardson, L.L., Pedigo, C. and Handel, M.A. (2000) Expression of
deoxyribonucleic acid repair enzymes during spermatogenesis in mice.
Biol. Reprod., 62, 789±796.
Robb, L., Li, R., Hartley, L. et al. (1998) Infertility in female mice lacking the
receptor for interleukin 11 is due to a defective uterine response to
implantation. Nature Med., 4, 303±308.
Robertson, K.M., O'Donnell, L., Jones, M.E.E. et al. (1999) Impairment of
spermatogenesis in mice lacking a functional aromatase (cyp 19) gene.
Proc. Natl Acad. Sci. USA, 96, 7986±7991.
Rocchietti-March, M., Weinbauer, G.F., Page, D.C. et al. (2000) Dazl protein
expression in adult rat testis is up-regulated at meiosis and not hormonally
regulated. Int. J. Androl., 23, 51±56.
Roest, H.P., van Klaveren, J., de Wit, J. et al. (1996) Inactivation of the HR6B
ubiquitin-conjugating DNA repair enzyme in mice causes male sterility
associated with chromatin modi®cation. Cell, 86, 799±810.
Rosenberg, M.P., Aversa, C.R., Wallace, R. et al. (1995) Expression of the vMos oncogene in male meiotic germ cells of transgenic mice results in
metaphase arrest. Cell Growth Diff., 6, 325±336.
Ross, A.J., Waymire, K.G., Moss, J.E. et al. (1998) Testicular degeneration in
bcl-w-de®cient mice. Nature Genet., 18, 251±256.
Rotter, V., Schwartz, D., Almon, E. et al. (1993) Mice with reduced levels of
p53 protein exhibit the testicular giant-cell degenerative syndrome. Proc.
Natl Acad. Sci. USA, 90, 9075±9079.
Rucker, E.B., Dierisseau, P., Wagner, K.U. et al. (2000) Bcl-x and bax
regulate mouse primordial germ cell survival and apoptosis during
embryogenesis. Mol. Endocrinol., 14, 1038±1052.
Rudolph, K.L., Chang, S., Lee H.-W. et al. (1999) Longevity, stress response
and cancer in aging telomerase-de®cient mice. Cell, 96, 701±712.
Rudolph-Owen, L.A., Cannon, P. and Matrisian, L.M. (1998) Overexpression
of the matrix metalloproteinase matrilysin results in premature mammary
gland differentiation and male infertility. Mol. Biol. Cell, 9, 421±435.
Ruggiu, M., Speed, R., Taggart, M. et al. (1997) The mouse Dazla gene
encodes a cytoplasmic protein essential for gametogenesis. Nature, 389,
73±77.
Ruggiu, M., Saunders, P.T.K. and Cooke, H.J. (2000) Dynamic subcellular
distribution of the DAZL protein is con®ned to primate male germ cells. J.
Androl., 21, 470±477.
Sassone-Corsi, P. (2000) CREM: a master-switch regulating the balance
between differentiation and apoptosis in male germ cells. Mol. Reprod.
Dev., 56, 228±229.
Seligman, J. and Page, D.C. (1998) The Dazh gene is expressed in male and
female embryonic gonads before germ cell sex differentiation. Biochem.
Biophys. Res. Commun., 245, 878±882.
Selva, D.M., Tirado, O.M., Toran, N. et al. (2000) Meiotic arrest and germ cell
apoptosis in androgen-binding protein transgenic mice. Endocrinology,
141, 1168±1177.
Shackleton, S., Lloyd, D.J., Jackson, S.N. et al. (2000) LMNA, encoding
lamin A/C, is mutated in partial lipodystrophy. Nature Genet., 24, 153±
156.
Shamsadin, R., Adham, I.M., Nayernia, K. et al. (1999) Males de®cient for
germ-cell cyritestin are infertile. Biol. Reprod., 61, 1445±1451.
Shan, Z., Hirschmann, P., Seebacher, T. et al. (1996) A SPGY copy
homologous to the mouse gene Dazla and the Drosophila gene boule is
autosomal and expressed only in the human male gonad. Hum. Mol.
Genet., 5, 2005±2011.
Shao, C., Deng, L., Henegariu, O. et al. (2000) Chromosome instability
contributes to loss of heterozygosity in mice lacking p53. Proc. Natl Acad.
Sci. USA, 97, 7405±7410.
Sharan, S.K., Morimatsu, M., Albrecht, U. et al. (1997) Embryonic lethality
and radiation hypersensitivity mediated by Rad51 in mice lacking Brca2.
Nature, 386, 804±810.
Sicinski, P., Donaher, J.L., Geng, Y. et al. (1996) Cyclin D2 is an FSHresponsive gene involved in gonadal cell proliferation and oncogenesis.
Nature, 384, 470±474.
Simoni, M. (1994) Transgenic animals in male reproduction research. Exp.
Clin. Endocrinol., 102, 419±433.
Solari, A.J. (1974) The behaviour of the XY pair in mammals. Int. Rev. Cytol.,
38, 273±317.
Steger, K., Klonisch, T., Gavenis, K. et al. (1999) Round spermatids show
normal testis-speci®c H1t but reduced cAMP-responsive element
modulator and transition protein 1 expression in men with roundspermatid maturation arrest. J. Androl., 20, 747±754.
Steger, R.W., Chandrashekar, V., Zhao, W. et al. (1998) Neuroendocrine and
reproductive functions in male mice with targeted disruption of the
prolactin gene. Endocrinology, 139, 3691±3695.
Sun, C., Skaletsky, H., Birren, B. et al. (1999) An azoospermic man with a de
novo point mutation in the Y-chromosomal gene USP9Y. Nature Genet.,
23, 429±432.
Suzuki, M., Abe, K., Yoshinaga, K. et al. (1996) Speci®c arrest of
spermatogenesis caused by apoptotic cell death in transgenic mice.
Genes Cells, 1, 1077±1086.
209
D.Escalier
Swain, A., Narvaez, V., Burgoyne, P. et al. (1998) Dax1 antagonizes Sry
action in mammalian sex determination. Nature, 391, 761±767.
Tanaka, S.S., Toyooka, Y., Akasu, R. et al. (2000) The mouse homolog of
Drosophila Vasa is required for the development of male germ cells.
Genes Dev., 14, 841±853.
Tapanainen, J.S., Vaskivuo, T., Aittomaki, K. et al. (1998) Inactivating FSH
receptor mutations and gonadal dysfunction. Mol. Cell. Endocrinol., 145,
129±135.
Teixeira, L.T., Kiyokawa, H., Peng, X.D. et al. (2000) p27Kip1-de®cient mice
exhibit accelerated growth hormone-releasing hormone (GHRH)-induced
somatotrope proliferation and adenoma formation. Oncogene, 19, 1875±
1884.
TheÂpot, D., Weitzman, J.B., Barra, J. et al. (2000) Targeted disruption of the
murine jun D gene results in multiple defects in male reproduction
function. Development, 127, 143±153.
Tomboe, M. et al. (2000) J. Clin. Endocrinol. Metab., 85, 4013±4018.
Toscani, A., Mettus, R.V., Coupland, R. et al. (1997) Arrest of
spermatogenesis and defective breast development in mice lacking Amyb. Nature, 386, 713±717.
Tourtellotte, W.G., Nagarajan, R., Auyeung, A. et al. (1999) Infertility
associated with incomplete spermatogenic arrest and oligozoospermia in
Egr-4-de®cient mice. Development, 126, 5061±5071.
Tourtellotte, W.G., Nagarajan, R., Bartke, A. et al. (2000) Functional
compensation by Egr4 in Egr1-dependent luteinizing hormone
regulation and Leydig cell steroidogenesis. Mol. Cell. Biol., 20, 5261±
5268.
Toyooka, Y., Tsunekawa, N., Takahashi, Y. et al. (2000) Expression and
intracellular localization of mouse Vasa-homologue protein during germ
cell development. Mech. Dev., 93, 139±149.
Tsai, M.-Y., Chang, S.-Y., Lo, H.-Y. et al. (2000) The expression of DAZL1 in
the ovary of human female fetus. Fertil. Steril., 73, 627±630.
Tsui, S.L., Dai, T., Roettger, S. et al. (2000a) Identi®cation of two novel
proteins that interact with germ-cell-speci®c RNA-binding proteins DAZ
and DAZL1. Genomics, 65, 266±273.
Tsui, S.L., Dai T.N., Warren, S.T. et al. (2000b) Association of the mouse
infertility factor DAZL1 with actively translating polyribosomes. Biol.
Reprod., 62, 1655±1660.
Tsuzuki, T., Fujii, Y., Sakumi, K. et al. (1996) Targeted disruption of the
Rad51 gene leads to lethality in embryonic mice. Proc. Natl Acad. Sci.
USA, 93, 6236±6240.
Upadhya, P., Birkenmeier, E.H., Birkenmeier, C.S. et al. (2000) Mutations in
NIMA-related kinase gene Nek1 cause pleiotropic effects including a
progressive polycystic kidney disease in mice. Proc. Natl Acad. Sci. USA,
97, 217±221.
Varmuza, S., Jurisicova, A., Okano, K. et al. (1999) Spermiogenesis impaired
in mice bearing a targeted mutation in the protein phosphatase 1C gamma
gene. Dev. Biol., 205, 98±110.
Venables, J.P., Elliot, D.J., Makarova, O.V. et al. (2000) RBMY, a probable
human spermatogenesis factor and other hnRNP G proteins interact with
Tra2b and affect splicing. Hum. Mol. Genet., 9, 685±694.
Ventela, S., Okabe, M., Tanaka, H. et al. (2000) Expression of green
¯uorescent protein under beta-actin promoter in living spermatogenic
cells of the mouse: stage speci®c regulation by FSH. Int. J. Androl., 23,
236±242.
Viglietto, G., Dolci, S., Bruni, P. et al. (2000) Glial cell line-derived
neurotrophic factor and neurturin can act as paracrine factors stimulating
DNA synthesis of Ret-expressing spermatogonia. Int. J. Oncol., 16, 689±
694.
Vogel, T., Speed, R.M., Teague, P. et al. (1999) Mice with Y chromosome
deletion and reduced Rbm genes on a heterozygous Dazl1 null background
mimic a human azoospermic factor phenotype. Hum. Reprod., 14, 3023±
3029.
Whitney, M.A., Royle, G., Low, M.J. et al. (1996) Germ cell defects and
hematopoietic hypersensitivity to g-interferon in mice with a targeted
disruption of the Fanconi Anemia C gene. Blood, 88, 49±58.
210
Wong, R.W.-C., Kwan, R.W.-P., Mak, P.H.-S. et al. (2000) Overexpression of
epidermal growth factor induced hypospermatogenesis in transgenic mice.
J. Biol. Chem., 275, 18297±18301.
Wu, J.,Y., Ribar, T.J., Cummings, D.E. et al. (2000) Spermiogenesis and
exchange of basic nuclear proteins are impaired in male germ cells
lacking Camk4. Nature Genet., 25, 448±452.
Wylie, C. (2000) Germ cells. Curr. Opin. Genet. Dev., 10, 410±413.
Xu, J., Qiu, Y., DeMayo, F.J. et al. (1998) Partial hormone resistance in mice
with disruption of the steroid receptor coactivator-1 (SRC-1) gene.
Science, 279, 1922±1925.
Xu, X., Toselli, P.A., Russell, L.D. et al. (1999) Globozoospermia in mice
lacking the casein kinase IIa¢ catalytic subunit. Nature Genet., 23, 118±
121.
Xu, Y., Ashley, T., Brainerd, E.E. et al. (1996) Targeted disruption of ATM
leads to growth retardation, chromosomal fragmentation during meiosis,
immune defects and thymic lymphoma. Genes Dev., 10, 2411±2422.
Yamasaki, L., Jacks, T., Bronson, R. et al. (1996) Tumor induction and tissue
atrophy in mice lacking E2F-1. Cell, 85, 537±548.
Yan, W., Samson, M., Jegou, B. et al. (2000) Bcl-w forms complexes with
Bax and Bak, and elevated ratios of Bax/Bcl-w and Bak/Bcl-w correspond
to spermatogonial and spermatocyte apoptosis in the testis. Mol.
Endocrinol., 14, 682±699.
Yanaka, N., Kobayashi, K., Wakimoto, K. et al. (2000) Insertional mutation of
the murine kisimo locus caused a defect in spermatogenesis. J. Biol.
Chem., 275, 14791±14794.
Yeung, C.H., Sonnenberg-Riethmacher, E. and Cooper, T.G. (1999) Infertile
spermatozoa of c-ros tyrosine kinase receptor knockout mice show
¯agellar angulation and maturational defects in cell volume regulatory
mechanisms. Biol. Reprod., 61, 1062±1069.
Yeung, C.-H., Wagenfeld, A., Nieschlag, E. et al. (2000) The cause of
infertility of male c-ros tyrosine kinase receptor knockout mice. Biol.
Reprod., 63, 612±618.
Ying, Y., Liu, X.M., Marble, A. et al. (2000) Requirement of Bmp8b for the
generation of primordial germ cells in the mouse. Mol. Endocrinol., 14,
1053±1063.
Yoshida, K., Kondoh, G., Matsuda, Y. et al. (1998) The mouse RecA-like gene
Dmc1 is required for homologous chromosome synapsis during meiosis.
Mol. Cell, 1, 707±718.
Yu, R.N., Ito, M., Saunders, T.L. et al. (1998) Role of Ahch in gonadal
development and gametogenesis. Nature Genet., 20, 353±357.
Yu, Y.E., Zhang, Y., Unni, E. et al. (2000) Abnormal spermatogenesis and
reduced fertility in transition nuclear protein 1-de®cient mice. Proc. Natl
Acad. Sci. USA, 97, 4683±4688.
Yuan, L., Liu, J.-G., Zhao, J. et al. (2000) The murine SCP3 gene is required
for synaptonemal complex assembly, chromosome synapsis and male
fertility. Mol. Cell, 5, 73±83.
Zhao, G.-Q., Deng, K., Labosky, P.A. et al. (1996) The gene encoding bone
morhogenetic protein 8B is required for the initiation and maintenance of
spermatogenesis in the mouse. Genes Dev., 10, 1657±1669.
Zhao, G.-Q., Liaw, L. and Hogan, B.L.M. (1998) Bone morphogenetic protein
8A plays a role in the maintenance of spermatogenesis and the integrity of
the epididymis. Development, 125, 1103±1112.
Zhong, J., Peters, A.H., Lee, K. et al. (1999) A double-stranded RNA binding
protein required for activation of repressed messages in mammalian germ
cells. Nature Genet., 22, 171±174.
Zhu, D., Dix, D.J. and Eddy, E.M. (1997) HSP70-2 is required for CDC2
kinase activity in meiosis of mouse spermatocytes. Development, 124,
3007±3014.
Zimmermann, S., Steding, G., Emmen, J.M. et al. (1999) Targeted disruption
of the Insl3 gene causes bilateral cryptorchidism. Mol. Endocrinol., 13,
681±691.
Zindy, F., Deursen, V., Grosveld, G. et al. (2000) INK4d-de®cient mice are
fertile despite testicular atrophy. Mol. Cell. Biol., 20, 372±378.
Received on August 10, 2000; accepted on December 4, 2000