You are on page 1of 15

Cellular and Molecular Life Sciences (2021) 78:3621–3635

https://doi.org/10.1007/s00018-021-03757-2 Cellular and Molecular Life Sciences

ORIGINAL ARTICLE

Targeted disruption of galectin 3 in mice delays the first wave


of spermatogenesis and increases germ cell apoptosis
Tao Lei1,8 · Sandra M. Blois2,3 · Nancy Freitag2,3,4 · Martin Bergmann5 · Sudhanshu Bhushan1 · Eva Wahle1 ·
Annie Chi‑Chun Huang6 · Hung‑Lin Chen6 · Michaela F. Hartmann7 · Stefan A. Wudy7 · Fu‑Tong Liu6 ·
Andreas Meinhardt1 · Monika Fijak1

Received: 15 October 2020 / Revised: 22 December 2020 / Accepted: 6 January 2021 / Published online: 28 January 2021
© The Author(s), under exclusive licence to Springer Nature Switzerland AG part of Springer Nature 2021

Abstract
Galectin 3 is a multifunctional lectin implicated in cellular proliferation, differentiation, adhesion, and apoptosis. This lectin is
broadly expressed in testicular somatic cells and germ cells, and is upregulated during testicular development. Since the role
of galectin 3 in testicular function remains elusive, we aimed to characterize the role of galectin 3 in testicular physiology. We
found that galectin 3 transgenic mice (Lgals3−/−) exhibited significantly decreased testicular weight in adulthood compared
to controls. The transgenic mice also exhibited a delay to the first wave of spermatogenesis, a decrease in the number of germ
cells at postnatal day 5 (P5) and P15, and defective Sertoli cell maturation. Mechanistically, we found that Insulin-like-3 (a
Leydig cell marker) and enzymes involved in steroid biosynthesis were significantly upregulated in adult Lgals3−/− testes.
These observations were accompanied by increased serum testosterone levels. To determine the underlying causes of the
testicular atrophy, we monitored cellular apoptosis. Indeed, adult Lgals3−/− testicular cells exhibited an elevated apoptosis
rate that is likely driven by downregulated Bcl-2 and upregulated Bax and Bak expression, molecules responsible for live/
death cell balance. Moreover, the percentage of testicular macrophages within C ­ D45+ cells was decreased in Lgals3−/− mice.
These data suggest that galectin 3 regulates spermatogenesis initiation and Sertoli cell maturation in part, by preventing
germ cells from undergoing apoptosis and regulating testosterone biosynthesis. Going forward, understanding the role of
galectin 3 in testicular physiology will add important insights into the factors governing the development of germ cells and
steroidogenesis and delineate novel biomarkers of testicular function.

Keywords Galectin 3 · Testis · Spermatogenesis · Sertoli cell maturation · Leydig cells · Apoptosis

Supplementary Information The online version contains


supplementary material available at https​://doi.org/10.1007/s0001​
8-021-03757​-2.

* Monika Fijak Berlin, Corporate Member of Freie Universität Berlin,


monika.fijak@anatomie.med.uni‑giessen.de Humboldt Universität zu Berlin, Berlin, Germany
5
1 Institute of Veterinary Anatomy, Histology, and Embryology,
Department of Anatomy and Cell Biology, Justus-Liebig-
Justus-Liebig-University of Giessen, Giessen, Germany
University of Giessen, Aulweg 123, 35385 Giessen,
6
Germany Institute of Biomedical Sciences, Academia Sinica, Taipei,
2 Taiwan
Department of Obstetrics and Fetal Medicine, AG
7
Glycoimmunology, University Medical Center Steroid Research and Mass Spectrometry Unit, Pediatric
Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Endocrinology and Diabetology, Center of Child
Germany and Adolescent Medicine, Justus-Liebig-University
3 of Giessen, Giessen, Germany
Experimental and Clinical Research Center, A Cooperation
8
between the Max Delbrück Center for Molecular Present Address: Department of Obstetrics and Gynecology,
Medicine in the Helmholtz Association, The Charité Union Hospital, Tongji Medical College, Huazhong
Universitätsmedizin Berlin, Berlin, Germany University of Science and Technology, Wuhan, China
4
Division of General Internal and Psychosomatic Medicine,
Berlin Institute of Health, Charité Universitätsmedizin

13
Vol.:(0123456789)
3622 T. Lei et al.

Introduction trophoblast-matrix interaction and in placental angiogen-


esis [22]. Deficiency in galectin 3 during murine preg-
Mammalian spermatogenesis is a complex process com- nancy leads to placental dysfunction, higher abortion rate
prised of pre-meiotic, meiotic, and spermiogenic stages and fetal growth restriction [23].
[1–3]. During the pre-meiotic stage, spermatogonia prolif- Since galectin 3 is often secreted into biological fluids,
erate and differentiate by mitosis before transforming into its role as an important biomarker of heart or autoimmune
meiotic spermatocytes [2]. Meiosis is a unique process of diseases is frequently discussed [24].
cell division, taking place only in germ cells and leading Taken together, it seems that galectin 3 is an important
to the formation of haploid gametes [4]. After two rounds lectin involved in testicular development and inflammation.
of cell division, the spermatocytes form round spermatids, Yet despite these observations, the testicular function of
which later—after undergoing several morphological and galectin 3 is unknown. In this study, we aimed to investigate
structural changes—transform into elongated spermatids the influence of galectin 3 depletion on key parameters of
[3, 5]. In mice, the first wave of spermatogenesis is ini- testicular function such as germ cell proliferation, differen-
tiated in a few days after birth, and then continues in a tiation and apoptosis; Sertoli cell maturation; Leydig cell
synchronized manner, up to post-natal day (P) 35 [6, 7]. steroidogenesis; and testicular macrophage numbers using
Germ cell types arise in a sequential pattern during the whole body galectin 3 knockout mice Lgals3−/−.
first wave of spermatogenesis in all cords [7]. At P5 the
seminiferous tubules are mainly comprised of spermato-
gonia and immature Sertoli cells [8]. Then, early sper- Materials and methods
matocytes emerge at P9, pachytene spermatocytes at P14
as meiosis is initiated, and round spermatids at P20 [9]. Animals
During this period, the Sertoli cells mature, morphologi-
cally diversify and become functional—as evidenced by Homozygous galectin-3 knockout mice (Lgals3−/−) were
the secretion of testicular fluid to the lumen [10, 11]. The bred on a C57/BL6 genetic background, as described previ-
precise regulation of the proliferation and differentiation ously [25]. Both Lgals3−/− and Lgals3+/+ mice were main-
of these somatic and germ cells during the first wave of tained under standard specific-pathogen-free conditions.
spermatogenesis is essential to ensuring normal fertility. Organ collection in this study was approved by the local
Galectin 3 is a 30-kDa lectin that binds to β-galactoside Animal Ethics Committee (Regierungspräsidium Giessen
[12]. Galectin 3 is involved in various cellular processes, GI Nr. 646_M). For euthanasia, animals were deeply anaes-
including proliferation, differentiation, macrophage activa- thetized by inhalation of 5% isoflurane and sacrificed by
tion, apoptosis regulation, and angiogenesis. Functionally, cervical dislocation.
galectin 3 is best characterized for its role in driving inflam- Testes were collected from neonatal (P5), pre-pubertal
mation, fibrosis, and immune responses [13–15]. This lectin (P15), pubertal (P20), and adult (P35, P50, P100) mice.
is widely expressed in different tissues and developmentally Both testes were removed, weighed, and either snap frozen
regulated. In human, murine and porcine testes galectin 3 in liquid nitrogen for RNA extraction and protein isolation
expression is up-regulated during testicular development or fixed in Bouin’s solution for paraffin embedding. Paraffin
[16–18]. Galectin 3 is expressed in the testis and epididymis, tissue Sects. (5 µm) were subjected to histological analysis
in ejaculated spermatozoa and extracellular vesicles in (hematoxylin–eosin staining) and immunostaining.
human seminal plasma, which serve as vehicles to transfer To measure steroid hormone concentrations, serum sam-
the lectin to the sperm surface during post-testicular matu- ples were collected from mice at P50.
ration. Sperm-bound galectin-3 then plays a critical role in
spermatozoa—zona pellucida binding [16, 19–21]. Gas chromatography tandem mass spectrometry
Furthermore, galectin 3 expression can be upregulated (GC–MS/MS) measurement of steroid hormones
upon treating cultured Sertoli cells with follicle stimulat-
ing hormone (FSH) or epidermal growth factor (EGF) [16]. Up to 100 µL serum was spiked with a cocktail of inter-
Galectin 3 expression also increases in the atrophied semi- nal standards containing [16,16,17-2H3] testosterone (d3-
niferous tubules after vasovasostomy in mice, which could T), [16,16,17-2H3] 5α-androstanediol-3α,17β (d3-AD),
result from testicular inflammation [17]. Indeed, we have [16,16,17-2H3] dihydrotestosterone (d3-DHT) [26]. After
observed galectin 3 upregulation in rat testes from animals equilibration, the samples were extracted with ethyl ace-
with testicular inflammation (unpublished observations). tate and then purified by gel chromatography on Sephadex
In a broader context of reproduction galectin 3 play LH-20 mini columns. Heptafluorobutyric anhydride (Sigma-
a role in embryo implantation, immune regulation, Aldrich) was used for derivatization. Gas chromatography
was performed on an Optima® 1-MS capillary column

13
Targeted disruption of galectin 3 in mice delays the first wave of spermatogenesis and increases… 3623

(25 m × 0.2 mm I.D., df 0.1 µm, Macherey–Nagel, Düren, 2 Zeiss microscope, Carl Zeiss, Göttingen, Germany) and
Germany) housed in a Thermo Scientific Trace 1310 Gas confocal (Leica TCS2, Wetzlar, Germany) microscope.
Chromatograph with a TriPlus RSH Autosampler cou-
pled to a TSQ 800 triple quadrupole MS (Thermo Scien- Histology, immunohistochemistry,
tific, Dreieich, Germany). Helium (1.0 mL/min) was used and morphometry
as the carrier gas. The steroids of interest were eluted at
a rate of 3 °C/min until the column temperature reached Tissue sections were deparaffinized in xylene, hydrated in
250 °C. Quantification was performed in the multiple reac- serial dilutions of ethanol, and treated for antigen retrieval
tion monitoring mode (MRM). The following MRM or by boiling for 20 min in 10 mM sodium citrate buffer. To
m/z ratios were measured for the analytes and their corre- block endogenous peroxidase activity, the sections were
sponding internal standards: m/z 665.1 (668.1) for T (d3- incubated in 1.2% ­H2O2 in methanol for 30 min at RT.
T), m/z 455.3/241.3 (458.3/244.4) for AD (d3-AD) and m/z Then, the sections were blocked with 5% goat serum (Vec-
414.1/185.2 (417.2/188.2) for DHT (d3-DHT). The limits tor Laboratories, Burlingame, USA) in TBST, and incubated
of the quantifications were 0.1 ng/ml for T and AD, and with the indicated primary antibody (Table 1) overnight at
0.03 ng/ml for DHT. 4 °C. An Envision System (DAKO, Hamburg, Germany) for
PCNA or Vectastain ABC Elite Kit (Vector Laboratories)
for HSD11b1 was used to detect primary antibody bind-
Immunofluorescence staining ing, according to the manufacturer`s instructions, combined
with DAB staining. The Sects. (5 µm) were stained with
Testicular paraffin-embedded sections were dewaxed in hematoxylin and eosin (H&E) and evaluated as previously
xylene, rehydrated in decreasing concentrations of ethanol, described [1].
and boiled for antigen retrieval for 20 min in 10 mM sodium For comparative analyses, age-matched animals were
citrate buffer. After blocking with 5% goat serum in TBS chosen. Sections were always processed simultaneously. All
containing 0.1% Tween 20 (TBST), the sections were incu- images were obtained under a Leica DM LB microscope
bated overnight with the indicated primary antibody at 4 ℃ (Leica, Wetzlar, Germany).
(Table 1). The sections were then washed in TBST, and incu- Quantitative microscopic analyses were performed using
bated with the appropriate secondary antibody at room tem- a Leica DM LB microscope (Leica, Wetzlar, Germany).
perature (RT) for 1 h. The nuclei were counterstained with For each mouse, 100 seminiferous tubules in Lgals3−/− and
Topro 3 (Life Technologies, Darmstadt, Germany). Images Lgals3+/+ testicular sections at P15 and P20 were analyzed.
were captured under a conventional fluorescence (Axioplan Each tubule was classified according to lumen formation and

Table 1  Detailed list of antibodies and antibody dilutions used in this study
Primary antibodies Manufacturer Catalogue no Dilution

Rabbit monoclonal anti-Bak Cell signaling technology, Germany 12,105 1:1000*


Rabbit polyclonal anti-Bax Cell signaling technology, Germany 2772 1:1000*
Rabbit monoclonal anti-Bcl-2 Cell signaling technology, Germany 2870 1:1000*
Mouse monoclonal anti-β-actin Ac-15 Sigma-Aldrich, Germany A5441 1:5000*
Rabbit monoclonal anti-galectin 3 GeneTex, USA GTX62084 1:500**
Rat monoclonal anti-germ cell specific antigen (TRA98) Abcam, UK ab82527 1:1000**
Rabbit polyclonal anti-HSD11B1 Abcam, UK ab39364 1:100**
Rabbit polyclonal anti-Insl3 Biorbyt Ltd, UK orb648755 1:100*
Rabbit polyclonal anti-PCNA Abcam, UK ab18197 1:500**
Rabbit polyclonal anti-phospho-histone H3 (Ser10) Cell signaling technology, Germany 9701 1:200**
Rabbit monoclonal anti-Sox9 Abcam, UK ab185230 1:250**
Rabbit monoclonal anti—StAR Cell signaling technology, Germany 8449 1:1000*
Secondary antibodies Manufacturer Catalogue No Dilution
Alexa Fluor 448 goat anti-rabbit IgG Thermo fisher scientific, USA A11008 1:1000**
Alexa Fluor 546 goat anti-rat IgG Thermo fisher scientific, USA A11081 1:1000**
HRP-labelled goat anti-rabbit IgG MP biomedicals, USA 855,676 1:10,000*
HRP-labelled sheep anti-mouse IgG Sigma-Aldrich, Germany A5906 1:10,000*

Antibody dilution used for *Western blotting or **Immunofluorescence/Immunohistochemistry

13
3624 T. Lei et al.

the most differentiated germ cell stage it contained (Sper— Equal amounts of protein were separated by 10% SDS–pol-
spermatogonia, L/Z—leptotene/zygotene spermatocyte, yacrylamide gel electrophoresis and electro-blotted onto
P—pachytene spermatocyte, RS—round spermatid). The nitrocellulose membranes (GE Healthcare, Freiburg, Ger-
percentage of seminiferous tubules of each class was calcu- many). The efficiency of the transfer was monitored by
lated for each mouse and then plotted against age. Ponceau S staining. The membranes were blocked with 5%
BSA in TBST for 1 h before incubation overnight with the
TUNEL assay appropriate primary antibody (Table 1). Then, the mem-
branes were washed and incubated with the appropriate
Apoptosis was quantified by the TUNEL method (ApopTag secondary HRP-conjugated antibody (Table 1). The sig-
7100 Kit, Chemicon, Hofheim, Germany) according to the nals were visualized using ECL (Millipore Corporation,
manufacturer’s instructions. Counterstaining was carried Billerica, USA) and analyzed by densitometry (Fusion FX,
out with hematoxylin. Counting was performed in a blinded Witec AG, Luzern, Switzerland).
fashion. All tubules of one testicle cross-section were ana-
lyzed by counting the total number of tubules as well as
the total number of tubules with at least one TUNEL-posi- RNA extraction and quantitative PCR
tive nucleus. Results are presented as percentage (positive
tubules/all tubules × 100) (n = 3 per group). Total RNA was isolated using an RNeasy Mini kit (Qiagen,
Hilden, Germany) according to the manufacturer’s instruc-
Western blotting tions. Quantitative PCR was performed on a CFX96 Touch
thermal cycler (Bio-Rad, Munich, Germany) using iTaq Uni-
Testes or cells were homogenized in ice-cold RIPA buffer versal SYBR Green Supermix (Bio-Rad, Munich, Germany).
(50 mM Tris pH 8.0, 1% Nonidet P 40, 0.5% deoxycho- The primer sequences and amplicon sizes are provided in
late, 0.1% SDS, 150 mM NaCl) containing a 1% protease Table 2. Relative gene expression was calculated by using
inhibitor cocktail (Sigma-Aldrich, Steinheim, Germany). the ΔΔCt method [27].

Table 2  List of PCR primers Gene Primer (5` → 3`) Entrez gene ID Amplicon
used in this study size (bp)

Actb F: TGA​CAG​GAT​GCA​GAA​GGA​GAT​ 11,461 156


R: TAC​TCC​TGC​TTG​CTG​ATC​CAC​
Amh F: CGA​GCT​CTT​GCT​GAA​GTT​CC 11,705 302
R: TGA​AAC​AGC​GGG​AAT​CAG​AG
Cyp11a1 F: CCA​GTG​TCC​CCA​TGC​TCA​AC 13,070 74
R: TGC​ATG​GTC​CTT​CCA​GGT​CT
Cyp17a1 F: GCC​CAA​GTC​AAA​GAC​ACC​TAAT​ 13,074 159
R: GTA​CCC​AGG​CGA​AGA​GAA​TA
Ddx4 F: GGT​CCA​AAA​GTG​ACA​TAT​ATA​CCC​ 13,206 140
R: TTG​GTT​GAT​CAG​TTC​TCG​AGT​
Gapdh F: TGA​CGT​GCC​GCC​TGG​AGA​AA 14,433 98
R: AGT​GTA​GCC​CAA​GAT​GCC​CTT​CAG​
Hprt F: CTG​GTG​AAA​AGG​ACCTC​ 15,452 110
R: CTG​AAG​TAC​TCA​TTA​TAG​TCAAG​
Hsd3b1 F: TGG​ACA​AAG​TAT​TCC​GAC​CAGA​ 15,492 250
R: GGC​ACA​CTT​GCT​TGA​ACA​CAG​
Insl3 F: TGC​AGT​GGC​TAG​AGC​AGA​GA 16,336 151
R: GTG​CAG​CCA​GTA​AGA​CAG​CA
Lgals3 F: GAT​CAC​AAT​CAT​GGG​CAC​AG 16,854 100
R: ATT​GAA​GCG​GGG​GTT​AAA​GT
Star F: TGC​CCA​TCA​TTT​CAT​TCA​TCCTT​ 76,205 232
R: AAA​AGC​GGT​TTC​TCA​CTC​TCC​
Wt1 F: ACC​CAG​GCT​GCA​ATA​AGA​GA 22,431 230
R: CCT​GGT​GTG​GGT​CTT​CAG​AT

13
Targeted disruption of galectin 3 in mice delays the first wave of spermatogenesis and increases… 3625

Transfection of MLTC‑1 cell line with Lgals3 siRNA (clone REA293; Miltenyi Biotech). Finally, the cells were
washed in washing buffer. Data were collected for 700,000
A mouse Leydig tumor cell line (MLTC-1) was cultured events using a MACSQuant Analyzer 10 flow cytometer
in ATCC-formulated RPMI-1640 (Gibco, Darmstadt, Ger- and analyzed using MACSQuantify software version 2.11
many) supplemented with 10% fetal bovine serum, 100 U/ (Miltenyi Biotech).
mL penicillin, and 0.1 mg/mL streptomycin. Cell cultures
were maintained in a 5% ­CO2 humidified atmosphere at
Statistical analyses
37 °C. The cell line was kindly provided by Prof. Thomas
Linn and Dr. Qingkui Jiang (Clinical Research Unit, Cen-
The data represent the means ± SEM. Comparisons between
tre of Internal Medicine, Justus-Liebig-University, Giessen,
two groups were made by two-tailed t test. P values < 0.05
Germany). A commercially available, pre-designed short
were considered statistically significant. All tests were per-
interfering RNA (siRNA) against Lgals3 and Silencer®
formed using GraphPad Prism 5 software (GraphPad Soft-
select negative control siRNA was purchased from Life
ware, San Diego, USA).
Technologies GmbH (Darmstadt, Germany). Transient trans-
fection was performed using L ­ ipofectamine® RNAiMAX
Transfection Reagent (Thermo Fisher Scientific, Waltham,
USA), according to the manufacturer’s instructions. Briefly, Results
1 × 105 MLTC-1 cells were seeded in a 12-well plate and
transfected at 60–80% confluency with 15 pmol siRNA and Loss of galectin 3 delays the first wave
4.5 µl Lipofectamine® RNAiMAX per well. After 48 h incu- of spermatogenesis in Lgals3−/− mice
bation, the cells were treated with 1 IU/ml hCG (Predalon
5000 I.E. Organon, Oberschleissheim, Germany) for 24 h. To investigate the effects of endogenous galectin 3 on tes-
The supernatants and cell lysates were collected to measure ticular development, we harnessed Lgals3−/− mice [25].
testosterone levels and Western blot analyses, respectively. These mice have been previously extensively studied in the
context of fibrosis, inflammation, heart failure, atheroscle-
Testosterone measurement rosis or cancer but not for the effects of galectin 3 deficiency
on male reproduction and fertility [24]. In our first analy-
The levels of testosterone in the cell culture supernatants ses, testis weights were determined from mice at different
were measured using a testosterone ELISA kit (IBL Interna- time points after birth and in adulthood. Testicular weights
tional, Hamburg, Germany), according to the manufacturer’s increased progressively with age in mice of all genotypes
protocol. but were significantly decreased in adult Lgals3 −/− mice
(P50 and P100) compared to age-matched Lgals3+/+ con-
Flow cytometry trols (Fig. 1a).
To characterize testicular development and the onset of
Decapsulated testes were incubated with 1.2 mg/ml col- spermatogenesis in Lgals3−/− testes, we performed a his-
lagenase A and 15 U/ml DNase (Roche Diagnostics, Man- tological analysis of the testes at P5, P15 and P20. At P5,
nheim, Germany) in PBS in a shaking water bath at 34 °C the seminiferous tubules from Lgals3+/+ and Lgals3−/− tes-
for 15 min. The enzymes were inactivated by adding ice-cold tes were mainly composed of Sertoli cells and spermato-
PBS, and the tubule fragments were allowed to settle for gonia (Fig. 1b i–ii). We also observed gonocytes (G) in
4 min. Then, the supernatant was filtered and centrifuged at both genotypes (Fig. 1b i–ii). By P15, meiosis progressed
300 × g for 10 min at 4 °C. The pellet was washed with PBS such that pachytene spermatocytes (P) were now present
and the erythrocytes were depleted by osmotic lysis using in the Lgals3+/+ testes. However, most of the differentiated
red blood cell lysis buffer (Qiagen, Hilden, Germany) for germ cells in Lgals3−/− mice were composed of zygotene
5 min at RT. The final cell suspension was washed in wash- spermatocytes (Z), with numerous tubules containing only
ing buffer (0.5% BSA and 2 mM EDTA in PBS) and then spermatogonia (Fig. 1b iii–iv). Finally, by P20, we observed
processed directly for flow cytometric macrophage staining. signs that the next step of spermatogenesis had occurred
Interstitial cells (1 × 106) were incubated with the following in Lgals3+/+ mice, as evidenced by the presence of round
fluorochrome conjugated recombinant REAfinity antibod- spermatids (RS) in the seminiferous tubules (Fig. 1b v)
ies: CD45 -VioGreen (clone REA737), CD11b -VioBright indicating complete passage through meiosis. By contrast,
FITC (clone REA592), Ly6C -PE (clone REA796), and most tubules in age-matched Lgals3−/− testes contained only
F4/80 -APC (clone REA126) (all from Miltenyi Biotech, pachytene spermatocytes, a mid-meiotic stage (Fig. 1b vi)
Bergisch-Gladbach, Germany). Background staining was As such, it seems that loss of galectin 3 delays the first wave
evaluated using the appropriate REA control antibodies of spermatogenesis in Lgals3−/− mice.

13
3626 T. Lei et al.

Fig. 1  The first wave of spermatogenesis is delayed in Lgals3−/− was quantified according to the most differentiated germ cell stage
mice. a Testicular weight (mg) collected from Lgals3+/+ and visible (G gonocytes, L leptotene spermatocyte, P pachytene sper-
Lgals3−/− mice at postnatal (P) days 5, 15, 20, 35, 50 and 100; matocyte, RS round spermatid, S Sertoli cell, Sper spermatogonia,
(n = 4, * P < 0.05). b Representative histology of mouse testes from Z zygotene spermatocyte) at P15 (c) and P20 (d); (n = 3, *P < 0.05,
Lgals3+/+ (i, iii, v) and Lgals3−/− mice (ii, iv, vi) at P5 (i, ii), P15 (iii, **P < 0.01, ***P < 0.001). The data represent the means ± SEM
iv) and P20 (v, vi). (C and D) The percentage of tubule cross sections

To confirm these findings, we aimed to quantify sper- tubules containing less-differentiated stages of spermato-
matogenesis in testes from Lgals3+/+ and Lgals3−/− animals genesis (such as leptotene/zygotene spermatocytes or sper-
at P15 and P20 (Fig. 1c, d). To do so, we determined the matogonia) was increased in Lgals3−/− testes compared to
percentage of seminiferous tubules containing the most Lgals3+/+ testes (14% vs. 3.7% and 18% vs. 0%, respectively;
advanced stage of spermatogenesis in each section. At P15, Fig. 1d). Taken together, these data illustrate that the first
we found that the percentage of tubules containing pachy- wave of spermatogenesis in Lgals3−/− testes is delayed.
tene spermatocytes was significantly lower in Lgals3−/− tes-
tes as compared to Lgals3+/+ testes (9% vs. 23%, respec- Germ cell number is reduced in neonatal Lgals3−/−
tively; Fig. 1c). Subsequently, at P20, the percentage of testes, independently of cellular proliferation
tubules containing round spermatids and pachytene sper-
matocytes in Lgals3−/− testes was also significantly lower We next wanted to investigate the underlying causes of
compared with Lgals3+/+ testes (3.6% vs. 17% and 53% vs. this apparent delay of the first wave of spermatogenesis in
82%, respectively; Fig. 1d). Conversely, the percentage of Lgals3−/− testes. To do so, we assayed germ cell numbers

13
Targeted disruption of galectin 3 in mice delays the first wave of spermatogenesis and increases… 3627

and germ and somatic cell proliferation by immunostaining Next we studied the expression of proliferating cell
with the germ cell marker Tra98 and the mitosis marker nuclear antigen (PCNA)—a marker of cell proliferation.
phospho-histone H3 Ser10 (PH3) during early postnatal tes- At P5, PCNA staining revealed no overt differences in
tis development. We found that Lgals3−/− testes had nearly the proliferation of testicular cells between Lgals3+/+ and
a twofold lower number of germ cells at P5 compared to Lgals3−/− mice (Supplementary Fig. 1). By contrast, several
Lgals3+/+ control testes (Fig. 2a–g). seminiferous tubules were found without proliferating cells
We also monitored Ddx4 (VASA) expression, which in Lgals3−/− testes at P15 and P20 (Supplementary Fig. 1).
serves as a germ cell marker during germ cell differen- Together, these findings point to a reduced number of gono-
tiation [28]. Here we found that Ddx4 mRNA expression cytes and spermatogonia that is independent of proliferation,
was downregulated in neonatal (P5) and prepubescent in the neonatal Lgals3−/− testis.
(P15) Lgals3−/− testes compared to control Lgals3+/+ tes-
tes (Fig. 2h), which strongly supports a reduction in germ Germ cells are less frequent in the seminiferous
cell numbers in Lgals3−/− testes. By contrast, the number of tubules of neonatal and pubertal ­Lgals3−/− testes
dividing germ cells (PH3- and Tra98-positive) and somatic
cells (PH3-positive) in Lgals3−/− testes was unaffected The PCNA staining results described above showed in sev-
(Fig. 2f, g). eral tubules no sign of cell proliferation in P15 and P20

Fig. 2  Testicular germ cell numbers are reduced in neonatal (a cell proliferation marker). Nuclei were counterstained with Topro
Lgals3−/− testes. a–d Identification of germ cell numbers in Lgals3+/+ 3 (blue). e–g The numbers of germ cells (e), PH3-positive germ cells
and Lgals3−/− testes using Tra98 immunofluorescence staining (red) (f) and somatic cells (g) were normalized to the total cross-section
at P5 (a–d). Phosphorylated histone 3 staining (PH3, green; b, c, d) area ­(mm2) in both Lgals3+/+ and Lgals3−/− testes. h Ddx4 mRNA
and Tra98 staining were used to distinguish between dividing germ expression (germ cell marker) of P5 and P15 testes was normal-
and somatic cells (a, c, d). Part d shows the magnified areas iden- ized to Actb and Gapdh; (n = 3–4, *P < 0.05). The data represent the
tified by the white boxes in part (c). The thin arrow indicates germ means ± SEM
cells; the thick arrow indicates somatic cells, both positive for PH3

13
3628 T. Lei et al.

Lgals3−/− testes. To decipher a possible influence of endog- suggests that the number of Sertoli cells is unaffected in
enous galectin 3 on germ cell development, we stained testes Lgals3−/− mice. As such, while Lgals3−/− testes display
from neonatal (P5), pre-pubertal (P15) and pubertal (P20) altered Sertoli cell maturation, the overall number of Sertoli
Lgals3+/+ and Lgals3−/− mice with the Sertoli cell marker cells is unaffected.
Sox9 and the germ cell marker Tra98. Our quantitative
analyses revealed that in P5 Lgals3−/− testes, the number of
Lgals3−/− testicular cells exhibit an increased rate
seminiferous tubules without germ cells was significantly
of apoptosis accompanied by mild atrophy
increased as compared to Lgals3+/+ testes (46% vs. 13%,
respectively). In Lgals3+/+ testes some tubules were entirely
To determine the underlying causes of testicular atrophy
free of germ cells (Fig. 3a–e), an observation not seen from
in adult Lgals3−/− mice, we performed TUNEL staining
P15 onwards when all seminiferous tubules showed germ
to detect the level of cellular apoptosis. Quantifications of
cells. Conversely, 8% of tubules at P15 and ~ 15% of tubules
TUNEL-positive cells revealed higher numbers of apoptotic
at P20 in Lgals3−/− mice had no germ cells (Fig. 3e). By
cells in adult Lgals3−/− testes compared to Lgals3+/+ testes at
P35, however, all seminiferous tubules in Lgals3−/− tes-
P5, P35, P50 and P100 (Fig. 4a–c). Due to the fact that ~ 15%
tes contained germ cells. These results indicate that the
of tubules at P20 in Lgals3−/− mice contained no germ cells
number of seminiferous tubules containing germ cells in
analysis of TUNEL positive cells had a bias and was conse-
Lgals3−/− mice is reduced during the neonatal and prepu-
quently not included.
bescent periods.
Galectin 3 can stabilize the mitochondrial membrane by
binding to the anti-apoptotic protein Bcl-2; it can also inhibit
Lgals3−/− testes display defects in Sertoli cell
cytochrome c release by blocking the activity of the pro-
maturation
apoptotic proteins Bak and Bax [30–32] (Fig. 4d). We thus
investigated the expression of these pro- and anti-apoptotic
Sertoli cell maturation and concomitant fluid secretion
proteins in Lgals3−/− and Lgals3+/+ testes at P50. Bcl-2 was
results in the formation of the lumen of the seminiferous
downregulated, while Bak and Bax were upregulated in
tubules during testicular development [29]. To characterize
Lgals3−/− testes, as compared to Lgals3+/+ testes (Fig. 4e–h).
the effects of Lgals3 gene disruption on Sertoli cell matura-
These data indicate that an increase in the number of apop-
tion, we analyzed lumen formation at P15 and performed a
totic cells in adult Lgals3−/− mouse testes results from an
quantitative analysis of the Sertoli cell maturation marker
imbalance in anti-/pro-apoptotic protein expression. This
anti-Müllerian hormone (Amh) [11]. The percentage of
perturbation might contribute to mild testicular atrophy in
tubules containing a lumen in Lgals3−/− testes was two-fold
Lgals3−/− mice.
lower compared to in Lgals3+/+ control tubules (Fig. 3f–g).
Moreover, our results showed that Amh mRNA was highly
expressed in the neonatal period (P5) in testes from both Gonadal Insl3 and steroidogenic enzymes
Lgals3+/+ and Lgals−/− mice (Fig. 3h) and then decreased at and circulating testosterone levels are upregulated
P15 before reaching extremely low levels at P20 and P35 in in Lgals3−/− mice
both strains. These findings confirm the progressive matura-
tion process of Sertoli cells (Fig. 3h). Interestingly, although Production of galectin 3 in granulosa cells is regulated by
slightly decreased compared to P5, the levels of Amh mRNA luteotropic hormone (LH), which indicates an involvement
expression in Lgals3−/− testes at P15 were three-fold higher of galectin 3 in steroidogenesis [33]. Similar to granulosa
as compared to Lgals3+/+ control testes, indicating a delay to cells, the predominant function of testicular Leydig cells
Sertoli cell maturation in Lgals3−/− testes (Fig. 3h). is sex hormone production (testosterone) through a steroi-
We next studied the effects of galectin 3 on Sertoli cell dogenic process [34]. To investigate whether Lgals3 gene
number, and the ratio between Sertoli and germ cells in the ablation affects Leydig cell function, we analyzed the gene
developing testes. To do so, we performed an immunostain- and protein expression of Insulin-like factor 3 (Insl3) and
ing using Sox9 and Tra98 antibodies at P15. Although not steroidogenic enzymes (Cyp11a1, Hsd3b1 and Cyp17a1) in
quantified, the immunostaining revealed no obvious differ- adult Lgals3−/− and Lgals3+/+ mice by quantitative RT-PCR
ence in Sox9 expression between Lgals3−/− and Lgals3+/+ and Western blotting.
testes at P15 (Supplementary Fig. 2). Compared to Lgals3+/+ mice, the Cyp11a1 and Hsd3b1
Wilms’ tumor gene (Wt1) is considered a stable marker mRNA expression levels were significantly upregu-
of Sertoli cells during testicular development [11]. Our lated in adult Lgals3−/− mice at P35 and P100 (Fig. 5a);
analyses of Wt1 mRNA expression showed that the expres- Cyp17a1 mRNA expression was significantly increased in
sion levels in Lgals3−/− testes were comparable to those in Lgals3−/− mice testes at P50 and P100 (Fig. 5a).
Lgals3+/+ testes at P15 (Supplementary Fig. 2). This finding

13
Targeted disruption of galectin 3 in mice delays the first wave of spermatogenesis and increases… 3629

Fig. 3  Testes from Lgals3−/− mice exhibit a higher number of semi- Lgals3+/+ and Lgals3−/− mice at P15. The lower panel (iii, iv, v, vi)
niferous tubules without germ cells and altered maturation of Sertoli shows the magnified areas from (i) and (ii) containing seminiferous
cells. a–d Representative immunofluorescent labeling of germ cells tubules with (iii, v) or without (iv, vi) a lumen in Lgals3+/+ (i) and
with Tra98 (red; a, c, d) and Sertoli cells with Sox9 (green; b–d) Lgals3−/− (ii) testes, respectively. f–g The percentage of seminiferous
in testes from Lgals3+/+ and Lgals3−/− mice at P5. Part d shows the tubules without (f) and with (g) a lumen was quantified by counting at
magnified areas identified by the white boxes in part (c). The white least 100 seminiferous tubule cross sections in each testis. (H) Rela-
stars indicate the seminiferous tubules without germ cells. The enu- tive Amh mRNA expression in testes from Lgals3+/+ and Lgals3−/−
meration of tubules containing germ cells was performed on P5, P15 mice at P5, P15, P20 and P35 was normalized to Actb and Gapdh;
and P20 (i). At least 100 seminiferous tubules were counted in each (n = 3, *P < 0.05, **P < 0.01). The data represent the means ± SEM
testis; (n = 3, *P < 0.05). e Representative HE staining of testes from

Expression of Insl3 in adult testes is an indicator of the regulatory protein StAR were also elevated in Lgals3−/− tes-
number and differentiation status of the Leydig cells [35]. tes compared to control Lgals3+/+ testes at P50 (Fig. 5c).
In Lgals3 −/− mice testes, Insl3 mRNA expression was By using Leydig cell staining with an antibody against
increased at P35, P50 and P100 (Fig. 5b). In agreement 11β-hydroxysteroid dehydrogenase type 1 (HSD11B1) no
with the mRNA data, Insl3 and the steroidogenic acute

13
3630 T. Lei et al.

Fig. 4  Testicular cells in Lgals3−/− mice experience increased apop- by binding to Bcl-2 and inhibiting cytochrome c release. Bcl-2 inhib-
tosis due to an imbalance of anti- and pro-apoptotic proteins. a, b its activity of pro-apoptotic proteins Bak and Bax. Western blot e and
TUNEL staining (arrows) of testicular sections from Lgals3+/+ mice densitometric analysis of Bcl-2 f Bak g and Bax h expression in testes
and Lgals3−/− mice at P50. c The numbers of apoptotic germ cells from Lgals3+/+ and Lgals3−/− mice at P50 is shown; (n = 3, *P < 0.05;
were quantified in 100 tubular cross sections in testes from Lgals3+/+ **P < 0.01; ***P < 0.001). β-actin was used as a loading control. The
and Lgals3−/− mice at P5, P15, P35, P50 and P100. d A scheme is data represent the means ± SEM
illustrating how galectin 3 can stabilize the mitochondrial membrane

differences in the cell numbers between transgenic and WT in which we depleted Lgals3 with siRNA. The efficiency of
animal groups at P50 were apparent (Supplementary Fig. 3). Lgals3 siRNA silencing is shown in Supplementary Fig. 4.
Next, we investigated whether the increased expression of We stimulated testosterone production by incubating the
steroidogenic enzymes affected the concentration of steroids cells with hCG [36], and found that StAR protein levels
in the serum. To do so, we used a GC–MS/MS method to and testosterone secretion were upregulated in both Lgals3-
measure testosterone, androstandiol-3a, 17b (AD) and dihy- depleted and control cells (Fig. 5e–f). However, Lgals3
drotestosterone (DHT) levels in sera taken from Lgals3+/+ silencing led to a stronger increase in StAR expression and
and Lgals3−/− mice. Here, testosterone and AD levels were testosterone production under hCG stimulation compared to
increased in adult Lgals3−/− mice as compared to Lgals3+/+ control cells (Fig. 5e–f). These results indicate an inhibitory
mice (Fig. 5d); no significant change was found in DHT sera effect of endogenous galectin 3 on testicular steroidogenesis.
levels (Fig. 5d). Taken together, these results suggest that sex
hormone production is increased in Lgals3−/− mice. Loss of Lgals3 results in a depletion of testicular
macrophages
Lgals3 depletion in cultured Leydig tumor cells
upregulates StAR and testosterone production Galectin 3 is considered a chemoattractant for mac-
rophages under physiological conditions, and can also
Our data thus far suggest that galectin 3 affects the steroido- accelerate M2 macrophage infiltration in tumor environ-
genic activity of Leydig cells. To understand how galectin 3 ments [37, 38]. Moreover, the macrophages possess a
affects Leydig cell function, we determined the level of tes- unique function in the maintenance of testicular immune
tosterone secretion in a cultured Leydig cell line (MLTC-1) privilege and support spermatogonial differentiation and

13
Targeted disruption of galectin 3 in mice delays the first wave of spermatogenesis and increases… 3631

Fig. 5  Expression of steroidogenic enzymes and Insl3 is upregulated one, androstandiol-3a, 17b, and dihydrotestosterone (DHT) concen-
in Lgals3−/− testes. a–b The relative mRNA expression of Cyp11a1, trations in the sera from Lgals3+/+ and Lgals3−/− mice at P50, deter-
Hsd3b1, Cyp17a1 (A) and Insl3 b in Lgals3+/+ and Lgals3−/− testes mined by GC–MS/MS; (n = 3–4, *P < 0.05, **P < 0.01). e–f StAR
was analyzed by real-time RT-PCR at P35, P50 and P100. The right expression and testosterone levels in galectin 3-depleted MLTC-1
panel shows the main pathway of steroidogenesis in rodents, with cells stimulated with 1 IU/ml hCG for 24 h. StAR protein expression
the corresponding enzymes highlighted by a dotted frame. c West- in the cell lysates e and the testosterone concentrations in conditioned
ern blot analysis of StAR and Insl3 protein expression in testes from media (F) were determined by Western blotting and ELISA, respec-
Lgals3+/+ and Lgals3−/− mice at P50; (n = 3, *P < 0.05, ***P < 0.001, tively. Representative data are shown; (n = 3–4, *P < 0.05, **P < 0.01,
****P < 0.0001). β-actin was used as a loading control. (D) Testoster- ***P < 0.001, ****P < 0.0001). The data represent the means ± SEM

Leydig cell steroidogenesis [39, 40]. Importantly for our by 11% in adult Lgals3 −/− mice compared to control
research, testicular macrophages express galectin 3 [18]. Lgals3+/+ (Fig. 6a–b). In sum, these data support that the
In our final analyses, we thus investigated whether the endogenous galectin 3 regulates the number of testicular
macrophage population was affected in Lgals3−/− testes. macrophages and their depletion may lead to disturbances
To do so, we performed flow cytometry using an extended in the spermatogenesis (via affecting spermatogonia) and
antibody panel (CD45, Ly6C, F4/80 and CD11b) to target steroidogenesis.
testicular macrophages (Supplementary Fig. 5). Within the Overall, our results indicate that endogenous galectin
­ D45+ leukocytes, we defined res-
population of testicular C 3 plays an important regulatory role in the development
ident macrophages as ­Ly6C−F4/80+CD11b+. The percent- of germ cells, maturation of Sertoli cells, steroidogenesis
age of testicular macrophages was significantly decreased

13
3632 T. Lei et al.

Fig. 6  The percentage of testicular macrophages in the population and Lgals3−/− testes at P50. A representative dot plot a and the cor-
of ­CD45+ leukocytes is decreased in Lgals3−/− mice. a–b Testicular responding quantification b is shown; (n = 4–6, *P < 0.05). The data
macrophage frequency was assessed by flow cytometry in Lgals3+/+ represent the means ± SEM

and maintenance of the macrophages niche, all comprising seems to have a more complex multilayered influence on
essential testicular functions. reproduction. In this regard, in human seminal plasma
galectin 3 is associated with extracellular vesicles, which
are responsible for immunosuppression and regulation of
Discussion sperm function in the female reproductive tract [19]. Fur-
thermore, the acrosomal region of capacitated human sper-
Galectin 3 is widely expressed in testicular cells includ- matozoa shows strong galectin 3 labelling, which mediates
ing Sertoli, peritubular, Leydig, and germ cells and its zona pellucida binding and affects fertilization in vitro [21].
upregulation is associated with testicular maturation [16, Moreover, in our study we observed altered Sertoli cell
18]. However, the role of endogenous galectin 3 in male maturation kinetics in Lgals3−/− testes, whereby the lumen
gonadal development is unknown. Our results show for the formation and the loss of Amh mRNA expression was
first time that mice lacking galectin 3 expression experience delayed compared to WT testes. In maturating Sertoli cells
a delay to the first wave of spermatogenesis, as evidenced by AMH expression is progressively reduced reaching almost
a decrease in the number of germ cells during the neonatal undetectable levels during transition to adulthood [46].
period and defects in Sertoli cell maturation. Moreover, we Galectin 3 expression in Sertoli cells is regulated by several
found evidence for increased Leydig cell marker and steroi- factors, including EGF, TNF, FSH, and adult germ cells,
dogenic enzyme expression, elevated testosterone produc- which suggests that galectin 3 expression may affect Sertoli
tion, an increased rate of germ cell apoptosis, and lower cell function [16]. Our data support that loss of galectin 3 in
numbers of testicular macrophages in adult Lgals3−/− testes. Sertoli cells could cause a delay to Sertoli cell maturation
Overall, these changes are accompanied by mild testicular during the neonatal period, followed by a decrease in the
atrophy. Together, these findings all point to an important number of germ cells. Abnormal development, proliferation
role of endogenous galectin 3 in testicular physiology. and/or maturation of Sertoli cells in early life could lead to
Our initial analyses identified a significant reduction in testicular dysfunction in adult life [11]. Such disruption in
testicular weight in adult Lgals3−/− mice compared to age- the Sertoli cell maturation has been associated with testicu-
matched WT controls. We propose that the delay to the first lar dysgenesis syndrome [11, 46]. Conversely, the absence of
wave of spermatogenesis in these transgenic animals may be germ cells in animal models and in human patients results in
responsible for this decrease in organ mass. Similar observa- a delay to Sertoli cell maturation, but has no effect on their
tions have been shown for regulatory genes, such as Rhox13, final maturation (reviewed in [11]).
TR4, Rnf138, and Tap73 where a delay of the first wave We also observed an increase in the number of
of spermatogenesis is accompanied by a reduced testicular tubules without germ cells in both neonatal and pubertal
weight that persists into adulthood in mice [41–44]. Lgals3−/− testes, which mirrors the reduced number of
In addition, Lgals3−/− mice produce lower numbers of germ cells in the neonatal period. Chiarini-Garcia et al.
germ cells during the neonatal period. This is similar to mice reported that undifferentiated spermatogonia can migrate
lacking a functional Taf4b gene that are also characterized to ensure the uniform distribution of germ cell progeny at
by reduced germ cell numbers during late embryogenesis the basement of the seminiferous tubules [47]. A study by
and the neonatal period, which subsequently led to a delay Jahnukainen et al. showed that after irradiation, spermato-
to meiotic initiation [45]. Beside its influence on germ cell genesis can recover in single testicular lobules in primate
development or spermatogenic differentiation, galectin 3 testes, indicating that spermatogenic stem cells are capable

13
Targeted disruption of galectin 3 in mice delays the first wave of spermatogenesis and increases… 3633

of re-colonizing their respective loop of the seminiferous percentage of testicular macrophages within the population
tubules [48]. Therefore, we hypothesize that re-colonization of ­CD45+ leukocytes in adult Lgals3−/− mice. Interestingly,
by undifferentiated spermatogonia may explain our obser- subpopulations of testicular macrophages called “peritubular
vation that many tubules were seen without germ cells in macrophages” due to their close proximity to peritubular
neonatal and prepubescent Lgals3−/− testes, while they were cells near the seminiferous tubule wall, express factors such
eventually replenished in adulthood. However, testicular as CSF1 which are involved in spermatogonial proliferation
weight is affected in a long term and persists into adulthood, and differentiation, a finding that can explain reduced num-
an observation that can be explained by the higher number bers of spermatogonia in macrophage depleted testis [39]. It
of apoptotic germ cell in Lgals3−/− testis. can therefore be hypothesized that the reduced numbers of
In female mice, galectin 3 expression is increased dur- testicular macrophages in Lgals3−/− testis at least partially
ing luteolysis and is associated with a loss of progesterone contribute to the mild testicular atrophy seen in adulthood
synthesis in vivo, which can be reversed by administration in these mice. On the other hand, interstitial macrophages,
of hCG [33]. In this line, galectin 3 is downregulated in which are intimately connected to Leydig cells produce fac-
hCG-stimulated human granulosa cells [33]. In male mice, tors, e.g. 25-hydroxycholesterol, which could influence the
galectin 3 is expressed in Leydig cells [49]. Interestingly, we steroidogenesis of Leydig cells [60]. 25-hydroxycholesterol
found an upregulated expression of the Leydig cell marker from testicular macrophages is involved in the differentia-
Insl3 and steroidogenic enzymes (Cyp11a1, Hsd3b1 and tion of Leydig cells to fully functional steroidogenic cells
Cyp17a1) in adult Lgals3−/− testes with concomitant ele- that occurs during post-natal maturation of the testis [61], a
vated serum levels of testosterone and DHT. To determine process in which macrophage numbers increase [62]. Here,
the relationship between galectin 3 and testosterone produc- we investigated the total number of macrophages in adult
tion, we knocked down galectin 3 in the MLTC-1 Leydig testis without further division into subpopulations. How-
cell line using Lgals3 siRNA. Following hCG stimulation, ever, based on our in vitro results using a galectin-3 depleted
StAR expression and testosterone secretion were found MLTC-1 cell line, the increased expression of StAR protein
increased in these cells. These data indicate that Lgals3 has followed by an enhanced secretion of testosterone points
an inhibitory effect on hCG-stimulated testosterone produc- to a direct influence of this lectin on Leydig cell steroido-
tion in Leydig cells, although in our study we cannot exclude genesis, independent of an interaction with macrophages.
that high levels of LH may also play a role in the increased Furthermore, the testosterone concentration in the serum
androgen levels in Lgals3−/− mice. is increased in Lgals3−/− mice which also suggests a com-
We also revealed an increased number of apoptotic cells pensatory mechanism to maintain normal spermatogenesis.
in adult Lgals3−/− testes that is likely due to a perturbed Furthermore, these findings are in line with other studies
Bcl-2, Bax and Bak expression ratio. Data from previous demonstrating an increased expression of Star, Cyp11a1 and
studies have suggested that galectin 3 can form heterodimers Cyp11b1 in the thymus followed by higher levels of corticos-
with Bcl-2 and thus stabilize the mitochondrial membrane terone in sera and thymus of galectin 3 deficient mice [63].
[31, 50]. Bcl-2 also inhibits the oligomerization of Bax/Bak, Critically, it needs to be noted that our approach to utilize
which is associated with mitochondrial membrane permea- a whole body knockout of Lgals3−/−does not fully allow to
bilization and cell apoptosis [51, 52]. Diao et al. recently determine an impact of galectin 3 at the local testicular level.
reported that Lgals3 downregulation by siRNA in pituitary Yet, the broad expression of galectin-3 in most testicular
tumor cells leads to an increase in Bax and caspase 3 expres- cells would render it almost very difficult to deplete this
sion, and thus an elevated apoptotic rate [53]. Moreover, lectin from the male gonad using cell-specific knock outs.
increased galectin 3 expression levels serve as a poor prog- In conclusion, our data show that Lgals3−/− mice expe-
nosis marker in hepatocellular carcinoma, bladder cancer, rience a delay of the first wave of spermatogenesis. The
and pancreatic carcinoma — all related to the anti-apoptotic resulting impairment to testicular development ultimately
properties of galectin 3 [54–58]. Thus, the increased num- causes mild testicular atrophy due to an increased rate
bers of apoptotic cells in Lgals3−/− testes in this study seems of germ cell apoptosis and a decreased number of mac-
to be the result of an imbalanced anti- and pro-apoptotic rophages in adult testes. Increased expression of steroi-
protein expression due to the absence of endogenous galectin dogenic enzymes in Lgals3−/− testes, as well as functional
3, an effect leading to testis shrinkage and lower number of in vitro data indicate that galectin 3 is involved in andro-
germ cells in adulthood. gen biosynthesis. Our results thus underline an essential
Sano et al. found that galectin 3 acts as a chemoattractant role for galectin 3 in archetypical testicular functions.
for macrophages under physiological conditions [38]. In con-
trast, the galectin 3 inhibitor lactose can restrain the infiltra- Acknowledgements The authors would like to thank Prof. Ralf Mid-
dendorff and Sabine Tasch for providing the PCNA antibody and
tion of macrophages and neutrophils and induce pancreatic acknowledge Prof. Thomas Linn and Dr. Qingkui Jiang for offering the
edema in acute pancreatitis [59]. Here, we found a decreased

13
3634 T. Lei et al.

MLTC-1 cell line. We are grateful to Yahia Almousa for assistance with relevance to disorders of testis function in adulthood. Repro-
the fixation protocol and tissue collection. We appreciate the support duction 125:769–784. https​://doi.org/10.1530/rep.0.12507​69
from our research assistants Petra Moschansky, Maria Daniltchenko 12. Yang R-Y, Rabinovich GA, Liu F-T (2008) Galectins: structure,
and Gudrun Koch. function and therapeutic potential. Expert Rev Mol Med 10:e17.
https​://doi.org/10.1017/S1462​39940​80007​19
Funding The financial support of the China Scholarship Council to Tao 13. Shimura T, Takenaka Y, Tsutsumi S et al (2004) Galectin-3,
Lei and of the Medical Faculty of Justus-Liebig University is gratefully a novel binding partner of beta-catenin. Cancer Res 64:6363–
acknowledged. This work was supported by grants from the Deutsche 6367. https​://doi.org/10.1158/0008-5472.CAN-04-1816
Forschungsgemeinschaft (DFG) BL1115/2-1 and Heisenberg Program 14. Li P, Liu S, Lu M et al (2016) Hematopoietic-derived galectin-3
(BL1115/3-1-BL1115/7-1) to S.M.B. causes cellular and systemic insulin resistance. Cell 167:973-
984.e12. https​://doi.org/10.1016/j.cell.2016.10.025
15. de Oliveira FL, Gatto M, Bassi N et al (2015) Galectin-3 in
Data availability The datasets generated during and analysed during
autoimmunity and autoimmune diseases. Exp Biol Med (May-
the current study are available from the corresponding author on rea-
wood) 240:1019–1028. https​: //doi.org/10.1177/15353​7 0215​
sonable request.
59382​6
16. Deschildre C, Ji JW, Chater S et al (2007) Expression of galectin-3
Compliance with ethical standards and its regulation in the testes. Int J Androl 30:28–40. https​://doi.
org/10.1111/j.1365-2605.2006.00707​.x
Conflict of interests The author(s) declare(s) that they have no com- 17. Haddad Kashani H, Moshkdanian G, Atlasi MA et al (2013)
peting interests. Expression of galectin-3 as a testis inflammatory marker in vasec-
tomised mice. Cell J 15:11–18
Ethics approval All procedures involving animals were carried out in 18. Özbek M, Hitit M, Yıldırım N et al (2018) Expression pattern
strict accordance with guidelines for care and use of experimental ani- of galectin-1 and galectin-3 in rat testes and epididymis during
mals of the German law of welfare. Organ collection from mice was postnatal development. Acta Histochem 120:814–827. https:​ //doi.
approved by the local Animal Ethics Committee (Regierungspräsidium org/10.1016/j.acthi​s.2018.09.006
Giessen GI Nr. 646_M). 19. Jones JL, Saraswati S, Block AS et al (2010) Galectin-3 is associ-
ated with prostasomes in human semen. Glycoconj J 27:227–236.
https​://doi.org/10.1007/s1071​9-009-9262-9
20. Kim H, Ahn M, Moon C et al (2008) Immunohistochemical study
of galectin-3 in mature and immature bull testis and epididymis. J
Vet Sci 9:339–344. https​://doi.org/10.4142/jvs.2008.9.4.339
References 21. Mei S, Chen P, Lee C-L et al (2019) The role of galectin-3 in
spermatozoa-zona pellucida binding and its association with
1. Russell LD, Ettlin RA, Hikim APS, Clegg ED (1993) Histologi- fertilization in vitro. Mol Hum Reprod 25:458–470. https​://doi.
cal and histopathological evaluation of the testis. Int J Androl org/10.1093/moleh​r/gaz03​0
16:83–83. https​://doi.org/10.1111/j.1365-2605.1993.tb011​56.x 22. Blois SM, Dveksler G, Vasta GR et al (2019) Pregnancy galecti-
2. Yoshida S (2010) Stem cells in mammalian spermatogenesis. nology: insights into a complex network of glycan binding pro-
Dev Growth Differ 52:311–317. https​://doi.org/10.1111/j.1440- teins. Front Immunol 10:1–15. https​://doi.org/10.3389/fimmu​
169X.2010.01174​.x .2019.01166​
3. Cheng CY, Mruk DD (2010) The biology of spermatogenesis: 23. Freitag N, Tirado-Gonzalez I, Barrientos G et al (2020) Galectin-3
the past, present and future. Philos Trans R Soc Lond B Biol deficiency in pregnancy increases the risk of fetal growth restric-
Sci 365:1459–1463. https​://doi.org/10.1098/rstb.2010.0024 tion (FGR) via placental insufficiency. Cell Death Dis. https:​ //doi.
4. De Rooij DG, Russell LD (2000) All you wanted to know about org/10.1038/s4141​9-020-02791​-5
spermatogonia but were afraid to ask. J Androl 6512:776–798. 24. Hara A, Niwa M, Noguchi K et al (2020) Galectin-3 as a next-
https​://doi.org/10.1002/j.1939-4640.2000.tb034​08.x generation biomarker for detecting early stage of various diseases.
5. Song H-W, Wilkinson MF (2014) Transcriptional control Biomolecules 10:1–19. https​://doi.org/10.3390/biom1​00303​89
of spermatogonial maintenance and differentiation. Semin 25. Hsu DK, Yang RY, Pan Z et al (2000) Targeted disruption of
Cell Dev Biol 30:14–26. https ​ : //doi.org/10.1016/j.semcd​ the galectin-3 gene results in attenuated peritoneal inflammatory
b.2014.02.005 responses. Am J Pathol 156:1073–1083. https​://doi.org/10.1016/
6. Itman C, Mendis S, Barakat B, Loveland KL (2006) All in the S0002​-9440(10)64975​-9
family: TGF-beta family action in testis development. Reproduc- 26. Wudy SA, Schuler G, Sánchez-Guijo A, Hartmann MF (2018) The
tion 132:233–246. https​://doi.org/10.1530/rep.1.01075​ art of measuring steroids: principles and practice of current hor-
7. Sung WK, Komatsu M, Jagiello GM (1986) The kinetics of the monal steroid analysis. J Steroid Biochem Mol Biol 179:88–103.
first wave of spermatogenesis in the newborn male mouse. Gamete https​://doi.org/10.1016/j.jsbmb​.2017.09.003
Res 14:245–254. https​://doi.org/10.1002/mrd.11201​40308​ 27. Fijak M, Damm L-J, Wenzel J-P et al (2015) Influence of testos-
8. Vergouwen RP, Huiskamp R, Bas RJ et al (1993) Postnatal devel- terone on inflammatory response in testicular cells and expression
opment of testicular cell populations in mice. J Reprod Fertil of transcription factor Foxp3 in T cells. Am J Reprod Immunol
99:479–485. https​://doi.org/10.1530/jrf.0.09904​79 74:12–25. https​://doi.org/10.1111/aji.12363​
9. Bellvé AR, Cavicchia JC, Millette CF et al (1977) Spermato- 28. Niedenberger BA, Busada JT, Geyer CB (2015) Marker
genic cells of the prepuberal mouse. Isolation and morphologi- expression reveals heterogeneity of spermatogonia in the neo-
cal characterization. J Cell Biol 74:68–85 natal mouse testis. Reproduction 149:329–338. https​: //doi.
10. França LR, Hess RA, Dufour JM et al (2016) The Sertoli cell: org/10.1530/REP-14-0653
one hundred fifty years of beauty and plasticity. Andrology 29. Auharek SA, de França LR (2010) Postnatal testis develop-
4:189–212. https​://doi.org/10.1111/andr.12165​ ment, Sertoli cell proliferation and number of different sper-
11. Sharpe RM, McKinnell C, Kivlin C, Fisher JS (2003) Prolif- matogonial types in C57BL/6J mice made transiently hypo- and
eration and functional maturation of Sertoli cells, and their

13
Targeted disruption of galectin 3 in mice delays the first wave of spermatogenesis and increases… 3635

hyperthyroidic during the neonatal period. J Anat 216:577–588. 48. Jahnukainen K, Ehmcke J, Quader MA et al (2011) Testicular
https​://doi.org/10.1111/j.1469-7580.2010.01219​.x recovery after irradiation differs in prepubertal and pubertal
30. Ahmed H, AlSadek DMM (2015) Galectin-3 as a potential non-human primates, and can be enhanced by autologous germ
target to prevent cancer metastasis. Clin Med Insights Oncol cell transplantation. Hum Reprod 26:1945–1954. https​://doi.
9:113–121. https​://doi.org/10.4137/CMO.S2946​2 org/10.1093/humre​p/der16​0
31. Lichtenstein RG, Rabinovich GA (2013) Glycobiology of cell 49. Khorsandi L, Orazizadeh M (2011) Immunolocalization of
death: when glycans and lectins govern cell fate. Cell Death galectin-3 in mouse testicular tissue. Iran J Basic Med Sci
Differ 20:976–986. https​://doi.org/10.1038/cdd.2013.50 14:349–353
32. Nakahara S, Oka N, Raz A (2005) On the role of galectin-3 50. Yang RY, Hsu DK, Liu FT (1996) Expression of galectin-3
in cancer apoptosis. Apoptosis 10:267–275. https ​ : //doi. modulates T-cell growth and apoptosis. Proc Natl Acad Sci U
org/10.1007/s1049​5-005-0801-y S A 93:6737–6742. https​://doi.org/10.1073/pnas.93.13.6737
33. Nio-Kobayashi J, Boswell L, Amano M et al (2014) The loss of 51. Shamas-Din A, Kale J, Leber B, Andrews DW (2013) Mechanisms
luteal progesterone production in women is associated with a of action of Bcl-2 family proteins. Cold Spring Harb Perspect Biol
galectin switch via α2,6-sialylation of glycoconjugates. J Clin 5:a008714. https​://doi.org/10.1101/cshpe​rspec​t.a0087​14
Endocrinol Metab 99:4616–4624. https​: //doi.org/10.1210/ 52. Andreu-Fernández V, Sancho M, Genovés A et al (2017)
jc.2014-2716 Bax transmembrane domain interacts with prosurvival Bcl-2
34. Zirkin BR, Papadopoulos V (2018) Leydig cells: formation, proteins in biological membranes. Proc Natl Acad Sci USA
function, and regulation. Biol Reprod 99:101–111. https​://doi. 114:310–315. https​://doi.org/10.1073/pnas.16123​22114​
org/10.1093/biolr​e/ioy05​9 53. Diao B, Liu Y, Xu G-Z et al (2018) The role of galectin-3 in the
35. Ivell R, Anand-ivell R (2009) Biology of insulin-like factor 3 in tumorigenesis and progression of pituitary tumors. Oncol Lett
human reproduction. Hum Reprod Update 15:463–476. https​:// 15:4919–4925. https​://doi.org/10.3892/ol.2018.7931
doi.org/10.1093/humup​d/dmp01​1 54. Eisa NH, Ebrahim MA, Ragab M et al (2015) Galectin-3 and
36. Casarini L, Santi D, Brigante G, Simoni M (2018) Two hor- matrix metalloproteinase-9: perspective in management of hepa-
mones for one receptor: evolution, biochemistry, actions, and tocellular carcinoma. J Oncol Pharm Pract 21:323–330. https​://
pathophysiology of LH and hCG. Endocr Rev 39:549–592. https​ doi.org/10.1177/10781​55214​53269​8
://doi.org/10.1210/er.2018-00065​ 55. Xie L, Ni W-K, Chen X-D et al (2012) The expressions and
37. Jia W, Kidoya H, Yamakawa D et al (2013) Galectin-3 acceler- clinical significances of tissue and serum galectin-3 in pancre-
ates M2 macrophage infiltration and angiogenesis in tumors. atic carcinoma. J Cancer Res Clin Oncol 138:1035–1043. https​
Am J Pathol 182:1821–1831. https​: //doi.org/10.1016/j.ajpat​ ://doi.org/10.1007/s0043​2-012-1178-2
h.2013.01.017 56. Gendy HE, Madkour B, Abdelaty S et al (2014) Diagnostic and
38. Sano H, Hsu DK, Yu L et al (2000) Human galectin-3 is a novel prognostic significance of serum and tissue galectin 3 expres-
chemoattractant for monocytes and macrophages. J Immunol sion in patients with carcinoma of the bladder. Curr Urol 7:185–
165:2156–2164 190. https​://doi.org/10.1159/00036​5673
39. DeFalco T, Potter SJ, Williams AV et al (2015) Macrophages 57. Fang T, Liu D-D, Ning H-M et al (2018) Modified citrus pec-
contribute to the spermatogonial niche in the adult tes- tin inhibited bladder tumor growth through downregulation of
tis. Cell Rep 12:1107–1119. https​: //doi.org/10.1016/j.celre​ galectin-3. Acta Pharmacol Sin. https​://doi.org/10.1038/s4140​
p.2015.07.015 1-018-0004-z
40. Mossadegh-Keller N, Sieweke MH (2018) Testicular mac- 58. Campo VL, Marchiori MF, Rodrigues LC, Dias-Baruffi M
rophages: guardians of fertility. Cell Immunol 330:120–125. (2016) Synthetic glycoconjugates inhibitors of tumor-related
https​://doi.org/10.1016/j.celli​mm.2018.03.009 galectin-3: an update. Glycoconj J 33:853–876. https​: //doi.
41. Busada JT, Velte EK, Serra N et al (2016) Rhox13 is required org/10.1007/s1071​9-016-9721-z
for a quantitatively normal first wave of spermatogenesis in 59. Pan L-L, Deng Y-Y, Wang R et al (2018) Lactose induces phe-
mice. Reproduction 152:379–388. https​: //doi.org/10.1530/ notypic and functional changes of neutrophils and macrophages
REP-16-0268 to alleviate acute pancreatitis in mice. Front Immunol 9:751.
42. Mu X, Lee Y-F, Liu N-C et al (2004) Targeted inactivation of https​://doi.org/10.3389/fimmu​.2018.00751​
testicular nuclear orphan receptor 4 delays and disrupts late 60. Meinhardt A, Wang M, Schulz C, Bhushan S (2018) Microenvi-
meiotic prophase and subsequent meiotic divisions of spermato- ronmental signals govern the cellular identity of testicular mac-
genesis. Mol Cell Biol 24:5887–5899. https​://doi.org/10.1128/ rophages. J Leukoc Biol 104:757–766. https​://doi.org/10.1002/
MCB.24.13.5887-5899.2004 JLB.3MR03​18-086RR​
43. Holembowski L, Kramer D, Riedel D et al (2014) Tap73 is 61. Lukyanenko YO, Chen JJ, Hutson JC (2001) Production of
essential for germ cell adhesion and maturation in testis. J Cell 25-hydroxycholesterol by testicular macrophages and its
Biol 204:1173–1190. https​://doi.org/10.1083/jcb.20130​6066 effects on Leydig cells. Biol Reprod 64:790–796. https​://doi.
44. Xu L, Lu Y, Han D et al (2017) Rnf138 deficiency promotes org/10.1095/biolr​eprod​64.3.790
apoptosis of spermatogonia in juvenile male mice. Cell Death 62. Gaytan F, Bellido C, Aguilar E, van Rooijen N (1994) Require-
Dis 8:e2795. https​://doi.org/10.1038/cddis​.2017.110 ment for testicular macrophages in Leydig cell proliferation and
45. Lovasco LA, Gustafson EA, Seymour KA et al (2015) TAF4b is differentiation during prepubertal development in rats. J Reprod
required for mouse spermatogonial stem cell development. Stem Fertil 102:393–399. https​://doi.org/10.1530/jrf.0.10203​93
Cells 33:1267–1276. https​://doi.org/10.1002/stem.1914.TAF4b​ 63. Geenen V, Makara GB, Lepletier A et al (2018) Lack of galec-
46. Sansone A, Kliesch S, Isidori AM, Schlatt S (2019) AMH and tin-3 disrupts thymus homeostasis in association to increase
INSL3 in testicular and extragonadal pathophysiology: what of local and systemic glucocorticoid levels and steroidogenic
do we know? Andrology 7:131–138. https​://doi.org/10.1111/ machinery. Front Edocrinol 9:1–13. https​: //doi.org/10.3389/
andr.12597​ fendo​.2018.00365​
47. Chiarini-Garcia H, Hornick JR, Griswold MD, Russell LD
(2001) Distribution of type a spermatogonia in the mouse is not Publisher’s Note Springer Nature remains neutral with regard to
random. Biol Reprod 65:1179–1185. https​://doi.org/10.1095/ jurisdictional claims in published maps and institutional affiliations.
biolr​eprod​65.4.1179

13

You might also like