ArticlePDF Available

Human Menstrual Blood Stem Cell-Derived Granulosa Cells Participate in Ovarian Follicle Formation in a Rat Model of Premature Ovarian Failure In Vivo

Authors:

Abstract and Figures

We recently reported the application of human menstrual blood stem cells' (HuMenSCs) transplantation as a treatment modality in a rat model of premature ovarian failure (POF). We continued to investigate further in this respect. Female rats were injected intraperitoneally with 36 mg/kg busulfan. HuMenSCs were obtained, grown, and analyzed for immunophenotypic features at passage three. The cells were labeled with CM-Dil and infused into the rats. There were four groups: normal, negative control, treatment, and Sham. One month after treatment, the ovaries were collected and weighed. Histological sections were prepared from the ovary and HuMenSCs were tracking. Subsequently, we examined the changes of expression of Bax and B cell lymphoma 2 (Bcl2) genes by real-time polymerase chain reaction assay. One month after HuMenSCs transplantation, these cells were located in the ovarian interstitium and granulosa cells (GCs). The number of TUNEL-positive cells significantly decreased in the treatment group. Also the expression level of Bax genes, unlike Bcl2 gene, significantly decreased compared with negative and sham groups. In our study, HuMenSCs were tracked in ovarian tissues within 2 months after transplantation, and they differentiated into GCs. Therefore, the use of these cells can be a practical and low-cost method for the treatment of POF patients.
Content may be subject to copyright.
Human Menstrual Blood Stem Cell-Derived Granulosa Cells
Participate in Ovarian Follicle Formation in a Rat Model
of Premature Ovarian Failure In Vivo
Parastoo Noory,
1
Shadan Navid,
2
Bagher Minaee Zanganeh,
1
Ali Talebi,
3,4
Maryam Borhani-Haghighi,
1
Keykavos Gholami,
1
Marjan Dehghan Manshadi,
1
and Mehdi Abbasi
1
Abstract
We recently reported the application of human menstrual blood stem cells’ (HuMenSCs) transplantation as a
treatment modality in a rat model of premature ovarian failure (POF). We continued to investigate further in this
respect. Female rats were injected intraperitoneally with 36 mg/kg busulfan. HuMenSCs were obtained, grown,
and analyzed for immunophenotypic features at passage three. The cells were labeled with CM-Dil and infused
into the rats. There were four groups: normal, negative control, treatment, and Sham. One month after treat-
ment, the ovaries were collected and weighed. Histological sections were prepared from the ovary and Hu-
MenSCs were tracking. Subsequently, we examined the changes of expression of Bax and B cell lymphoma 2
(Bcl2) genes by real-time polymerase chain reaction assay. One month after HuMenSCs transplantation, these
cells were located in the ovarian interstitium and granulosa cells (GCs). The number of TUNEL-positive cells
significantly decreased in the treatment group. Also the expression level of Bax genes, unlike Bcl2 gene,
significantly decreased compared with negative and sham groups. In our study, HuMenSCs were tracked in
ovarian tissues within 2 months after transplantation, and they differentiated into GCs. Therefore, the use of
these cells can be a practical and low-cost method for the treatment of POF patients.
Keywords: POF, HuMenSCs, busulfan, Bax, Bcl2, ovary
Introduction
Menopause or the last menstrual cycle of women
occurs at an average age of 50.7 years. Menopause
before the age of 40 is called premature ovarian failure (POF)
(Goswami and Conway, 2007). One percent of women
under the age of 40 and 0.1% of women under the age of 30
experience POF (Meskhi and Seif, 2006). Patients with
POF have symptoms such as stopping follicular ovarian
activity, a follicle-stimulating hormone (FSH) concentra-
tiongreaterthan20to40mIU/mLinthepresenceofpri-
mary or secondary amenorrhea, hypergonadotropinemia,
and hypoestrogenemia (Falsetti et al., 1999; Kovanci and
Schutt, 2015).
Symptoms of POF patients are like physiological men-
opause, such as: infertility with palpitations, heat intoler-
ance, flushes, anxiety, depression, and fatigue. In addition
to infertility, hormone defects may cause neurological,
metabolic, or cardiovascular complications, and ultimately
lead to osteoporosis (Beck-Peccoz and Persani, 2006).
POF in each stage reduces the number of initial primor-
dial follicles, increases apoptosis or follicular degeneration,
and induces inability of follicles in response to gonado-
tropin stimulation (Welt, 2008). Several factors contribute
to the regulation of apoptosis and guarantee the survival of
preovulation follicles, including: gonadotropins, estrogen,
growth factors, cytokines, reorganization of the actin cy-
toskeleton, and nitric oxide and any changes in them can
lead to early ovarian failure. In contrast, tumor necrosis
factor-a,F
as
ligand, and androgens are stimulants of apo-
ptosis (Sinha and Kuruba, 2007).
POF can be due to several factors, such as infection,
metabolic disease, autoimmune disorders, or iatrogenic
cause such as radiation, chemotherapy, or physical damage
to the ovary (Chapman et al., 2015). One of the side effects
of chemotherapy or radiation therapy in treating patients
1
Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
2
Department of Anatomy, Gonabad University of Medical Sciences, Gonabad, Iran.
3
School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
4
Clinical Research Development Unit, Bahar Hospital, Shahroud University of Medical Sciences, Shahroud, Iran.
CELLULAR REPROGRAMMING
Volume 21, Number 5, 2019
ªMary Ann Liebert, Inc.
DOI: 10.1089/cell.2019.0020
249
Downloaded by 77.111.247.69 from www.liebertpub.com at 10/22/19. For personal use only.
with cancer is to increase the likelihood of ovarian damage
and, consequently, infertility (Schmidt et al., 2005). There are
several ways to preserve women’s fertility before treatment
with chemotherapeutic agents. The common method is hor-
monal stimulation of the ovary followed by in vitro fertiliza-
tion, and finally embryo cryopreservation. The second method
is ovarian tissue cryopreservation and its transplantation after
treatment. Nevertheless, both methods delay the treatment of
cancer, and cryopreserved tissue transplantation also has the
potential of reintroducing cancer cells (Xu et al., 2009).
One of the other methods is hormonal replacement ther-
apy (HRT). But the results of several recent studies have
indicated that HRT increases the risk of breast cancer, heart
attacks, and stroke. Therefore, it is recommended that
treatment of menopausal women with HRT be stopped
(Shelling, 2010).
In recent years, several studies have been done on the
therapeutic potential of stem cells. Mesenchymal stem cells
(MSCs) are capable of self-renewal, and have the capability
of differentiating into three germ layers and therapeutic
potential. Multipotent stem cells can now be obtained from
various sources such as adipose tissue, amniotic fluid, um-
bilical cord, bone marrow etc. (Ding et al., 2011; Lai et al.,
2015; Lv et al., 2018).
Studies with MSCs, such as bone marrow stem cells (Lee
et al., 2007), umbilical stem cells (Song et al., 2016), adi-
pose tissue stem cells (Sun et al., 2013), and amniotic fluid
stem cells (AFSCs) (Xiao et al., 2016), have shown that
these cells have the ability to restore ovarian function and
prolonged fertility in chemotherapy agents-induced labora-
tory animals. However, difficult access with invasive pro-
cedures and low proliferation capacity, has limited their
application.
Recently, endometrial-derived, highly proliferative stem
cell population has been identified in menstrual blood
(Xiang et al., 2017). Human menstrual blood stem cells
(HuMenSCs) were first extracted and described by Gargett
(2004). HuMenSCs have characteristics, such as spindle-
shaped appearance in culture, differentiation into three
germinal layers, and expression of surface markers similar
to those of the bone marrow-derived MSCs (Chen et al.,
2017). Previous studies have shown that HuMenSCs ex-
press some of the pluripotency markers, including Oct-4,
SSEA-4, nanog, c-kit, and STRO-1, as well as some of the
specific markers of MSCs, such as CD9, CD29, CD44,
CD49f, CD90, CD105, and CD117 (Lin et al., 2011; Ro-
drigues et al., 2012).
These cells are able to differentiate into chrondrogenic,
adipogenic, osteogenic, neurogenic, endothelial, pulmonary
epithelial, hepatic/pancreatic, and cardiogenic cell lineages
(Borlongan et al., 2010).
The therapeutic potential of HuMenSCs has been dem-
onstrated in several disease models, such as Duchenne
muscular dystrophy (Cui et al., 2007), stroke (Borlongan
et al., 2010), diabetes (Santamaria et al., 2011), myocardial
infarction (Zhang et al., 2013), and hepatic failure (Chen
et al., 2017). Previous studies indicated that Menstrual-
derived mesenchymal cells have a strong potential for re-
storing the function of cardiovascular disorders through
cardiomyogenesis in vitro (Hida et al., 2008).
Previous clinical studies have shown that alkylating
agents have the highest risk of infertility (Oktem and Oktay,
2007b). Cyclophosphamide, chlorambucil, melphalan, bu-
sulfan, nitrogen mustard, and procarbazine can be men-
tioned as alkylating agents (Oktem and Oktay, 2007a).
Busulfan is one of them that affects the reproductive process
of rat by its cytotoxic effect on the ovary (Sakurada et al.,
2009). In a previous study on rats treated with busulfan, it
has been reported that the number of oogonia reduces during
germ cell proliferation, resulting in a decrease in the number
of primordial follicles in the ovary (Shirota et al., 2003). In
another study, Brinster et al. (2003) reported that busulfan
targets granulosa cells (GCs) in the follicle.
According to the therapeutic properties and noninvasive
and easy access to HuMenSCs, we injected these cells
through the tail vein into chemotherapy-induced POF rat
and then measured restorative effects on ovarian function
with TUNEL assay and real-time technique. Since the death
of GCs occurs during the process of injection of busulfan as
chemotherapy agent, we decided to measure the differenti-
ation of these cells into ovarian-like cells (particularly GCs).
Materials and Methods
Experimental animals
Forty female Wistar albino rats (200–250 g, 6–8 week-old)
were purchased from Pharmacy Faculty of Tehran University
of Medical Sciences, Tehran, Iran. All procedures were ap-
proved by the Ethics Committee of Tehran University of
Medical Sciences, which corresponds to the national and
institutional guidelines for animal care and use.
Animal model establishment
and experimental grouping
To establish the POF model of chemotherapy-induced
ovarian damage, adult female rats were administered bu-
sulfan (Sigma, St. Louis, MO) at a dose of 36 mg/kg through
intraperitoneal injection. To confirm the POF model 7 days
after injection, the ovaries were collected. After Hematox-
ylin and Eosin (H&E) staining, the samples were examined
under light microscope and images were taken from the
slides for POF confirmation.
After establishing the POF model, we randomly divided
the rats into four equal groups (n=8): The control group
consisted of normal control rats that received no treatment. In
the negative control group, the rats were administered
busulfan. In the treatment group, after 7 days, POF rats were
injected intravenously with HuMenSCs (1·10
6
cells per
200 lL) in 1 mL phosphate-buffered saline (PBS; Sigma,
Steinheim, Germany). In Sham group, POF rats were injected
intravenously with 1mL PBS through the tail vein.
Isolation and culture of cells
HuMenSCs was collected from 20- to 30-year-old five
healthy women on the second day of the period with the
help of a special cup. This study was approved by the Ethics
Committee of Tehran University of Medical Sciences (ref.
no. 25110, approved 28 April 2014) and was performed
according to national and international guidelines.
HuMenSCs were isolated and centrifuged at 1500 gfor 10
minutes to obtain cellular pellets. The cellular pellets were
cultured in a T25 flask containing DMEM/F12 supple-
mented with 10% fetal bovine serum (Gibco), 100 mg/mL
250 NOORY ET AL.
Downloaded by 77.111.247.69 from www.liebertpub.com at 10/22/19. For personal use only.
streptomycin, and 100 U/mL penicillin (both from Gibco) in
a humidified incubator at 37C with 5% carbon dioxide. The
culture medium was changed every 3 days. When the cells
reached 80%–90% confluence, they were detached using
trypsin–EDTA (Gibco) for 5 minutes and passaged to cul-
ture flasks and observed under an inverted microscope
(Olympus, Tokyo, Japan) for evaluation of morphologic
features. For transplantation, the cells were prepared after 14
days of culture in passage 3 and were labeled with DiI
(Invitrogen, Carlsbad, CA).
For transplantation of these cells to the rats, vaginal
smears were obtained from rats daily. Only rats showing at
least two consecutive normal 4- to 5-day vaginal estrus
cycles were used. The injection of these cells was done in
the first cycle of estrus.
Flow cytometry for identification of HuMenSCs’
characterization and confirmation by the Homing assay
in ovary after transplantation
HuMenSCs were characterized by flow cytometry analysis
of specific surface antigen expression. The suspension cells
were incubated with FITC-conjugated monoclonal antibodies
against CD90, CD44, CD34, CD45, CD146, CD105, CD73,
CD10, and CD29 (all from eBioscience, CA), with a 1:100
dilution for 1 hour at room temperature. We used an isotype
antibody (mouse IgG1-FITC; BD Biosciences) as a negative
control for the measurement of nonspecific binding. The
expression levels of OCT4 and C-kit genes were assessed
(Abcam) followed by a 30-minute incubation with FITC-
conjugated sheep anti-rabbit antibody (Sina Biotech, Iran).
Finally, the cells were analyzed by FC500 flow cytometry
(based on the method used in our previously published arti-
cles) (Manshadi et al., 2019; Rajabi et al., 2018).
In addition, we used flow cytometry technique for homing
assessment 14 days after transplantation to the POF rat to
confirm the survival of HuMenSCs. Briefly, ovary samples
were collected and homogenized with collagenase IV (In-
vitrogen). The suspension cells were centrifuged and exposed
to lysis buffer. Samples were suspended in PBS and evaluated
for red fluorescence CM-Dil (570nm). The percentage of the
Dil cells in ovary was analyzed by flow cytometry. Our
finding was presented by FlowJo software (Navid et al., 2017;
Szilvassy et al., 2001).
Hoechst staining
One month after the cell injection, ovaries were collected,
fixed and embedded in paraffin, and cut into 5 lm sections.
The slides were then permeabilized by 0.02% Triton X-100
(Merck, Germany) and were then stained with Hoechst dye
(33258, Sigma, 25MG) to label the nuclei. The slides were
visualized with an inverted microscope (Olympus).
Ovarian follicle counts and histological analysis
One month after transplantation, the ovaries were col-
lected from all groups and the surrounding fat tissues were
removed and weighed. Ovaries were then embedded in
paraffin, serial sections (5 lm) were taken from five con-
secutive 100 lm intervals in the middle third of each ovary
and stained with H&E (Muskhelishvili et al., 2005; Picut
et al., 2008). Slides were observed using a light microscope.
The number of oocyte-containing follicles with a distinct
oocyte nucleus at each developmental stage was classified
and counted. The follicles were classified as follows: pri-
mordial follicle, primary follicle, secondary follicle, early
antral follicles, and preovulatory follicles (Myers et al.,
2004; Pedersen et al., 1968).
Tunel assay
To detect apoptosis in the cell of ovaries, TUNEL stain-
ing kits (Fluorescein Roche, 11684795910) were used on
paraffin-embedded ovaries according to the manufacturer’s
instructions. Briefly, 2 lm sections were cut and the slides
were deparaffinized and washed thrice with PBS for 5 min-
utes, immersed twice in 10% aqueous hydrogen peroxide for
10 minutes, and incubated with proteinase K at 37Cfor30
minutes. The slides were then exposed to 3% Triton X-100
for about 10 minutes at room temperature. Twenty-five mi-
croliters of terminal deoxynucleotidyl transferase (TdT) was
added to the samples, and the whole setting was incubated for
2 hours at 37C in a humidified atmosphere in the dark.
After washing thrice with PBS, the nuclei were stained
using 5 lg/mL propidium iodide (PI; Invitrogen) for a few
seconds. Apoptotic cells in the ovary were stained green.
Images were observed with fluorescence microscope
(Olympus). The percentage of TUNEL-positive cells was
determined by counting five random fields from each sam-
ple. The results are expressed as the percentages of apo-
ptotic cells in each section (Kraupp et al., 1995).
Real-time polymerase chain reaction
One month after transplantation, expression levels of ap-
optosis regulator (Bax, proapoptotic) and B cell lymphoma 2
(Bcl2, antiapoptotic) were examined by real-time polymerase
chain reaction (PCR). Total RNA was isolated from ovaries
using TRIzol reagent (Ready Mini Kit, Qiagen), according to
the manufacturer’s instruction. To ensure cDNA, 1 lgRNA
was used to prepare a single-strand cDNA using Oligo (dT)
primer (MWG-Biotech, Germany) and reverse transcription
enzyme (Fermentas), based on the protocol.
Each PCR was performed using PCR master mix and SYBR
GreenontheABI(AppliedBiosystems)StepOnemachine
(Sequences Detection Systems, Foster City, CA), according to
the manufacturer’s protocol. The ratio of expression of the genes
examined in this study was evaluated using the comparative CT
method (DDCT) and glyceraldehyde-3-phosphate dehydroge-
nase (GAPDH) used as internal control. The nucleotide se-
quences of primers are listed in Table 1 (Navid et al., 2017).
Statistical analysis
All data were expressed as mean standard deviation.
Statistical analysis of the results of all data were performed
using one-way analysis of variance (ANOVA) followed by
Tukey’s post hoc test. p£0.05 was considered statistically
significant.
Results
HuMenSCs morphology
The cells were cultured in cell culture flasks and rapidly
proliferated in vitro. In passage 0, the cells with a spindle
HUMENSC-DERIVED GRANULOSA CELLS PARTICIPATE IN PREMATURE OVARIAN FAILURE 251
Downloaded by 77.111.247.69 from www.liebertpub.com at 10/22/19. For personal use only.
fibroblast-like morphology was observed (Fig. 1a) and these
cells obtained a homologous single layer of the specific
mesenchymal colonies (passage 3) (Fig. 1b).
Immunophenotypic characterization of HuMenSCs
After three passages, the flow cytometry analysis of the
cells revealed that HuMenSCs typically express CD44,
CD90 CD34, CD146, CD105, CD73, CD10, and OCT4
(specific MSC markers) but they failed to express c-kit.
Also, the lack of expression of CD45, CD34, and CD29
cultured cells showed a nonhematopoietic stem cell origin
(based on our article published in the microscopy research
and techniques journal and Reproductive Biology journal).
Specifications of POF-induced rats
Initially, the POF model was confirmed. One week after
injection of busulfan, ovaries of POF rats and normal rats
were collected and cut and subjected to H&E staining for
pathological evaluation. According to observations, ovarian
size in the POF group was smaller than the normal group.
Microscopic examination showed that in POF-induced rats,
the ovaries were atrophied, as a result of busulfan toxicity,
and they contain fibrous tissue, follicular atresia, and a small
number of follicles with damaged oocytes in all stages of
development as a result of follicular evacuation. In contrast,
ovaries of normal rats contain a large number of follicles
with healthy oocytes in each stages of development (Fig. 2).
Tracing of HuMenSCs in the ovaries with fluorescent
microscopy and flow cytometry
HuMenSCslabeledwithDil(redfluorescence)weretrans-
planted into the busulfan-induced female rats after a week.
The rat ovary examination by fluorescence microscopy con-
firmed HuMenSC’s implantation. According to our results,
HuMenSCs labeled with Dil showed fluorescent signals
1 month after injection. These cells localized particularly into
GCs of follicle (Fig. 3a–c). Moreover, cell homing was as-
sessed by flow cytometry. Dil (red fluorescence)-positive
HuMenSCs were observed along the injection tract after 2
months in the ovaries of POF rat. So, this date indicates that
Dil-positive HuMenSCs could survive transplantation within
the POF rat ovaries for at least 2 months in vivo.However,
HuMenSCs were capable of in vivo survival after implanta-
tion (Fig. 3e).
Thus, our finding showed that HuMenSC transplantation
can play a pivotal role in the improved structure and
function of the ovaries in the rat. Also, Dil-positive Hu-
MenSCs could not be detected along the injection tract in
the ovaries of POF rat.
Ovarian weight and follicle number increased
after HuMenSC transplantation
One month after injection of HuMenSCs, H&E staining
showed an increase in the number of follicles at each stage of
development in the treatment group (Fig. 4a–c). Therefore, in-
travenous treatment significantly reduced ovarian damage.
There was no significant difference between the number of
sham group and negative control group in all stages of fol-
licular evolution, and both groups showed a significant de-
crease compared with the normal group. This decrease was
significant for primordial follicle ( p£0.0001 vs. negative con-
trol and sham group), secondary follicle ( p<0.1 vs. negative
control and sham group), but this decrease was not signifi-
cant for the primary, early antral, and preovulatory follicles.
After 1 month, the number of primordial follicles in the
treatment group (293.66 37.16) was significantly in-
creased compared with the negative group (164 4.35) and
sham group (189 26.6). The number of secondary folli-
cles in the treatment group (47.33 1.52) were increased
compared with the negative group (27.33 5.50) and the
sham group (21.33 1.52). Also, the number of primary
Table 1. The Primers Sequence Bax, Bcl2, GAPDH of Genes
Gene name Sequence Product size (bp) Annealing temp
Bcl2 For: 5¢-GCAAACTGGTGCTCAAGG-3¢183 56.63
Rev: 5¢-CAGCCACAAAGATGGTCA-3¢
Bax For: 5¢-GAGTGGGATACTGGAGATGAAG-3¢233 57.4
Rev: 5¢-TGGTAGCGACGAGAGAAGTCC-3¢
GAPDH For: 5¢-AAGTTCAACGGCACAGTCAAGG-3¢121 61.58
Rev: 5¢-CATACTCAGCACCAGCATCACC-3¢
Bcl2, B cell lymphoma 2; GAPDH, glyceraldehyde-3-phosphate dehydrogenase.
FIG. 1. Morphology characteris-
tics of HuMenSCs. (a) Spindle-
shaped HuMenSCs sticking to the
flask floor in early stages of
growth: passage 0 (b) Colony-
forming HuMenSCs with multi-
faceted appearance after 2 weeks:
passage3 (inverted microscope,
scale bar =200 lm). HuMenSCs,
human menstrual blood stem cells.
252 NOORY ET AL.
Downloaded by 77.111.247.69 from www.liebertpub.com at 10/22/19. For personal use only.
follicles (182.66 29.95), early antral follicles (23 5.29),
and preovulatory follicles (8.332.8) in the treatment
group increased compared with the negative groups, but
this increase was not significant (Fig. 4e).
To examine the effect of HuMenSCs on ovarian function,
ovarian weight changes were evaluated in all four groups.
There were no mortality due to the transplantation. One
month after injection of HuMenSCs, the ovaries of all four
groups were collected and weighed. The weight of the
ovaries in the negative control and sham group showed a
significant difference relative to the positive control and
treatment group. Ovarian weight increased in the treatment
group and was close to the normal (positive control) group.
The weight of ovaries in the treatment group (41.4 6.95),
compared with the negative control group (20.88 5.14),
indicated a significant increase ( p£0.001) and also showed
a significant increase ( p£0.01) compared with the sham
group (25.09 4.66) (Fig. 4f ).
The morphological changes in the ovaries have shown that
Dil-positive HuMenSCs cells could increase a higher number
of follicles at each stage of development in the ovaries in the
treatment group compared with the negative group (POF). In
addition, ovarian weight changes in the treatment group in
comparison with the negative group confirmed this result.
TUNEL assay
Since the alkylating agents (busulfan) induce ovarian
degeneration through GC’s apoptosis, we evaluated the
protective effects of HuMenSCs against apoptosis in ovaries
FIG. 2. Microscopic morphology
of ovarian after induction of POF
(H&E staining, scale bar =200
lm). (a) Follicular atresia and de-
pletion, 7 days after intraperitoneal
injection of busulfan. (b) Ovaries
of normal rats contain a large
number of follicles with healthy
oocytes in each stages of develop-
ment. H&E, Hematoxylin and Eo-
sin; POF, premature ovarian
failure.
FIG. 3. In vivo homing of CM-Dil-labeled HuMenSCs in busulfan-injured ovarian. (a) Nuclei were labeled with Hoechst.
(b) CM-Dil-labeled HuMenSCs. (c) Merged. HuMenSCs were traced in ovarian, 1 month after injection using CM-Dil-
labeling. These cells were located in the ovarian interstitium and GCs (arrow) (Fluorescent microscope, scale bar =200 lm).
(d) Cell homing was assessed by flow cytometry for red fluorescence CM-Dil (570 nm) 2 months after transplantation
in vivo. GCs, granulosa cells.
HUMENSC-DERIVED GRANULOSA CELLS PARTICIPATE IN PREMATURE OVARIAN FAILURE 253
Downloaded by 77.111.247.69 from www.liebertpub.com at 10/22/19. For personal use only.
of POF rat model. One month after the HuMenSC’s trans-
plantation, the TUNEL staining was performed and then the
number of TUNEL-positive cells was calculated in each
sections. There was no significant difference between the
number of TUNEL-positive cells in the sham group and the
negative group, and both groups showed a significant in-
crease ( p<0.0001) in comparison with the normal group
(Fig. 5). The mean number of TUNEL-positive cells in the
treatment group (23.58% 1.12%) was significantly lower
than the negative control group (46.67% 0.89%) and sham
group (45.49% 0.83%) ( p<0.0001).
Real-time PCR analysis
To further investigate the changes in apoptosis after bu-
sulfan induction, 1 month after injection of HuMenSCs, the
changes in the expression of Bax and Bcl2 genes involved
in apoptosis were investigated by quantitative real-time
PCR. As shown in Figure 6, in the negative control and
sham groups, the expression of Bax ( proapoptosis) and Bcl2
(antiapoptotic) genes was significantly ( p<0.0001) differ-
ent from that of the normal group, indicating an increase in
the level of apoptosis in the negative control group after
busulfan induction. Also, there was no significant difference
in the expression of these genes in the sham group compared
with the negative control group, which indicates that the
ovarian function is not improved in the sham group and the
negative group without cell therapy in these rats.
The graph indicates that the level of expression of these
genes in the negative group and sham group is at one level,
and not seen any difference in terms of reducing or in-
creasing of them. In the treatment groups, the level of ex-
pression of Bax gene was significantly decreased
(p<0.0001) compared with the negative group and sham
group. However, the expression of Bcl2 gene increased in
the treatment group in comparison with the negative group
and sham group, but this increase was not significant. These
changes indicate that the reduction in apoptosis after cell
therapy was mostly through a reduction in the expression of
Bax genes in the treatment group (Fig. 6).
Discussion
Previous study clearly demonstrated that HuMenSCs can
play an important role in the treatment of POF rat ( Man-
shadi et al., 2019), where HuMenSCs can improve and re-
store ovarian function and reduce apoptosis in damaged
ovarian tissue caused by busulfan toxicity. We also found
that there was the presence of effective homing of infused
cells into the ovary is crucial in cell-based therapies.
Today, the use of MSCs in the treatment of a wide range
of diseases has been studied. These cells are multipotent,
capable of self-renewal and high proliferative potential, and
differentiate into mesodermic and nonmesodermic lineages
(Emmerson and Gargett, 2016). MSCs are nonhematopoietic,
stromal cells, which also have the capacity to differentiate
into various types of osteocytic, chondrocytic, and adipocytic
lineages (De Cesaris et al., 2017).
In addition to bone marrow, other fibroblast-like stem
cells are found in other tissues including, circulating blood,
cord blood, placenta, amniotic fluid, heart, skeletal muscle,
adipose tissue, synovial tissue, and pancreas. In other words,
FIG. 4. (a–d) Ovarian sections of rats stained with H&E, 1 month after transplantation. H&E staining shows the follicular
atresia and evacuation in the negative control (a) and sham (c) groups, which indicates that recovery was not achieved after
1 month and 1 week after the injection of busulfan. While in the treatment group (d) follicular atresia has been improved
1 month after transplantation and follicular reserves are preserved. Positive group (b) (scale bars =200 lm). (e, f ) Com-
parison of ovarian weight and the number of follicles in four groups 1 month after injection. Data are expressed as
mean SD (****p£0.0001). ns; SD, standard deviation.
254 NOORY ET AL.
Downloaded by 77.111.247.69 from www.liebertpub.com at 10/22/19. For personal use only.
it can be hypothesized that all organs containing connective
tissue also contain MSCs (Kalervo Va
¨a
¨na
¨nen, 2005). Due to
the power of differentiation of MSCs into different tissues,
there is interest in using them to replace damaged tissues.
Given modern advances in gene therapy and tissue engi-
neering, they can be useful in improving the quality of life in
the future (Oreffo et al., 2005). These cells contribute to the
improvement of many diseases by the secretion of paracrine
agents, exosomes, secretomes, or even mitochondria (Bianco,
2014).
Previous studies have shown the therapeutic potential of
some MSCs on the ovary of laboratory animals in POF
model, including adipose tissue stem cells (Sun et al., 2013),
AFSCs (Ding et al., 2017; Pan et al., 2017; Xiao et al., 2016),
umbilical stem cells (Li et al., 2017; Song et al., 2016), and
bone marrow stem cells (Fu et al., 2008; Lee et al., 2007). All
of these have proven the improvement of ovarian function
after transplantation of cells in the POF model. Also, our
study showed improvement of some of the factors involved in
ovarian function after transplantation of mesenchyma-like
FIG. 5. TUNEL staining in ovary tissue sections after 1 month. TUNEL-positive cells labeled light, and nuclei labeled
dark (PI). (a–c) normal group, (d–f) negative control group, (g–i) sham group, and (j–l) treatment group. TUNEL-positive
cells are further restricted to proliferating granulosa and theca cells and lead to early depletion of ovarian follicles. TUNEL-
positive cells decreased in treatment group 1 month after transplantation of HuMenSCs (fluorescence microscope, scale
bar =10 lm). (m) There was no significant difference between the number of positive TUNELcells in the sham group and
the negative group. Apoptosis decreased in the treatment group, and showed a significant difference compared with three
groups: normal, negative control, and sham (***p<0.0001).
FIG. 6. Changes in the expression of Bax and Bcl2 genes in 4 groups 1 month after transplantation. In the negative control
and sham groups, the expression of Bax and Bcl2 genes was significantly different from that of the normal group. There was
no significant difference in the expression of these genes in the sham group compared with the negative control group. The
expression of Bax genes was significantly decreased, compared with the negative group and sham group and there was no
significant difference in Bax expression between treatment and normal groups (****p<0.0001). Bcl2, B cell lymphoma 2.
HUMENSC-DERIVED GRANULOSA CELLS PARTICIPATE IN PREMATURE OVARIAN FAILURE 255
Downloaded by 77.111.247.69 from www.liebertpub.com at 10/22/19. For personal use only.
stem cells extracted from menstrual blood on rat POF model.
Moreover, It has been accepted that effective homing of in-
fused cells into the ovary is crucial in cell-based therapies.
Immunohistochemistry studies by Meirow et al. demon-
strated positive staining of apoptosis in pre GCs in patients
treated with cisplatin (Meirow et al., 1998). Chemotherapy
agents lead to apoptosis by stimulating p53 activity and its
downstream genes such as Bim and Casp9 (Happo et al.,
2010; Xiao et al., 2016). The present study proved the de-
generative changes busulfan induced in the ovarian tissue of
POF rat model as in previous studies (Lai et al., 2015; Tan
et al., 2010). Like previous studies, a significant reduction in
ovarian weight was also observed in POF (Chen et al., 2015;
Ding et al., 2017; Liu et al., 2014; Wang et al., 2017).
In 2008, Patel and Silva stated that menstrual blood stem
cells express the embryonic and multipotent markers, such
as Oct-4, SSEA-4, and c-kit, and markers of mesenchymal
cells, including CD90, CD166, and CD105, but, they do not
indicate the expression of hematopoietic cell markers, in-
cluding CD31 (endothelial), CD34 (hematopoietic and en-
dothelial stem cells), and CD45 (leukocyte) (Patel and Silva,
2008). The main advantage of these cells is easy, periodic,
and noninvasive access as compared with other MSCs.
Previous studies have shown that HuMenSCs have a low
level of immunogenic reactions and tumor formation (Azedi
et al., 2017; Borlongan et al., 2010). Our results indicate that
HuMenSCs express more than 85% of the specific mesen-
chymal markers, such as CD90 and CD44.
The therapeutic potential of HuMenSCs has been proven
in several disease models, such as Duchenne muscular
dystrophy (Cui et al., 2007), stroke (Borlongan et al., 2010;
Rodrigues et al., 2012), type 1 diabetes (Santamaria et al.,
2011; Wu et al., 2014), hepatic failure (Chen et al., 2017),
acute lung injury (Xiang et al., 2017), and myocardial in-
farction (Zhang et al., 2013). In the present study, we in-
vestigated the capacity of self-renewal and therapeutic
potentialofHuMenSCsontheovariantissueinPOF
model. Stem cells isolated from human menstrual blood
had fibroblast-like properties, adhesion to the flask, and high
proliferative potential, as demonstrated in previous studies
(Lai et al., 2015; Liu et al., 2014; Wang et al., 2017).
In 2017, Wang et al. (2017) showed that HuMenSCs la-
beled with GFP showed fluorescent signals in the interstitial
tissue 7 days after injection, but the fluorescence signal was
not observed 21 days after injection. HuMenSCs labeled
with DiO (green fluorescent) were observed 14 days after
injection in the ovary in POF mice (Liu et al., 2014).
Similarly, Dongmei Lai et al. examined the tracing of
GFP-positive cells by using live imaging, 6 hours to 14 days
after the HuMenSC injection in the POF model mice. They
observed that these cells first entered the pelvic organs 6 to 12
hours after the injection, and entered the chest 24 hours after
the injection and then, and observed weak signals 7 days after
injection in the pelvic organs. Immunofluorescence studies
after infusion of GFP-stained cells, confirmed the presence of
these cells 2 months after injection into the ovarian stromal
tissue (Lai et al., 2015).
Our findings indicate that the presence of DiI-labeled
HuMenSCs (red fluorescence) could be detected in the
ovary a month after the injection by using a fluorescence
microscope and 2 months after the HuMenSCs injection by
flow cytometry in the POF rat model. On the other hand, we
confirmed the presence of HuMenSCs at least 2 months after
transplantation into the ovaries of rat POF in vivo.
Also, the implantation assessment showed that these cells
are located in addition to interstitium of the ovary in GCs.
Thus, it can be said that these cells are likely to be differ-
entiated into GCs and, keeping in mind the process of re-
generation, evolution, and secretion of inhibitory hormones,
maintains FSH level low and prevents follicular evacuation
(based on the article accepted in the microscopy research
and techniques journal).
One month after injection of HuMenSCs, the weight of
the ovaries increased significantly, indicating implantation
and protective effect of these cells on ovarian tissue. Pre-
vious studies on HuMenSCs (Liu et al., 2014; Wang et al.,
2017), hAMSCs (human amniotic stem cells) (Ding et al.,
2017), have also noted this increase.
Our study shows a significant decrease in follicles at all
stages of development after POF induction compared with
the normal group. but, in the treatment group, the number of
these follicles increased in all stages compared with the
sham and negative groups and this increase was significant
for primordial and secondary follicles. Nonimprovement in
the sham group indicates that the ovarian function is not
improving spontaneously and is due to the therapeutic and
regenerative potential of these cells after migration to the
ovarian tissue.
Previous studies show an increase in the number of folli-
cles, 8 weeks and 21 days, respectively, after intravenous
injection of HuMenSCs into POF mice model (Lai et al.,
2015; Wang et al., 2017). This increase has also been re-
ported in other MSCs, including AFSCs (Ding et al., 2017),
umbilical MSCs (Li et al., 2017; Song et al., 2016), and bone
marrow MSCs (BMMSCs) (Fu et al., 2008) for in situ in-
jection. Sun et al. (2013) showed that adipose tissue stem
cells, both intravenous injection and in situ injection, increase
the number of follicles in all stages, 1 month after injection.
Perez et al. (1997) showed that chemotherapeutic agent-
exposed germ cells begin apoptosis due to the activation of
several death signaling pathways, including ceramide, Bax,
and caspase. Previous studies have identified the effect of
different alkylating agents on increased levels of GCs apo-
ptosis (Chen et al., 2015; Sun et al., 2013; Wang et al., 2017;
Yang et al., 2012). Our study also confirmed an increase
in apoptosis after induction of busulfan. However, after
transplantation of intravenous menstrual stem cells, apo-
ptosis significantly decreased. Also, in the negative control
and sham groups, the number of apoptotic cells in one level
was observed, indicating that the improvement in the
treatment group in the course of 1 month is caused by the
injection of stem cells and their therapeutic potential.
Guan-Yu Xiao et al. stated that exosomes derived from
AFSCs, through micro-RNAs (in which both miR-146a and
miR 10a are highly enriched) and their potential target genes
(involved in apoptosis), indicate antiapoptotic effects on
damaged GCs and also prevention of follicular atresia, es-
pecially the primordial follicles, in 72 hours after induction
of POF on mice (Xiao et al., 2016). Zhen Wang studies
showed that HuMenSCs and HuMenSC-derived conditioned
media (CM) exerted a protective effect and antiapoptotic
role on damaged ovaries through FGF2 secretion, which
also reduce the fibrosis in the ovarian interstitium and in-
crease follicle growth (Wang et al., 2017). Three recent
256 NOORY ET AL.
Downloaded by 77.111.247.69 from www.liebertpub.com at 10/22/19. For personal use only.
studies have described the mechanisms of action of MSCs,
especially HuMenSCs, as well as their effect on ovarian
tissue and their protective effect.
We also examined the expression of Bax and Bcl2 genes
for further studies. Our study showed increased expression
of Bax and decreased expression of Bcl2, thereby increasing
the level of apoptosis in the POF model, as in the previous
study (Chen et al., 2015). Findings of Fu et al. regarding the
cytokines secretion from bone marrow stem cells in the
culture medium and the increased level of expression of
Bcl2 in vivo, showed that the reduction of apoptosis in GCs
after BMMSC transplantation in POF model, could be due
to upstream adjustment of Bcl2 genes (Fu et al., 2008). In
the present study, the reduction of the BAX gene expression
level significantly suggests that HuMenSCs can, by para-
crine factors, induce upregulation of Bax and Bcl2 genes
and antiapoptotic effects on GCs.
Conclusions
Our further research strongly proves the effectiveness of
stem cells in the treatment, regeneration, and improvement of
the function of busulfan-induced POF. Given the easy and
noninvasive access of these cells, they can be applied as a
new and effective approach for the treatment of POF patients.
Acknowledgment
The authors thank the staff of Tehran University of
Medical Sciences for animal care.
Ethics Approval and Consent to Participate
Animal experiments were approved by the Ethics Com-
mittee of Tehran University of Medical Sciences and all
procedures were performed in accordance with the univer-
sity’s guidelines. In the present work, we used animal model
considering all the rights based on the Ethics Committee of
Medical Faculty of Tehran University.
Author Disclosure Statement
The authors declare they have no conflicting financial
interests.
Funding Information
The authors are grateful to Tehran University of Medical
Sciences for their funding support.
References
Azedi, F., Kazemnejad, S., Zarnani, A.H., Soleimani, S., Sho-
jaei, A., and Arasteh, S. (2017). Comparative capability of
menstrual blood versus bone marrow derived stem cells in
neural differentiation. Mol. Biol. Rep. 44, 169–182.
Beck-Peccoz, P., and Persani, L. (2006). Premature ovarian
failure. Orphanet J. Rare Dis. 1, 9.
Bianco, P. (2014). ‘‘Mesenchymal’ stem cells. Ann. Rev. Cell
Dev. Biol. 30, 677–704.
Borlongan, C.V., Kaneko, Y., Maki, M., Yu, S.-J., Ali, M.,
Allickson, J.G., Sanberg, C.D., Kuzmin-Nichols, N., and
Sanberg, P.R. (2010). Menstrual blood cells display stem
cell–like phenotypic markers and exert neuroprotection fol-
lowing transplantation in experimental stroke. Stem Cells.
Dev. 19, 439–452.
Brinster, C.J., Ryu, B.-Y., Avarbock, M.R., Karagenc, L.,
Brinster, R.L., and Orwig, K.E. (2003). Restoration of fertility
by germ cell transplantation requires effective recipient
preparation. Biol. Reprod. 69, 412–420.
Chapman, C., Cree, L., and Shelling, A.N. (2015). The genetics of
premature ovarian failure: Currentperspectives.Int.J.Womens
Health 7, 799.
Chen, L., Xiang, B., Wang, X., and Xiang, C. (2017a). Exosomes
derived from human menstrual blood-derived stem cells alle-
viate fulminant hepatic failure. Stem Cell Res. Ther. 8, 9.
Chen, L., Zhang, C., Chen, L., Wang, X., Xiang, B., Wu, X.,
Guo, Y., Mou, X., Yuan, L., and Chen, B. (2017b). Human
menstrual blood-derived stem cells ameliorate liver fibrosis in
mice by targeting hepatic stellate cells via paracrine mediators.
Stem Cells Transl. Med. 6, 272–284.
Chen, W., Xu, X., Wang, L., Bai, G., and Xiang, W. (2015).
Low expression of Mfn2 is associated with mitochondrial
damage and apoptosis of ovarian tissues in the premature
ovarian failure model. PLoS One 10, e0136421.
Cui, C.-H., Uyama, T., Miyado, K., Terai, M., Kyo, S., Kiyono,
T., and Umezawa, A. (2007). Menstrual blood-derived cells
confer human dystrophin expression in the murine model of
Duchenne muscular dystrophy via cell fusion and myogenic
transdifferentiation. Mol. Biol. Cell 18, 1586–1594.
De Cesaris, V., Grolli, S., Bresciani, C., Conti, V., Basini, G.,
Parmigiani, E., and Bigliardi, E. (2017). Isolation, prolifera-
tion and characterization of endometrial canine stem cells.
Reprod. Domest. Anim. 52, 235–242.
Ding, C., Li, H., Wang, Y., Wang, F., Wu, H., Chen, R., Lv, J.,
Wang, W., and Huang, B. (2017). Different therapeutic ef-
fects of cells derived from human amniotic membrane on
premature ovarian aging depend on distinct cellular biological
characteristics. Stem Cell Res. Ther. 8, 173.
Ding, D.-C., Shyu, W.-C., and Lin, S.-Z. (2011). Mesenchymal
stem cells. Cell Transplant. 20, 5–14.
Emmerson, S.J., and Gargett, C.E. (2016). Endometrial mes-
enchymal stem cells as a cell based therapy for pelvic organ
prolapse. World J Stem Cells 8, 202.
Falsetti, L., Scalchi, S., Villani, M.T., and Bugari, G. (1999).
Premature ovarian failure. Gynecol. Endocrinol. 13, 189–195.
Fu, X.-F., He Y, Xie, C., and Liu, W. (2008). Bone marrow
mesenchymal stem cell transplantation improves ovarian
function and structure in rats with chemotherapy-induced
ovarian damage. Cytotherapy 10, 353–363.
Gargett, C.E. (2004). Stem cells in gynaecology. Aust. N. Z. J.
Obstet. Gynaecol. 44, 380–386.
Goswami, D., and Conway, G.S. (2007). Premature ovarian
failure. Horm. Res. Paediatr. 68, 196–202.
Happo, L., Cragg, M.S., Phipson, B., Haga, J.M., Jansen, E.S.,
Herold, M.J., Dewson, G., Michalak, E.M., Vandenberg, C.J.,
and Smyth, G.K. (2010). Maximal killing of lymphoma cells
by DNA damage–inducing therapy requires not only the p53
targets Puma and Noxa, but also Bim. Blood 116, 5256–5267.
Hida, N., Nishiyama, N., Miyoshi, S., Kira, S., Segawa, K.,
Uyama, T., Mori, T., Miyado, K., Ikegami, Y., and Cui,
C.H. (2008). Novel cardiac precursor-like cells from hu-
man menstrual blood-derived mesenchymal cells. Stem
Cells 26, 1695–1704.
Kalervo Va
¨a
¨na
¨nen, H. (2005). Mesenchymal stem cells. Ann.
Med. 37, 469–479.
HUMENSC-DERIVED GRANULOSA CELLS PARTICIPATE IN PREMATURE OVARIAN FAILURE 257
Downloaded by 77.111.247.69 from www.liebertpub.com at 10/22/19. For personal use only.
Kovanci, E., and Schutt, A.K. (2015). Premature ovarian fail-
ure. Obstet. Gynecol. Clin. 42, 153–161.
Kraupp, B.G., Ruttkay-Nedecky, B., Koudelka, H., Bukowska,
K., Bursch, W., and Schulte-Hermann, R. (1995). In situ
detection of fragmented DNA (TUNEL assay) fails to dis-
criminate among apoptosis, necrosis, and autolytic cell death:
A cautionary note. Hepatology 21, 1465–1468.
Lai, D., Wang, F., Yao, X., Zhang, Q., Wu, X., and Xiang, C.
(2015). Human endometrial mesenchymal stem cells restore
ovarian function through improving the renewal of germline
stem cells in a mouse model of premature ovarian failure. J.
Transl. Med. 13, 155.
Lee, H.-J., Selesniemi, K., Niikura, Y., Niikura, T., Klein, R.,
Dombkowski, D.M., and Tilly, J.L. (2007). Bone marrow
transplantation generates immature oocytes and rescues
long-term fertility in a preclinical mouse model of chemotherapy-
induced premature ovarian failure. J. Clin. Oncol. 25, 3198–3204.
Li, J., Mao, Q.X., He, J.J., She, H.Q., Zhang, Z., and Yin, C.Y.
(2017). Human umbilical cord mesenchymal stem cells im-
prove the reserve function of perimenopausal ovary via a
paracrine mechanism. Stem Cell Res. Ther. 8, 55.
Lin, J., Xiang, D., Zhang, J., Allickson, J., and Xiang, C. (2011).
Plasticity of human menstrual blood stem cells derived from
the endometrium. J. Zhejiang Univ. Sci. B 12, 372–380.
Liu, T., Huang, Y., Zhang, J., Qin, W., Chi, H., Chen, J., Yu, Z.,
and Chen, C. (2014). Transplantation of human menstrual
blood stem cells to treat premature ovarian failure in mouse
model. Stem Cells Dev. 23, 1548–1557.
Lv, H., Hu, Y., Cui, Z., and Jia, H. (2018). Human menstrual
blood: A renewable and sustainable source of stem cells for
regenerative medicine. Stem Cell Res. Ther. 9, 325.
Manshadi, M.D., Navid, S., Hoshino, Y., Daneshi, E., Noory,
P., and Abbasi, M. (2019). The effects of human menstrual
blood stem cells-derived granulosa cells on ovarian follicle
formation in a rat model of premature ovarian failure. Mi-
crosc. Res. Tech. 82, 635–642.
Meirow, D., Nugent, D., Epstein, M., Livni, N., and Gosden, R.G.
(1998). An in-vitro study of the effects of chemotherapy on human
primordial follicles. In Human Reproduction, 13–14. (Oxford
Univ Press Great Clarendon St, Oxford OX2 6DP, England).
Meskhi, A., and Seif, M.W. (2006). Premature ovarian failure.
Curr. Opin. Obstet. Gynecol. 18, 418–426.
Muskhelishvili, L., Wingard, S.K., and Latendresse, J.R.
(2005). Proliferating cell nuclear antigen—A marker for
ovarian follicle counts. Toxicol. Pathol. 33, 365–368.
Myers, M., Britt, K.L., Wreford, N.G., Ebling, F.J., and Kerr,
J.B. (2004). Methods for quantifying follicular numbers
within the mouse ovary. Reproduction 127, 569–580.
Navid, S., Abbasi, M., and Hoshino, Y. (2017a). The effects of
melatonin on colonization of neonate spermatogonial mouse
stem cells in a three-dimensional soft agar culture system.
Stem Cell Res. Ther. 8, 233.
Navid, S., Rastegar, T., Baazm, M., Alizadeh, R., Talebi, A.,
Gholami, K., Khosravi-Farsani, S., Koruji, M., and Abbasi,
M. (2017b). In vitro effects of melatonin on colonization of
neonate mouse spermatogonial stem cells. Syst. Biol. Reprod.
Med. 63, 370–381.
Oktem, O., and Oktay, K. (2007a). A novel ovarian xeno-
grafting model to characterize the impact of chemotherapy
agents on human primordial follicle reserve. Cancer Res. 67,
10159–10162.
Oktem, O., and Oktay, K. (2007b). Quantitative assessment of
the impact of chemotherapy on ovarian follicle reserve and
stromal function. Cancer 110, 2222–2229.
Oreffo, R.O.C., Cooper, C., Mason, C., and Clements, M.
(2005). Mesenchymal stem cells. Stem Cell Rev. 1, 169–178.
Pan, Y., Zhang, L., Zhang, X., Hu, C., and Liu, R. (2017).
Biological and biomechanical analysis of two types of mes-
enchymal stem cells for intervention in chemotherapy-induced
ovarian dysfunction. Arch. Gynecol. Obstet. 295, 247–252.
Patel, A.N., and Silva, F. (2008). Menstrual blood stromal cells:
The potential for regenerative medicine. Regen. Med. 3, 443–
444.
Pedersen, T., and Peters, H. (1968). Proposal for a classification
of oocytes and follicles in the mouse ovary. J. Reprod. Fertil.
17, 555–557.
Perez, G.I., Knudson, C.M., Leykin, L., Korsmeyer, S.J., and
Tilly, J.L. (1997). Apoptosis-associated signaling pathways
are required for chemotherapy-mediated female germ cell
destruction. Nat. Med. 3, 1228–1232.
Picut, C.A., Swanson, C.L., Scully, K.L., Roseman, V.C., Parker,
R.F., and Remick, A.K. (2008). Ovarian follicle counts using
proliferating cell nuclear antigen (PCNA) and semi-automated
image analysis in rats. Toxicol. Pathol. 36, 674–679.
Rajabi, Z., Yazdekhasti, H., Mugahi, S.M.H.N., Abbasi, M.,
Kazemnejad, S., Shirazi, A., Majidi, M., and Zarnani, A.-H.
(2018). Mouse preantral follicle growth in 3D co-culture
system using human menstrual blood mesenchymal stem cell.
Reprod. Biol. 18, 122–131.
Rodrigues, M.C.O., Voltarelli, J., Sanberg, P.R., Allickson,
J.G., Kuzmin-Nichols, N., Garbuzova-Davis, S., and Bor-
longan, C.V. (2012). Recent progress in cell therapy for basal
ganglia disorders with emphasis on menstrual blood trans-
plantation in stroke. Neurosci. Biobehav. Rev. 36, 177–190.
Sakurada, Y., Kudo, S., Iwasaki, S., Miyata, Y., Nishi, M., and
Masumoto, Y. (2009). Collaborative work on evaluation of
ovarian toxicity 5) Two-or four-week repeated-dose studies
and fertility study of busulfan in female rats. J. Toxicol. Sci.
34, SP65–SP72.
Santamaria, X., Massasa, E.E., Feng, Y., Wolff, E., and Taylor,
H.S. (2011). Derivation of insulin producing cells from hu-
man endometrial stromal stem cells and use in the treatment
of murine diabetes. Mol. Ther. 19, 2065–2071.
Schmidt, K.L.T., Andersen, C.Y., Loft, A., Byskov, A.G., Ernst,
E., and Andersen, A.N. (2005). Follow-up of ovarian function
post-chemotherapy following ovarian cryopreservation and
transplantation. Hum. Reprod. 20, 3539–3546.
Shelling, A.N. (2010). Premature ovarian failure. Reproduction
140, 633–641.
Shirota, M., Soda, S., Katoh, C., Asai, S., Sato, M., Ohta, R.,
Watanabe, G., Taya, K., and Shirota, K. (2003). Effects of
reduction of the number of primordial follicles on follicular
development to achieve puberty in female rats. Reproduction
125, 85–94.
Sinha, P., and Kuruba, N. (2007). Premature ovarian failure. J.
Obstet. Gynaecol. 27, 16–19.
Song, D., Zhong, Y., Qian, C., Zou, Q., Ou, J., Shi, Y., Gao, L.,
Wang, G., Liu, Z., and Li, H. (2016). Human umbilical cord
mesenchymal stem cells therapy in cyclophosphamide-
induced premature ovarian failure rat model. Biomed. Res.
Int. 2016, 2517514.
Sun, M., Wang, S., Li, Y., Yu, L., Gu, F., Wang, C., and Yao,
Y. (2013). Adipose-derived stem cells improved mouse ovary
function after chemotherapy-induced ovary failure. Stem Cell
Res. Ther. 4, 80.
Szilvassy, S.J., Meyerrose, T.E., Ragland, P.L., and Grimes, B.
(2001). Differential homing and engraftment properties of
hematopoietic progenitor cells from murine bone marrow,
258 NOORY ET AL.
Downloaded by 77.111.247.69 from www.liebertpub.com at 10/22/19. For personal use only.
mobilized peripheral blood, and fetal liver. Blood 98, 2108–
2115.
Tan, S.-J., Yeh, Y.-C., Shang, W.-J., Wu, G.-J., Liu, J.-Y., and
Chen, C.-H. (2010). Protective effect of a gonadotropin-
releasing hormone analogue on chemotherapeutic agent-
induced ovarian gonadotoxicity: A mouse model. Eur. J.
Obstet. Gynecol. Reprod. Biol. 149, 182–185.
Wang,Z.,Wang,Y.,Yang,T.,Li,J.,andYang,X.(2017).
Study of the reparative effects of menstrual-derived stem
cells on premature ovarian failure in mice. Stem Cell Res.
Ther. 8, 11.
Welt, C.K. (2008). Primary ovarian insufficiency: A more ac-
curate term for premature ovarian failure. Clin. Endocrinol.
68, 499–509.
Wu, X., Luo, Y., Chen, J., Pan, R., Xiang, B., Du, X., Xiang,
L., Shao, J., and Xiang, C. (2014). Transplantation of
human menstrual blood progenitor cells improves hyper-
glycemia by promoting endogenous progenitor differenti-
ation in type 1 diabetic mice. Stem Cells Dev. 23, 1245–
1257.
Xiang, B., Chen, L., Wang, X., Zhao, Y., Wang, Y., and Xiang,
C. (2017). Transplantation of menstrual blood-derived mes-
enchymal stem cells promotes the repair of LPS-induced
acute lung injury. Int. J. Mol. Sci. 18, 689.
Xiao, G.-Y., Cheng, C.-C., Chiang, Y.-S., Cheng, W.T.-K., Liu,
I-H., and Wu, S.-H. (2016). Exosomal miR-10a derived from
amniotic fluid stem cells preserves ovarian follicles after
chemotherapy. Sci. Rep. 6, 23120.
Xu, M., Barrett, S.E., West-Farrell, E., Kondapalli, L.A., Kie-
sewetter, S.E., Shea, L.D., and Woodruff, T.K. (2009).
In vitro grown human ovarian follicles from cancer patients
support oocyte growth. Hum. Reprod. 24, 2531–2540.
Yang, X., Zhou, Y., Peng, S., Wu, L., Lin H-Y, Wang, S., and
Wang, H. (2012). Differentially expressed plasma micro-
RNAs in premature ovarian failure patients and the potential
regulatory function of mir-23a in granulosa cell apoptosis.
Reproduction 144, 235–244.
Zhang, Z., Wang, J.-A., Xu, Y., Jiang, Z., Wu, R., Wang, L.,
Chen, P., Hu, X., and Yu, H. (2013). Menstrual blood derived
mesenchymal cells ameliorate cardiac fibrosis via inhibition
of endothelial to mesenchymal transition in myocardial in-
farction. Int. J. Cardiol. 168, 1711–1714.
Address correspondence to:
Mehdi Abbasi
Department of Anatomy
School of Medicine
Tehran University of Medical Sciences
Tehran 0098
Iran
E-mail: abbasima@tums.ac.ir
HUMENSC-DERIVED GRANULOSA CELLS PARTICIPATE IN PREMATURE OVARIAN FAILURE 259
Downloaded by 77.111.247.69 from www.liebertpub.com at 10/22/19. For personal use only.
... Men-MSCs and their derivatives have been used in various preclinical and clinical studies for illnesses like diabetes mellitus ( 37 , 38 ), multiple sclerosis ( 31 ), Asherman's syndrome ( 18 ), liver failure (39)(40)(41), myocardial infarction ( 42 , 43 ), Alzheimer's disease ( 44 ) and . In animal studies, transplantation of these cells has shown improvement in ovarian dysfunction and endometrial diseases (46)(47)(48)(49). These cells effectively reduced granulosa cell apoptosis and ovarian interstitial fibrosis, resulting in increased follicular numbers and normalization of sex hormone levels ( 48 , 50 ). ...
... One month after transplantation, the cells were discovered in the ovarian interstitium and granulosa cells. Their research concluded that these cells could be a viable and low-cost method for treating POF ( 47 ). Wang Z, et al. performed another study in which Men-MSCs were transplanted into POF models of mice through the tail vein. ...
Article
Full-text available
Background: Premature ovarian failure (POF) is a well-known cause of infertility, particularly in women under the age of 40. POF is also associated with elevated gonadotropin levels, amenorrhea and sex-hormone deficiency. Aim of the study: In this study, the therapeutic potential of autologous mesenchymal stromal cells obtained from menstrual blood (Men-MSCs) for patients with POF was evaluated. Methods: 15 POF patients were included in the study. The cultured Men-MSCs were confirmed by flow cytometry, karyotype, endotoxin and mycoplasma and were then injected into the patients' right ovary by vaginal ultrasound guidance and under general anesthesia and aseptic conditions. Changes in patients' anti-Müllerian hormone (AMH), antral follicle count (AFC), follicle-stimulating hormone (FSH), luteal hormone (LH), and estradiol (E2) levels, as well as general flushing and vaginal dryness were followed up to one year after treatment. Results: All patients were satisfied with a decrease in general flushing and vaginal dryness. 4 patients (2.9%) showed a spontaneous return of menstruation without additional pharmacological treatment. There was a significant difference in AFC (0 vs. 1 ± 0.92 count, p value ≤0.001%), FSH (74 ± 22.9 vs. 54.8 ± 17.5 mIU/mL, p-value ≤0.05%), E2 (10.2 ± 6 vs. 21.8 ± 11.5 pg/mL p-value ≤0.01%), LH (74 ± 22.9 vs. 54.8 ± 17.5 IU/L,p-value ≤0.01%) during 3 months post-injection. However, there were no significant changes in AMH (p-value ≥0.05%). There were also no significant differences in assessed parameters between 3 and 6 months after cell injection. Conclusion: According to the findings of this study, administration of Men-MSCs improved ovarian function and menstrual restoration in some POF patients.
... hMensSCs increased the ovarian weight, plasma E2 levels, and follicle numbers in mice [100]. Amniotic fluid stem cells can differentiate into granulosa cells, which inhibit follicular atresia and maintain healthy follicles [101]. ...
Article
Full-text available
Infertility is a condition defined by the failure to establish a clinical pregnancy after 12 months of regular, unprotected sexual intercourse or due to an impairment of a person’s capacity to reproduce either as an individual or with their partner. The authors have set out to succinctly investigate, explore, and assess infertility treatments, harnessing the potential of stem cells to effectively and safely treat infertility; in addition, this paper will present the legal and regulatory complexities at the heart of stem cell research, with an overview of the legislative state of affairs in six major European countries. For couples who cannot benefit from assisted reproductive technologies (ART) to treat their infertility, stem-cells-based approaches have been shown to be a highly promising approach. Nonetheless, lingering ethical and immunological uncertainties require more conclusive findings and data before such treatment avenues can become mainstream and be applied on a large scale. The isolation of human embryonic stem cells (ESCs) is ethically controversial, since their collection involves the destruction of human embryonic tissue. Overall, stem cell research has resulted in important new breakthroughs in the treatment of infertility. The effort to untangle the complex web of ethical and legal issues associated with such therapeutic approaches will have to rely on evidence-based, broadly shared standards, guidelines, and best practices to make sure that the procreative rights of patients can be effectively reconciled with the core values at the heart of medical ethics.
... The anti-apoptotic effects of MSCs in ovarian cells have been related to the production of certain factors (.e.g, VEGF, HGF, epidermal growth factor (EGF), and IGF-1), the secretion of exosomal miRNAs, the regulation of apoptosis-related genes, including Bcl-2, Bax, Caspase3, as well as the downregulation of ROS production [9,121,[124][125][126]. ...
Article
Full-text available
Premature ovarian insufficiency (POI) has far-reaching consequences on women's life quality. Due to the lack of full recognition of the etiology and complexity of this disease, there is no appropriate treatment for infected patients. Recently, stem cell therapy has attracted the attention of regenerative medicine scholars and offered promising outcomes for POI patients. Several kinds of stem cells, such as embryonic stem cells (ESCs), mesenchymal stem cells (MSCs), and induced pluripotent stem cells (iPSCs) have been used for the treatment of ovarian diseases. However, their potential protective mechanisms are still unknown. Undoubtedly, a better understanding of the therapeutic molecular and cellular mechanisms of stem cells will address uncover strategies to increase their clinical application for multiple disorders such as POI. This paper describes a detailed account of the potential properties of different types of stem cells and provides a comprehensive review of their protective mechanisms, particularly MSC, in POI disorder. In addition, ongoing challenges and several strategies to improve the efficacy of MSC in clinical use are addressed. Therefore, this review will provide proof-of-concept for further clinical application of stem cells in POI.
... Thanks to their multipotent differentiation abilities and paracrine properties, multipotent MSCs have established themselves as prime candidates for optimal treatment strategies in the field of regenerative medicine. Numerous studies have investigated MSCs from various sources such as human embryonic stem cells [93,94], ovarian cells [95], um- bilical cord [96][97][98][99][100][101][102][103][104][105][106], placenta [70,107,108], fetal liver [109], amnion [110][111][112][113][114][115][116][117], chorionic lamina [118], menstrual blood (endometrium) [119][120][121][122][123][124] and bone marrow [125][126][127][128]. MSCs derived from human umbilical cord blood in particular have been used most frequently in animal models. MSCs improve ovarian activity and combat premature ovarian insufficiency primarily through paracrine mechanisms [108]. ...
Article
Full-text available
Despite advancements in assisted reproductive technology (ART), achieving successful pregnancy rates remains challenging. Diminished ovarian reserve and premature ovarian insufficiency hinder IVF success—about 20% of in vitro fertilization (IVF) patients face a poor prognosis due to a low response, leading to higher cancellations and reduced birth rates. In an attempt to address the issue of premature ovarian insufficiency (POI), we conducted systematic PubMed and Web of Science research, using keywords “stem cells”, “extracellular vesicles”, “premature ovarian insufficiency”, “diminished ovarian reserve” and “exosomes”. Amid the complex ovarian dynamics and challenges like POI, stem cell therapy and particularly the use of extracellular vesicles (EVs), a great potential is shown. EVs trigger paracrine mechanisms via microRNAs and bioactive molecules, suppressing apoptosis, stimulating angiogenesis and activating latent regenerative potential. Key microRNAs influence estrogen secretion, proliferation and apoptosis resistance. Extracellular vesicles present a lot of possibilities for treating infertility, and understanding their molecular mechanisms is crucial for maximizing EVs’ therapeutic potential in addressing ovarian disorders and promoting reproductive health.
... Primers were designed by allele ID software. The sequences of the primers are shown in Table 1 [33]. ...
Article
Full-text available
Background In recent decades, in vitro fertilization (IVF) has been widely used as a method of assisted reproductive technology (ART) to improve fertility in individuals. To be more successful in this laboratory method, we used the presence of two common types of antioxidants (melatonin and vitamin C) simultaneously and exclusively in IVF medium. Methods The cumulus-oocyte complexes (COCs) were obtained from Gonadotropin-releasing hormone (GnRH) and Human Chorionic Gonadotropin (HMG) -stimulated mice. Subsequently, metaphase II (MII) oocytes were fertilized in vitro. In the experiment, the IVF medium was randomly divided into two equal groups: The control group did not receive any antioxidants. In the treatment group, 100 µM melatonin and 5 mM vitamin C were added to the IVF medium. Finally, oocytes and putative embryos transferred into developmental medium and cultured 120 h after IVF to the blastocyst stage. After and before IVF, oocytes and putative embryos were stained with dichlorodihydrofluorescein diacetate (DCFDA) and the H2O2 level was measured with an inverted fluorescence microscope using ImageJ software. At the end of the fifth day after IVF, the expression of Bax and B cell lymphoma 2 (Bcl2) was evaluated using real-time PCR. Results The levels of reactive oxygen species (ROS) in oocytes and putative embryos observed in the treatment group demonstrated a significant reduce compared to the control group (p ≤ 0.01. (.Furthermore, the number of embryos in the blastocycte stage(P < 0.05), the expression level of the Bcl2 (P < 0.05) gene, the Bax unlike gene, significantly increased compared with the control group. Conclusion We conclude that the presence of melatonin and vitamin C antioxidants simultaneously and exclusively in the IVF medium leads to a reduction in ROS and ,as a result, improves the growth of the embryo up to the blastocyst stage.
... They constitute a stem cell population that gathers both endometrial stromal fibroblasts and perivascular eMSCs, anticipating a similar identity to eMSCs obtained from endometrial biopsies (38, 95, 96). Preclinical studies investigating human Men-MSCs transplantation in rodent models of POI shed light into novel mechanisms of ovarian function restoration, such as renewal of OSC pool (93), amelioration of fibrosis via downregulation of TGF-β1/SMAD 2,4 pathway (97) and secretion of fibroblast growth factor 2 (FGF-2) (98), upregulation of extracellular matrix (ECM)-dependent focal adhesion kinase (FAK)/Akt signaling pathway (99), and most importantly, their potential to differentiate into granulosa cells (100,101) which is unique among MSCs. In conjunction with decreasing granulosa cell apoptosis, the latter underpins the restoration of normal follicle development (99) and hormonal function, which is observed not only in preclinical studies (97,99,100,102), but also in the only, up-to-now, conducted clinical study in POI (94). ...
Article
Full-text available
Premature Ovarian Insufficiency (POI) is a multi-factorial disorder that affects women of reproductive age. The condition is characterized by the loss of ovarian function before the age of 40 years and several factors have been identified to be implicated in its pathogenesis. Remarkably though, at least 50% of women have remaining follicles in their ovaries after the development of ovarian insufficiency. Population data show that approximately up to 3.7% of women worldwide suffer from POI and subsequent infertility. Currently, the treatment of POI-related infertility involves oocyte donation. However, many women with POI desire to conceive with their own ova. Therefore, experimental biological therapies, such as Platelet-Rich Plasma (PRP), Exosomes (exos) therapy, In vitro Activation (IVA), Stem Cell therapy, MicroRNAs and Mitochondrial Targeting Therapies are experimental treatment strategies that focus on activating oogenesis and folliculogenesis, by upregulating natural biochemical pathways (neo-folliculogenesis) and improving ovarian microenvironment. This mini-review aims at identifying the main advantages of these approaches and exploring whether they can underpin existing assisted reproductive technologies.
... The homing of transplanted MSCs to the ovary in POI animal models, which affects the successful transplantation of MSCs, has been mediated by different mechanisms and molecules [20][21][22][23]. Some studies have reported that human MSC-derived menstrual blood and skin were differentiated into GCs and ovarian stroma cells in an animal model of POI [8,13,24,25]. However, most researchers ...
Article
Full-text available
Background Primary ovarian insufficiency (POI) refers to the loss of ovarian function under the age of 40 and results in amenorrhea and infertility. Our previous studies have shown that transplantation of mesenchymal stem cells (MSCs) and MSC-derived exosomes in chemotherapy-induced POI mouse ovaries can reverse the POI and eventually achieve pregnancy. Based on our recent studies, MSC-derived exosomes have almost equal therapeutic potentials as transplanted MSCs. However, it is still unclear whether exosomes can completely replace MSCs in POI treatment. For the reliable application of cell-free treatment for POI patients using exosomes, there is a need to understand whether there is any outcome and effectiveness difference between MSC and MSC-derived exosome treatment. Methods Comparing the therapeutic effect of intravenous injection using MSCs and equal amounts of exosomes in a POI mouse model will reveal the difference between the two therapeutic resources. In this study, we induced POI in C57/BL6 mice by chemotherapy (CXT) using a standard protocol. We then injected four different doses of MSCs or equal amounts of commercialized MSC-derived exosomes by retro-orbital injection post-CXT. Result After MSC/exosome treatment, tissue and serum samples were harvested to analyze molecular changes after treatment, while other mice in parallel experiments underwent breeding experiments to compare the restoration of fertility. Both the MSC- and exosome-treated groups had a restored estrous cycle and serum hormone levels compared to untreated POI mice. The pregnancy rate in the MSC-treated group was 60–100% after treatment, while the pregnancy rate in the exosome-treated group was 30–50% after treatment. Interestingly, in terms of long-term effects, MSC-treated mice still showed a 60–80% pregnancy rate in the second round of breeding, while the exosome-treated group became infertile again in the second round of breeding. Conclusions Although there were some differences in the efficacy between MSC treatment and exosome treatment, both treatments were able to achieve pregnancy in the POI mouse model. In conclusion, we report that MSC-derived exosomes are a promising therapeutic option to restore ovarian function in POI conditions similar to treatment with MSCs.
... A randomized clinical trial from South Valley University proposed a hypothesis by using filgrastim to recover ovary functions, with 10 participants from 16 to 40 years (NCT02783937). Filgrastim is a granulocyte-colony stimulating factor (G-CSF) and has been approved to stimulate peripheral blood stem cell numbers (262) and potentially improve ovarian follicle formation (263). In ovarian failure, there are still some residual very small embryoniclike stem cells (VSELs) that serve as a backup pool for mature stem cells and are mobilized under stress conditions, which might be involved in gonadal rescue after exogenous/endogenous stimuli (264,265). ...
Article
Full-text available
Premature ovarian failure (POF) is an insidious cause of female infertility and a devastating condition for women. POF also has a strong familial and heterogeneous genetic background. Management of POF is complicated by the variable etiology and presentation, which are generally characterized by abnormal hormone levels, gene instability and ovarian dysgenesis. To date, abnormal regulation associated with POF has been found in a small number of genes, including autosomal and sex chromosomal genes in folliculogenesis, granulosa cells, and oocytes. Due to the complex genomic contributions, ascertaining the exact causative mechanisms has been challenging in POF, and many pathogenic genomic characteristics have yet to be elucidated. However, emerging research has provided new insights into genomic variation in POF as well as novel etiological factors, pathogenic mechanisms and therapeutic intervention approaches. Meanwhile, scattered studies of transcriptional regulation revealed that ovarian cell function also depends on specific biomarker gene expression, which can influence protein activities, thus causing POF. In this review, we summarized the latest research and issues related to the genomic basis for POF and focused on insights gained from their biological effects and pathogenic mechanisms in POF. The present integrated studies of genomic variants, gene expression and related protein abnormalities were structured to establish the role of etiological genes associated with POF. In addition, we describe the design of some ongoing clinical trials that may suggest safe, feasible and effective approaches to improve the diagnosis and therapy of POF, such as Filgrastim, goserelin, resveratrol, natural plant antitoxin, Kuntai capsule et al. Understanding the candidate genomic characteristics in POF is beneficial for the early diagnosis of POF and provides appropriate methods for prevention and drug treatment. Additional efforts to clarify the POF genetic background are necessary and are beneficial for researchers and clinicians regarding genetic counseling and clinical practice. Taken together, recent genomic explorations have shown great potential to elucidate POF management in women and are stepping from the bench to the bedside.
... MSC surface markers (CD29, CD44, CD49f, CD90, CD105 and CD117) and ESC markers (Oct4 and SSEA3/4) were highly expressed on the HuMenSC surface [57]. It confirmed the differentiation of HuMenSCs into ovarian-like cells (especially GCs) by injecting HuMenSCs into CTXinduced POF rats through the tail vein [58]. However, the source of HuMenSCs in menstrual blood is limited, and there is a risk of infection. ...
Article
Full-text available
Incidence of premature ovarian failure (POF) is higher with the increase of the pace of life. The etiology of POF is very complex, which is closely related to genes, immune diseases, drugs, surgery, and psychological factors. Ideal animal models and evaluation indexes are essential for drug development and mechanism research. In our review, we firstly summarize the modeling methods of different POF animal models and compare their advantages and disadvantages. Recently, stem cells are widely studied for tumor treatment and tissue repair with low immunogenicity, high homing ability, high ability to divide and self-renew. Hence, we secondly reviewed recently published data on transplantation of stem cells in the POF animal model and analyzed the possible mechanism of their function. With the further insights of immunological and gene therapy, the combination of stem cells with other therapies should be actively explored to promote the treatment of POF in the future. Our article may provide guidance and insight for POF animal model selection and new drug development. Graphical Abstract
Article
Full-text available
One of the major problems in today's world is the inability to have children. This has affected social health and led researchers to preclinical studies. Over several decades, numerous animal models in the biology of reproduction have entered the field of research. Different mechanisms and approaches induce different types of reproductive diseases. Since premature ovarian failure, azoospermia and polycystic ovary syndrome are important causes of inability of couples to have children, we decided to review the studies conducted in this field, appropriate laboratory methods and processes to create an animal model for premature failure. Introduce ovarian, azoospermia and polycystic ovary syndrome. In the present study, common methods along with the relevant protocols, along with the advantages and disadvantages of each model are presented.
Article
Full-text available
Abstract Stem cells (SCs) play an important role in autologous and even allogenic applications. Menstrual blood discharge has been identified as a valuable source of SCs which are referred to as menstrual blood-derived stem cells (MenSCs). Compared to SCs from bone marrow and adipose tissues, MenSCs come from body discharge and obtaining them is non-invasive to the body, they are easy to collect, and there are no ethical concerns. There is, hence, a growing interest in the functions of MenSCs and their potential applications in regenerative medicine. This review presents recent progress in research into MenSCs and their potential application. Clinical indications of using MenSCs for various regenerative medicine applications are emphasized, and future research is recommended to accelerate clinical applications of MenSCs.
Article
Full-text available
Background Melatonin is a pleiotropic hormone with powerful antioxidant activity both in vivo and in vitro. The present study aimed to investigate the effects of melatonin on the proliferation efficiency of neonatal mouse spermatogonial stem cells (SSCs) using a three-dimensional soft agar culture system (SACS) which has the capacity to induce development of SSCs similar to in vivo conditions. Methods SSCs were isolated from testes of neonate mice and their purities were assessed by flow cytometry using PLZF antibody. Isolated testicular cells were cultured in the upper layer of the SACS in αMEM medium in the absence or presence of melatonin extract for 4 weeks. Results The identity of colonies was confirmed by alkaline phosphatase staining and immunocytochemistry using PLZF and α6 integrin antibodies. The number and diameter of colonies of SSCs in the upper layer were evaluated at days 14 and 28 of culture. The number and diameter of colonies of SSCs were significantly higher in the melatonin group compared with the control group. The levels of expression of ID-4 and Plzf, unlike c-kit, were significantly higher in the melatonin group than in the control group. Conclusions Results of the present study show that supplementation of the culture medium (SACS) with 100 μM melatonin significantly decreased reactive oxygen species (ROS) production in the treated group compared with the control group, and increased SSC proliferation.
Article
Full-text available
We have recently reported that antioxidant supplements enhance the efficacy of cryopreserved spermatogonial stem cells. Melatonin is considered a free radical scavenger which has direct and indirect antioxidant effects in in vitro and in vivo microenvironments. Due to the anti-apoptotic properties of melatonin, researchers have proposed that melatonin may improve the efficiency of spermatogonial stem cell (SSC) transplantation. However, the appropriate methodology which facilitates SSC proliferation remains to be determined. Identification of a proper propagation system is essential for the future application of SSCs in the field of infertility. The aim of the present study was to investigate the effects of melatonin on the colonization of SSCs. SSCs were isolated from the testes of three to six day old mice, and their purities were assessed by cytometry using Plzf antibody. Isolated testicular cells were cultured in the absence or presence of melatonin extract for two weeks. Suppression of differentiation and maintenance of spermatogonial stem cells was confirmed by alkaline phosphatase staining and immunocytochemistry using Plzf antibody. The number and diameter of the colonies of SSCs were assessed during the 7(th) and 14(th) days of culture, and the expression of Id4, Plzf, and C-kit were evaluated using real-time PCR at the end of the culture period. The survival rate of the cultured cells in the presence of melatonin was significantly higher than the control group. The number and diameter of colonies also increased in the cells treated with melatonin. The results of our study suggest that culture of SSCs with 100 μM melatonin supplementation can increase SSCs proliferation significantly.
Article
Full-text available
Background Many reports have shown that various kinds of stem cells have the ability to recover premature ovarian aging (POA) function. Transplantation of human amniotic epithelial cells (hAECs) improves ovarian function damaged by chemotherapy in a mice model. Understanding of how to evaluate the distinct effects of adult stem cells in curing POA and how to choose stem cells in clinical application is lacking. Methods To build a different degrees of POA model, mice were administered different doses of cyclophosphamide: light dose (70 mg/kg, 2 weeks), medium dose (70 mg/kg, 1 week; 120 mg/kg, 1 week), and high dose (120 mg/kg, 2 weeks). Enzyme-linked immunosorbent assay detected serum levels of sex hormones, and hematoxylin and eosin staining allowed follicle counting and showed the ovarian tissue structure. DiIC18(5)-DS was employed to label human amniotic mesenchymal stem cells (hAMSCs) and hAECs for detecting the cellular retention time in ovaries by a live imaging system. Proliferation of human ovarian granule cells (ki67, AMH, FSHR, FOXL2, and CYP19A1) and immunological rejection of human peripheral blood mononuclear cells (CD4, CD11b, CD19, and CD56) were measured by flow cytometry (fluorescence-activated cell sorting (FACS)). Distinction of cellular biological characteristics between hAECs and hAMSCs was evaluated, such as collagen secretory level (collagen I, II, III, IV, and VI), telomerase activity, pluripotent markers tested by western blot, expression level of immune molecules (HLA-ABC and HLA-DR) analyzed by FACS, and cytokines (growth factors, chemotactic factors, apoptosis factors, and inflammatory factors) measured by a protein antibody array methodology. Results After hAMSCs and hAECs were transplanted into a different degrees of POA model, hAMSCs exerted better therapeutic activity on mouse ovarian function in the high-dose administration group, promoting the proliferation rate of ovarian granular cells from premature ovarian failure patients, but also provoking immune rejection. Meanwhile, our results showed that the biological characteristics of hAMSCs were superior to hAECs, but not to expression of immune molecules. Conclusions These results suggest that hAMSCs are a more effective cell type to improve ovarian function than hAECs. Meanwhile, this distinct effect is attributable to cellular biological characteristics of hAMSCs (telomerase activity, expression level of pluripotent markers, cytokine and collagen secretion) that are superior to hAECs, except for immunological rejection. Sufficient consideration of cell properties is warranted to move forward to more effective clinical therapy. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0613-3) contains supplementary material, which is available to authorized users.
Article
Full-text available
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are associated with high morbidity and mortality. Menstrual blood-derived stem cells (MenSCs) have been shown to be good therapeutic tools in diseases such as ovarian failure and cardiac fibrosis. However, relevant studies of MenSCs in ALI have not yet proceeded. We hypothesized that MenSC could attenuate the inflammation in lipopolysaccharide (LPS)-induced ALI and promote the repair of damaged lung. ALI model was induced by LPS in C57 mice, and saline or MenSCs were administered via tail vein after four hours. The MenSCs were subsequently detected in the lungs by a live imaging system. The MenSCs not only improved pulmonary microvascular permeability and attenuated histopathological damage, but also mediated the downregulation of IL-1β and the upregulation of IL-10 in bronchoalveolar lavage fluid (BALF) and the damaged lung. Immunohistochemistry revealed the increased expression of proliferating cell nuclear antigen (PCNA) and the reduced expression of caspase-3 indicating the beneficial effect of MenSCs. Keratinocyte growth factor (KGF) was also upregulated after MenSCs administrated. As shown using transwell co-culture, the MenSCs also could improve the viability of BEAS-2B cells and inhibit LPS-induced apoptosis. These findings suggest that MenSC-based therapies could be promising strategies for treating ALI.
Article
Full-text available
Background Human umbilical cord mesenchymal stem cells (hUCMSCs) are a type of pluripotent stem cell which are isolated from the umbilical cord of newborns. hUCMSCs have great therapeutic potential. We designed this experimental study in order to investigate whether the transplantation of hUCMSCs can improve the ovarian reserve function of perimenopausal rats and delay ovarian senescence. Method We selected naturally aging rats confirmed by vaginal smears as models of perimenopausal rats, divided into the control group and the treatment group, and selected young fertile female rats as normal controls. hUCMSCs were transplanted into rats of the treatment group through tail veins. Enzyme-linked immunosorbent assay (ELISA) detected serum levels of sex hormones, H&E staining showed ovarian tissue structure and allowed follicle counting, immunohistochemistry and western blot analysis revealed ovarian expression of hepatocyte growth factor (HGF), vascular endothelial cell growth factor (VEGF), and insulin-like growth factor-1 (IGF-1), polymerase chain reaction (PCR) and western blot analysis revealed hUCMSCs expression of HGF, VEGF, and IGF-1. ResultsAt time points of 14, 21, and 28 days after hUCMSCs transplantation, estradiol (E2) and anti-Müllerian hormone (AMH) increased while follicle-stimulating hormone (FSH) decreased; ovarian structure improved and follicle number increased; ovarian expression of HGF, VEGF, and IGF-1 protein elevated significantly. Meanwhile, PCR and western blot analysis indicated hUCMSCs have the capacity of secreting HGF, VEGF, and IGF-1 cytokines. Conclusions Our results suggest that hUCMSCs can promote ovarian expression of HGF, VEGF, and IGF-1 through secreting those cytokines, resulting in improving ovarian reserve function and withstanding ovarian senescence.
Article
Full-text available
Background Human menstrual blood-derived stem cells (MenSCs) are a novel source of MSCs that provide the advantage of being easy to collect and isolate. Exosomes contain some mRNAs and adhesion molecules that can potentially impact cellular and animal physiology. This study aimed to investigate the therapeutic potential of MenSC-derived exosomes (MenSC-Ex) on AML12 cells (in vitro) and D-GalN/LPS-induced FHF mice (in vivo). Methods Transmission electron microscopy and Western blot were used to identify MenSC-Ex. Antibody array was used to examine cytokine levels on MenSC-Ex. MenSC-Ex were treated in D-GalN/LPS-induced AML12 in vitro. Cell proliferation and apoptosis were measured. MenSC-Ex were injected into the tail veins of mice 24 h before treatment with D-GalN/LPS. Blood and liver tissues served as physiological and biochemical indexes. The number of liver mononuclear cells (MNCs) and the amount of the active apoptotic protein caspase-3 were determined to elaborate the mechanism of hepatoprotective activity. ResultsHuman menstrual blood-derived stem cell-derived exosomes (MenSC-Ex) are bi-lipid membrane vesicles that have a round, ball-like shape with a diameter of approximately 30–100 nm. Cytokine arrays have shown that MenSC-Ex expressed cytokines, including ICAM-1, angiopoietin-2, Axl, angiogenin, IGFBP-6, osteoprotegerin, IL-6, and IL-8. MenSC-Ex markedly improved liver function, enhanced survival rates, and inhibited liver cell apoptosis at 6 h after transplantation. MenSC-Ex migrated to sites of injury and to AML12 cells (a mouse hepatocyte cell line), respectively. Moreover, MenSC-Ex reduced the number of liver mononuclear cells (MNCs) and the amount of the active apoptotic protein caspase-3 in injured livers. Conclusions In conclusion, our results provide preliminary evidence for the anti-apoptotic capacity of MenSC-Ex in FHF and suggest that MenSC-Ex may be an alternative therapeutic approach to treat FHF.
Article
Many studies have reported that human endometrial mesenchymal stem cells (HuMenSCs) are capable of repairing damaged tissues. The aim of the present study was to investigate the effects of HuMenSCs transplantation as a treatment modality in premature ovarian failure (POF) associated with chemotherapy‐induced ovarian damage. HuMenSCs were isolated from menstrual blood samples of five women. After the in vitro culture of HuMenSCs, purity of the cells was assessed by cytometry using CD44, CD90, CD34, and CD45 FITC conjugate antibody. Twenty‐four female Wistar rats were randomly divided into four groups: negative control, positive control, sham, and treatment groups. The rat models of POF used in our study were established by injecting busulfan intraperitoneally into the rats during the first estrus cycle. HuMenSCs were transplanted by injection via the tail vein into the POF‐induced rats. Four weeks after POF induction, ovaries were collected and the levels of Amh, Fst, and Fshr expression in the granulosa cell (GC) layer, as well as plasma estradiol (E2) and progesterone (P4) levels were evaluated. Moreover, migration and localization of DiI‐labeled HuMenSCs were detected, and the labeled cells were found to be localized in GCs layer of immature follicles. In addition to DiI‐labelled HuMenSCs tracking, increased levels of expression of Amh and Fshr and Fst, and the high plasma levels of E2 and P4 confirmed that HuMenSC transplantation had a significant effect on follicle formation and ovulation in the treatment group compared with the negative control (POF) group. Our findings demonstrated that HuMenSCs have improved restorative effects on ovarian function. ELISA and real time PCR techniques were used to evaluate the various parameters in our study.
Article
Follicle culture provides a condition which can help investigators to evaluate various aspects of ovarian follicle growth and development and impact of different components and supplementations as well as presumably application of follicle culture approach in fertility preservation procedures. Mesenchymal Stem Cells (MSCs), particularly those isolated from menstrual blood has the potential to be used as a tool for improvement of fertility. In the current study, a 3D co-culture system with mice preantral follicles and human Menstrual Blood Mesenchymal Stem Cells (MenSCs) using either collagen or alginate beads was designed to investigate whether this system allows better preantral follicles growth and development. Results showed that MenSCs increase the indices of follicular growth including survival rate, diameter, and antrum formation as well as the rate of in vitro maturation (IVM) in both collagen and alginates beads. Although statistically not significant, alginate was found to be superior in terms of supporting survival rate and antrum formation. Hormone assay demonstrated that the amount of secreted 17 β-estradiol and progesterone in both 3D systems increased dramatically after 12 days, with the highest levels in system employing MenSCs. Data also demonstrated that relative expression of studied genes increased for Bmp15 and Gdf9 and decreased for Mater when follicles were cultured in the presence of MenSCs. Collectively, results of the present study showed that MenSCs could improve indices of follicular growth and maturation in vitro. Further studies are needed before a clinical application of MenSCs-induced IVM is considered.