Next Article in Journal
Muscle-Bone Crosstalk and Metabolic Dysregulation in Children and Young People Affected with Type 1 Diabetes: Mechanisms and Clinical Implications
Previous Article in Journal
The Emerging Role of Peroxisome Proliferator-Activated Receptors in Cancer Stemness
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Estrogen Receptor Regulates Male Satellite Cells in a Female, but Not Male, Environment

1
Lillehei Heart Institute and Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
2
Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
3
Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
*
Author to whom correspondence should be addressed.
Cells 2025, 14(20), 1606; https://doi.org/10.3390/cells14201606
Submission received: 23 August 2025 / Revised: 11 October 2025 / Accepted: 13 October 2025 / Published: 16 October 2025

Abstract

Skeletal muscle homeostasis is dependent on the satellite cell pool, which is regulated by numerous signaling pathways. Estradiol (E2) function via estrogen receptor alpha (ERα, Esr1) plays an important role in satellite cell regulation in females, being necessary for satellite cell maintenance, proliferation and differentiation. Here we investigate this signaling axis in male satellite cells. Male satellite cells express Esr1 mRNA at similar levels to female satellite cells, and E2 enhances the proliferation of male satellite cell-derived myoblasts in vitro. Deletion of Esr1 specifically in male satellite cells has no effect on satellite cell number, nor on their ability to self-renew after injury, during regeneration, or when transplanted into male hosts. However, Esr1 deletion severely reduces self-renewal of male satellite cells when transplanted into female hosts. These data suggest that male satellite cells are competent for E2-ERα signaling, but that this signaling is not efficacious in the male environment, though E2-ERα signaling does become necessary when the male cells are transplanted into a female environment.

1. Introduction

Skeletal muscle maintenance and regeneration is dependent on satellite cells [1,2], which express the transcription factor Pax7 [3] and reside between the basal lamina and the sarcolemma of the skeletal muscle myofiber [4]. In the unperturbed condition, satellite cells are deeply quiescent, but upon stimulation (muscle stress or damage), the satellite cells activate and enter the cell cycle, yielding daughter cells that contribute nuclei to muscle fibers or repopulate the satellite cell pool [5,6].
The satellite cell pool is regulated by a complex set of factors in their microenvironment [7,8], in addition to several intrinsic and systemic extrinsic factors that determine the balance between self-renewal vs. myogenic commitment, depth of quiescence, and overall pool size [9,10,11,12,13,14]. Disruption of this harmonic balance and/or signaling leads to loss of the skeletal muscle mass and improper tissue repair in case of damage [15,16,17,18].
Among the best studied systemic signals that change with age are the sex hormones [19,20]. 17β-Estradiol (E2), the predominant and most potent form of estrogen, has a well-described role in skeletal muscle maintenance in females [21]. Consequent to estrogen deficiency in women with ovarian failure, skeletal muscle strength is deleteriously affected [22,23], which has been reversed by hormonal treatment [24,25]. In addition, a significant reduction in the appendicular skeletal muscle mass has been reported in young postmenopausal women [26] with several studies having reported improvement in skeletal muscle mass and composition with hormonal replacement therapy, e.g., in a post-menopausal monozygotic twin controlled study [27,28] and in postmenopausal females [25,29,30]. Furthermore, estrogen-based steroids have long been used as an anabolic agent in the livestock industry [31].
We have previously shown that satellite cells of female mice express Esr1, the primary estrogen receptor, and that the homeostatic satellite cell pool size drops 30–70% when mice are subjected to ovariectomy (OVX) or when Esr1 is conditionally deleted in Pax7-expressing cells of female mice [32,33,34]. Looking at homeostatic satellite cell pool size in humans revealed a positive correlation between PAX7+ cell frequency and E2 levels in perimenopausal women [32]. In mice, the reduction in the satellite cell pool occurs within two weeks of removal of ovarian hormones by OVX, is correlated with apoptosis within the pool [33], and is reversed with E2 treatment [32,34]. Furthermore, we documented a deficient regenerative response to muscle injury in female mice with OVX [32] or Esr1 KO satellite cells [34]. Thus, both the major female sex hormone, E2, and the primary receptor, ERα, are needed in satellite cells for homeostatic maintenance of the satellite cell pool, and for optimal muscle regeneration in females.
A significant reduction in satellite cell frequency with aging is seen in some muscles [35,36,37,38,39], most strongly the tibialis anterior (TA) and other locomotory muscles, which are also under-exercised in standard laboratory conditions, in both males and females. Less of or no decline is seen in diaphragm and masseter, muscles that are more normally used [40,41]. In the case of females, supplementation with E2 increases the number of satellite cells in the TA and soleus and reverses a subset of age-related gene expression changes [40]. The direct regulation of the satellite cell compartment by E2 in females prompts the question of whether E2 might also regulate satellite cells in males. Males have quite low levels of circulating E2; however, in both males and females, estrogen is produced through the action of aromatase on testosterone, a hormone males have in systemic abundance. Recently overexpression of aromatase in skeletal muscle of male mice led to increased serum and tissue E2 resulting in anabolic effects on muscle [42]. Therefore, it is possible that niche cells expressing aromatase might convert testosterone into locally high levels of E2 in the vicinity of satellite cells in male mice, maintaining the satellite cell pool via E2 independently of systemic levels. In that case, the deletion of Esr1 in male satellite cells would be predicted to result in a decline in satellite cell number. We previously observed a significant deficit in engraftment when we transplanted WT female satellite cells into OVX hosts compared to sham-treated hosts, as well as when we transplanted Esr1 KO female cells into WT female recipients compared to Esr1-WT female cells into WT female recipients [32]. If male cells were dependent on E2, whether local or systemic, then Esr1 KO would reduce their engraftment into male as well as female recipients. On the other hand, if satellite cells in muscle of males were intrinsically independent of E2 regulations, the cells should graft equally well into female recipients with or without Esr1 deletion.
To date the majority of satellite cell transplantation studies have been performed in a sex-matched manner, consequently intrinsic differences or similarities in the response to sex hormones as environmental factors has not been robustly elucidated. Some data exists on fiber differentiation in transplantation assays, and it suggests that WT male and female satellite cells have similar potentials with regard to in vivo differentiation [43], but neither the self-renewal of the stem cell compartment nor its E2 dependence is known.
In the current study we test the hypothesis that estradiol signaling through ERα regulates the male satellite cell pool in a manner comparable to females. We employ in vitro culture of male satellite cell-derived myoblasts in the presence or absence of 17β-estradiol, and we perform sex-matched and reciprocal-sex satellite cell transplantation. Finally, we evaluate the self-renewal potential of male satellite cells with satellite cell-specific temporally regulated genetic ablation of the Esr1 in the context of transplantation.

2. Materials and Methods

2.1. Mice

All procedures were performed in accordance with a protocol (2307-41280A) approved (3 November 2023) by the Institutional Animal Care and Use Committees at the University of Minnesota. All experiments were conducted on adult male and female mice 3–6 months of age. Male and female Pax7-ZsGreen mice were used for sex comparison experiments [44]; Esr1FL/FL; Pax7CreERT2; Pax7-ZsGreen (experimental), and Esr1FL/FL; Pax7-ZsGreen (Control, lacking Cre) mice were generated in-house [32]. Transplant recipients were NSG-mdx4Cv mice [45].
To induce Esr1 knockout, all mice were treated for five consecutive days with 80 mg/kg IP tamoxifen (Sigma Aldrich, St. Louis, MO, USA) dissolved at a concentration 12.75 mg in 1 mL sunflower oil, after which mice were then maintained on tamoxifen diet (TD.140251 Envigo, Indianapolis, IA, USA) for the remaining duration of each experiment. For the transplantation experiments, recipient mice were treated starting on the day of transplantation. This knockout protocol overcomes the ability of any escapers to reconstitute the satellite cell pool [1,18].

2.2. Isolation of Primary Murine Myoblasts

Primary myoblasts were isolated from adult (3–4-month-old) male and female C57Bl6/J mice via lineage depletion using the Miltenyi satellite cell isolation kit, mouse, according to manufacturer’s instructions (Miltenyi Biotec, Gaithersburg, MD, USA, Cat #130-104-268). Briefly, hind limb muscles were dissected, minced in parallel with muscle fibers, and digested in Dispase II and Collagenase type 2 for 30 min at 37 °C. The resulting slurry was manually dissociated, 4 mL of DMEM added and cells filtered through a 100 µM cell strainer. Cells were pelleted via centrifugation at 500× g for 5 min. Cells were resuspended in 80 µL of sorting buffer (PBS, pH 7.2, 0.5% BSA, 2 mM EDTA) and 20 µL of the Lin–cell separation microbeads and incubated for 15 min at 4 °C before magnetic separation. Unlabeled cells were collected, centrifuged at 500× g for 5 min, resuspended in Myogenic Growth Medium (DMEM/F12 medium (11320033; Thermo Scientific, Waltham, MA, USA) with 20% FBS (PS-FB3, PEAK Serum, Wellington, CO, USA), 10 ng/mL human basic fibroblast growth factor [bFGF; 100-18C; Peprotech, Cranbury, NJ, USA], 1% Pen/Strep [15140122; Gibco], and 1% Glutamax [35050061; Gibco]), and pre-plated for 90 min on an uncoated 10 cm plate at 37 °C and 5% CO2. Non-adherent (satellite) cells were transferred to a 0.1% gelatin-coated plate and allowed to proliferate until ≥80% confluent and thereafter considered primary myoblasts.

2.3. MTT Proliferation Assay

Mouse primary myoblasts were plated into 0.1% gelatin-coated 96-well plate (4000 cells/well) with phenol red-free F12/DMEM (SH30272.01; Hyclone, Logan, UT, USA) supplemented with 20% CS-FBS (NB036790; Thermo Fisher Scientific), 10 ng/mL human basic fibroblast growth factor, 1% Pen/Strep, and 1% Glutamax. Myoblasts were treated with 17β-Estradiol (E2) or vehicle every 24 h at a concentration of 100 pM (27.24 pg/mL) E2. The MTT assay was performed according to manufacturer’s instructions (11465007001; Roche, Indianapolis, IN, USA). After 48 or 72 h, the MTT labeling reagent [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide in PBS] was added to each well (final concentration 0.5 mg/mL) and incubated for 4 h. Cells were solubilized with DMSO and the formazan product measured in a microplate reader at a 570 nm wavelength.

2.4. EdU Proliferation Assay

Mouse primary myoblasts were plated into 0.1% gelatin-coated 96-well plate (4000 cells/well) with phenol red free DMEM/F12 (SH30284.01; Hyclone) supplemented with 20% CS-FBS (NB036790; Thermo Fisher Scientific), 10 ng/mL human basic fibroblast growth factor, 1% Pen/Strep, and 1% Glutamax. Myoblasts were treated with E2 or vehicle every 24 h at a concentration of 100 pM (27.24 pg/mL) E2. The Click-iT EdU proliferation assay for microplates was performed according to manufacturer’s instructions (C10499; Thermo Fisher Scientific). After 72 h of treatment, 10 µM EdU was added to cells for 8 h, cells were subsequently fixed, washed, and incubated with the Click-iT reaction cocktail. Cells were then blocked and incubated with the Amplex UltraRed reaction mixture. Fluorescence was subsequently read on a microplate reader with excitation at 568 nm and emission at 585 nm.

2.5. Western Blots

Cells were lysed with RIPA buffer supplemented with protease inhibitor cocktail (Complete, Roche), and proteins were separated on 10% SDS-PAGE gels, then transferred to PVDF membranes. Antibodies were diluted in 3% skim milk in TBST and incubated overnight at 4 °C or 1 h at RT. An appropriate HRP conjugated secondary antibody was incubated for 1 h at RT. Membranes were then washed with TBST, and signal was visualized using Pierce ECL Western blotting substrate (Thermo Scientific) for the ER alpha and SuperSignal West Femto Maximum Sensitivity Substrate (Thermo Scientific) for the GAPDH. Antibodies used in the study: GAPDH-HRP (1:5000, 60004, Proteintech), Mouse ER alpha /NR3A1 Antibody (1: 1000, MAB57151 R&D systems)

2.6. Flow Cytometry/FACS Sorting

Flow cytometric isolation of satellite cells from bulk hind limb or individual muscle digests (e.g., TA, EDL, Sol, and GC) were performed as described in detail previously [32,41]. Briefly, muscles were dissected, minced in parallel with muscle fibers, and digested with collagenase type II and dispase (17101-015 and 17105-041, resp.; Gibco, Grand Island, NY, USA). For isolation of satellite cells from C57BL/6 mice, mononuclear cells were stained using an antibody mixture of 1 µL PE-Cy7 rat anti-mouse CD31 (clone 390; 561410; BD Biosciences, San Diego, CA, USA), 1 µL PE-Cy7 rat anti-mouse CD45 (clone 30-F11; 552848; BD Biosciences), 1 µL Biotin rat anti-mouse CD106 (clone 429 (MVCAM.A); 553331; BD Biosciences), 1 µL PE Streptavidin (554061; BD Biosciences), and 2 µL alpha7 integrin 647 (clone R2F2; AbLab; Vancouver, BC, Canada). Samples were incubated with antibody cocktail, washed, and resuspended with FACS staining medium (2% Fetal Bovine Serum [FBS; 16000044; Gibco] in phosphate-buffered saline [PBS]) containing 0.5 µg/mL propidium iodide (PI) for analysis on a FACSAriaII SORP (BD Biosciences, San Diego, CA, USA). Total satellite cells (Lin–, i.e., not PE-Cy7+; double-positive, i.e., VCAM1+, Integrin α7+ cells) were analyzed from the entire muscle sample.
To obtain satellite cells from Pax7-ZsGreen mice, mononuclear cells were isolated as described above and were incubated in FACS staining medium containing PI [45]. Absolute satellite cell counts by FACS were confirmed through gating ZsGreen+ cells according to the manufacturer’s instructions. Researchers were blinded during all FACS analyses.

2.7. TA Injury and Transplantation

Adult (3–6 months old) Esr1fl/fl; Pax7-ZsGreen male mice carrying Pax7-creERT2 or Cre-negative controls were anesthetized with ketamine and xylazine, both hind limbs shaved and sterilized using surgical betadine solution, the skin over the TA was opened with a scissor, and both TA muscles were exposed. For injury-only studies, 30 µL of 1.2% BaCl2 was injected with a Hamilton syringe and the skin was closed using nonabsorbable suture. For transplantation experiments, 48 h prior to transplantation of cells, 4-month-old recipient NSG-mdx4Cv and C57BL/6J mice were anesthetized with ketamine and xylazine and both hind limbs were subjected to 1200 cGy irradiation using an RS 2000 Biological Research Irradiator (Rad Source Technologies, Inc., Suwanee, GA, USA) with lead shields protecting the body and forelimbs. Twenty-four hours prior to transplant, 15 µL cardiotoxin (10 µM in PBS, Sigma, St. Louis, MO, USA) was injected into the TA under anesthesia as above. Twenty-four hours later, 300 ZsGreen+ satellite cells were collected by FACS from donor mice and transplanted in a volume of 10 µL PBS into each TA. Four weeks after transplantation, TA was prepared for FACS analysis as described above to count the number of ZsGreen+ cells.

2.8. RNA Isolation, Reverse Transcription, and qRT-PCR

Total RNA was extracted using a Qiazol reagent (Qiagen) as described in the manufacturer’s protocol. First strand cDNA was generated using the iScript cDNA synthesis kit (BioRad, Hercules, CA, USA). Relative quantification of estrogen receptors was determined by Taqman probes, ERa (ESR1—Mm00433149_m1), ERb (ESR2—Mm00599819_m1), GPER (Mm02620446_s1), and Androgen receptor (Mm0042688_m1) using FAM-based chemistry on a CFX Connect real-time PCR detection system (Bio-Rad). Gene expression was determined with the ∆Ct relative quantification method and shown as relative to B2M (Mm00437762_m1), Tbp (Mm00446973_m1), Hprt (Mm03024075_m1), or Gapdh (Mm99999915_g1).

2.9. Statistical Analysis

All data were analyzed with two-tailed unpaired Student t-tests for determining significant differences between two groups or one-way ANOVA with Tukey’s post hoc for determining significant differences between three or more groups; data was tested for normality using Kolmogorov–Smirnov test. Significance was considered at p values of ≤0.05. Data are presented as mean ± SEM. All statistical testing was performed using GraphPad Prism 8.0 (GraphPad Software Inc., San Diego, CA, USA).

3. Results

3.1. Male Primary Myoblasts Express Esr1 and Are Equally as Responsive to E2 as Female Cells

E2 signaling promotes the proliferation of primary myoblasts derived from satellite cells of female mice [34]. To characterize the response of male primary myoblasts to E2, we isolated satellite cells from male and female C57Bl/6J by MACS (Figure 1A). We assessed the expression of the estrogen receptors, ERα and G-Protein coupled estrogen receptor (GPER) (Figure 1B), and found them both expressed at to at least their respective levels in female myoblasts, suggesting potential activity of the pathway. To assess the ERα protein level, we sorted Intga7+/VCAM1+ cells by FACS from male and female C57Bl/6J mice and analyzed by Western blot. We found both male and female satellite cells have detectable ERα protein, slightly higher in male cells, similar to the Esr1 mRNA results (Figure 1C). Next, we passaged primary myoblasts from male and female (n = 4 male mice and 3 female mice) C57BL/6J mice into growth medium with charcoal-stripped serum, to eliminate any influence of background steroid hormones, adding either E2 or vehicle for up to 72 h. We found that male and female satellite cells responded equally well to E2 with regard to proliferation, as measured by MTT assay at 48 and 72 h post plating (Figure 1D). We confirmed this using an independent measure of proliferation, EdU incorporation, and found similar increases in EdU incorporation in male and female cells after 72 h of E2 treatment (Figure 1E).

3.2. Male and Female SC Engraft Similarly Regardless of the Sex of the Recipient

We have developed a transplantation-based assay to monitor self-renewal of satellite cells in which 300 Pax7-ZsGreen cells are transplanted into injured, irradiated tibialis anterior (TA) muscles of recipient mice [44,46]. One month after transplantation, the TA muscle is dissected, digested, then analyzed by FACS to count the number of Pax7-Zsgreen cells that self-renewed after transplantation to contribute to the satellite cell pool (Figure 2A and Figure S1A). We employed this assay to investigate the effect of sex as a determinant of engraftment into the satellite cell pool, by isolating Pax7-ZsGreen cells from males and females and transplanting into the same or the reciprocal sex. We found no significant differences, i.e., satellite cells from males and females engrafted into the Pax7+ pool to a similar extent. Interestingly we found more cells in the TA of the male recipients compared to female recipients, regardless of the sex of the donor (Figure 2B), a result that correlates with the larger muscle size, and larger endogenous pre-irradiation satellite cell pool size, of the male TA compared to the female [40,41]. We found similar results when we repeated the experiment using NSG-mdx4Cv recipients (Figure 2C), namely that male and female WT Pax7-ZsGreen cells engrafted into the satellite cell pool equally well; however, the average number of Pax7-ZsGreen cells after transplantation of 300 cells into NSG-mdx4Cv recipients was significantly lower than that seen in the WT C57BL6/J recipients, suggesting that the DMD pathology suppresses their ability to contribute, possibly by exposing transplanted cells to a more stressful environment (Figure S1A,B).

3.3. Effect of Satellite Cell-Specific Deletion of Esr1 in Males During Homeostasis

To characterize the importance of Esr1 signaling in the satellite cells, we used the inducible satellite cell-specific Esr1 knockout mouse [32]. Male mice carrying or lacking Pax7-creERT2 were treated with IP tamoxifen for 5 consecutive days, and two weeks after the last dose of tamoxifen, the hind limb muscles were dissected, enzymatically digested, then stained for satellite cell FACS analysis using the Lin(CD45/CD31)- Itga7+VCAM1+ strategy [47] (Figure S2A). To confirm recombination, we isolated the RNA from sorted cells and tested the expression of the Esr1, GPER, and Androgen receptor, AR, which showed a statistically significant decline in the expression of Esr1 in cre+ animals (Esr1-mutant satellite cells) relative to their respective cre-negative controls (Figure 3A). However, when we quantified satellite cells, neither absolute number nor density of satellite cells differed between the cre- and cre+ male mice in the TA, gastrocnemius, and soleus muscles (Figure 3B,C). We tested Esr1-knockout females side by side with the males, and found, as expected, a significant decline in the number of Pax7-ZsGreen+ cells in the muscles of cre+ females compared to cre- controls (Figure S2B).

3.4. Effect of Satellite Cell Esr1 Ablation on Satellite Cell Pool Response to Injury

To evaluate the necessity of ERα signaling in male satellite cells in the muscle’s response to injury, we unilaterally injured the TA muscles of male mice with BaCl2. Twenty-one days following the first injury, a second injury to the same muscle was induced and mice were euthanized 28 days later. The number of Pax7-ZsGreen+ cells was quantified from the injured and contralateral uninjured TA by FACS, revealing a statistically significant expansion of the satellite cells pool after injury, in both the Esr1 WT and KO mice (Figure 4A,B).

3.5. ERα Is Necessary for Male Satellite Cell Self-Renewal in the Female, but Not Male, Environment

Our previous results found a strong dependence of satellite cell engraftment on the availability of E2 in the host environment and an intact estrogen receptor (ERα, Esr1) [32]. We therefore tested whether ERα was necessary in male satellite cells, using the in vivo transplantation assay because it is the most stringent way to evaluate the self-renewal potential of satellite cells, putting tremendous expansion pressure on a small number of donor cells that are also called upon to generate fibers. For these studies, we used 300 Pax7-ZsGreen cells from donors Esr1FL/FL; +/− Pax7-CreERT2; Pax7-ZsGreen male mice, such that satellite cells would be marked with green fluorescence if they self-renewed and ERα could be conditionally eliminated only from satellite cells. ZsGreen+ cells were transplanted into irradiated and injured TA muscles of wild-type C57BL/6J females, with the recipients treated with tamoxifen to induce the recombination. The mice were kept on tamoxifen diet throughout the experiment to suppress any deletion escapers (Figure 5A) [18]. We found that deletion of the ERα in male cells significantly reduced their self-renewal potential in the female E2-replete host environment (Figure 5B and Figure S3A). We repeated the same assay with independent donors and the recipient mice were treated with the vehicle only instead, and in this paradigm, we found no difference in the absence of tamoxifen (Figure 5C), supporting that the decline in the self-renewal potential was solely due to ERα deletion.
We then repeated the same transplantation assay using C57BL/6J male recipients instead (Figure 5D), and here we found no difference in the self-renewal between the ERα WT and knockout cells, supporting the interpretation that the dependence of male satellite cells on ERα signaling occurs only in a female environment (Figure 5E and Figure S3B).

4. Discussion

Sex-specific differences in adult skeletal muscle including those in satellite cells have been reported [48,49,50,51,52]. While the effects of androgens in maintaining normal muscle physiology in males have been evaluated [53,54], the necessity or activity of estrogens in males is largely obscure.
We have shown in both aged males [41] and females [40] that although satellite cells decline in number in locomotory muscles with age, they do not lose their capacity to engraft and regenerate if transplanted into a young environment. On the other hand, self-renewal of young or old satellite cells is impaired when they are transplanted into an aged female environment, an effect that can be countered to some extent by the provision of hormone (E2) replacement therapy [40]. This demonstrated that non-cell-autonomous environmental factors dominated over cell-autonomous age in driving age-related regenerative deficiencies of satellite cells. We have also shown that in females, E2 is an important factor in setting the satellite cell pool size, with loss of E2 signaling via OVX or satellite cell-specific deletion of Esr1 leading to 30–50% decline in satellite cell number in most muscles [32]. The current study was undertaken to determine to what extent E2 signaling may be operative in male satellite cells, even though males have much lower levels of systemic E2 than females do. Estrogens are secreted primarily from the granulosa cells of the ovaries in females and the Sertoli cells of the testes in males [30,55], but in both sexes, additional tissues including adipose also endogenously produce estrogens [56,57]. Whether the low levels of E2 in males impact satellite cell maintenance or self-renewal was unknown. Some data exists on the response of male satellite cells to female sex hormones in the context of experimentally elevating E2 levels in males. Studies have reported the beneficial role of exogenously supplied estrogen in preventing disuse muscle atrophy in male rats [58] and a greater extent of 3-day post-injury (downhill running)-induced increase in Pax7+ cells number after estrogen administration, again in male rats [59]. These data do not distinguish between direct responses of satellite cells to estrogen and changes in the environment mediated by systemic estrogen delivery that indirectly impact satellite cells through other pathways; the potential effect of the low endogenous level of E2 in males was also not addressed. They are also not relevant to the hypothesis that male cells respond to locally produced E2 from the action of aromatase in niche cells on circulating testosterone. Here, we show that male satellite cells express Esr1 at levels equal to or greater than female satellite cells, suggesting the possibility of their direct regulation by E2, either systemic or local.
Our data shows that deletion of Esr1 in satellite cells has no effect on the homeostatic maintenance of pool size in males, nor does it reduce the expansion in pool size seen after severe BaCl2 injury in male TA muscles. The most stringent test of self-renewal potential is arguably the satellite cell transplantation assay, in which 300 satellite cells are delivered into a TA muscle that has been irradiated and injured with BaCl2 or cardiotoxin [41,45]. This forces the satellite cells to regenerate a volume of muscle that would normally harbor an order of magnitude more satellite cells than are being transplanted, in addition to repopulating it with the self-renewed pool. Three hundred cells is also within the linear range of this assay, which begins to show severe non-linearity at around 1000 cells or more [41], meaning that at 300 cells it is very sensitive to minor changes in self-renewal potential. Even under this very sensitive assay, we find no difference between Esr1 KO and WT satellite cells, when transplanted into males. On the other hand, when the same cells were transplanted into females, the Esr1 KO satellite cells showed a greatly diminished self-renewal potential. This clearly demonstrates that male cells are competent for estrogen signaling, but that it is apparently irrelevant for self-renewal in the normal male environment. These data clearly rule out the hypothesis that male satellite cells respond to local aromatase-generated E2 derived from circulating testosterone. They also rule out a male satellite cell dependence on circulating E2. We propose two possible explanations for the lack of functional effect with loss of Esr1 in males: (1) the levels of E2, being much lower in normal males, may simply be below the threshold necessary for physiological effect on satellite cells, or (2) another signaling pathway may be operating in males that makes E2 signaling redundant, in which case eliminating that pathway might expose an E2 dependence in males. Previous studies with cross-sex transplants that did not involve hormone signaling manipulation showed that male and female satellite cells produce roughly equivalent numbers of myofibers when transplanted into male or female hosts [43]. If the second proposition is correct, this presumably means that female satellite cells should also be competent to respond to the compensatory signaling pathway operating in males. The possibility that testosterone might constitute such a pathway is supported by the finding that testosterone administration in male subjects leads to elevated PAX7+ cell number [60].

5. Conclusions

In summary, these studies demonstrate that estradiol signaling via estrogen receptor alpha is fully functional in male satellite cells, although not necessary in a normal male environment, ruling out the hypothesis that testosterone is converted to E2 by local aromatase to regulate male satellite cells. However, when male satellite cells find themselves in a female environment, they are strongly dependent on E2 for self-renewal, showing that sexually dimorphic mechanisms mediated by the environment regulate satellite cell pool size.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/cells14201606/s1, Figure S1: FACS gating and comparing engraftment in WT vs. NSG-mdx4Cv recipients; Figure S2: FACS gating and normalization by muscle mass of Pax7-ZsGreen counts; Figure S3: Representative FACS plots for transplantation of Esr1FL/FL male cells.

Author Contributions

Investigation, A.S.S., B.P.S., and E.A.T.; formal analysis, A.S.S., B.P.S., E.A.T., D.A.L., and M.K.; writing—original draft preparation, A.S.S. and M.K.; writing—review and editing, D.A.L. and B.P.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by a grant from the National Institutes of Health/National Institute on Aging (R01 AG062899). B.P.S. was supported by T32 AR007612.

Institutional Review Board Statement

The animal study protocol was approved by the Institutional Animal Care and Use Committee of the University of Minnesota (2307-41280A, approved on 3 November 2023).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data associated with this study will be made available upon reasonable request to Michael Kyba.

Acknowledgments

We thank Cynthia Faraday for artwork.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ARAndrogen receptor
DMEMDulbeco’s modified Eagle medium
E217β-Estradiol
EdU5-ethynyl-2′-deoxyuridine
EREstrogen receptor
FBSFetal bovine serum
GPERG-Protein coupled estrogen receptor
KOKnockout
Lin–Lineage-negative
MTT3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
OVXOvariectomy
PBSPhosphate-buffered saline
PIPropidium iodide
SCSatellite cell
scERαKOSatellite cell Esr1 knockout
scERαWTSatellite cell Esr1 wild-type
SEMStandard error of the mean
TATibialis anterior (muscle)
WTWild-type

References

  1. Von Maltzahn, J.; Jones, A.E.; Parks, R.J.; Rudnicki, M.A. Pax7 is critical for the normal function of satellite cells in adult skeletal muscle. Proc. Natl. Acad. Sci. USA 2013, 110, 16474–16479. [Google Scholar] [CrossRef]
  2. Günther, S.; Kim, J.; Kostin, S.; Lepper, C.; Fan, C.-M.; Braun, T. Myf5-positive satellite cells contribute to Pax7-dependent long-term maintenance of adult muscle stem cells. Cell Stem Cell 2013, 13, 590–601. [Google Scholar] [CrossRef] [PubMed]
  3. Seale, P.; Sabourin, L.A.; Girgis-Gabardo, A.; Mansouri, A.; Gruss, P.; Rudnicki, M.A. Pax7 is required for the specification of myogenic satellite cells. Cell 2000, 102, 777–786. [Google Scholar] [CrossRef] [PubMed]
  4. Mauro, A. Satellite cell of skeletal muscle fibers. J. Biophys. Biochem. Cytol. 1961, 9, 493. [Google Scholar] [CrossRef]
  5. Collins, C.A.; Olsen, I.; Zammit, P.S.; Heslop, L.; Petrie, A.; Partridge, T.A.; Morgan, J.E. Stem cell function, self-renewal, and behavioral heterogeneity of cells from the adult muscle satellite cell niche. Cell 2005, 122, 289–301. [Google Scholar] [CrossRef]
  6. Yin, H.; Price, F.; Rudnicki, M.A. Satellite cells and the muscle stem cell niche. Physiol. Rev. 2013, 93, 23–67. [Google Scholar] [CrossRef] [PubMed]
  7. Verma, M.; Asakura, Y.; Murakonda, B.S.R.; Pengo, T.; Latroche, C.; Chazaud, B.; McLoon, L.K.; Asakura, A. Muscle satellite cell cross-talk with a vascular niche maintains quiescence via VEGF and notch signaling. Cell Stem Cell 2018, 23, 530–543.e9. [Google Scholar] [CrossRef] [PubMed]
  8. Webster, M.T.; Manor, U.; Lippincott-Schwartz, J.; Fan, C.-M. Intravital imaging reveals ghost fibers as architectural units guiding myogenic progenitors during regeneration. Cell Stem Cell 2016, 18, 243–252. [Google Scholar] [CrossRef]
  9. Chakkalakal, J.V.; Christensen, J.; Xiang, W.; Tierney, M.T.; Boscolo, F.S.; Sacco, A.; Brack, A.S. Early forming label-retaining muscle stem cells require p27kip1 for maintenance of the primitive state. Development 2014, 141, 1649–1659. [Google Scholar] [CrossRef]
  10. Conboy, I.M.; Conboy, M.J.; Wagers, A.J.; Girma, E.R.; Weissman, I.L.; Rando, T.A. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 2005, 433, 760–764. [Google Scholar] [CrossRef]
  11. Der Vartanian, A.; Quétin, M.; Michineau, S.; Auradé, F.; Hayashi, S.; Dubois, C.; Rocancourt, D.; Drayton-Libotte, B.; Szegedi, A.; Buckingham, M. PAX3 confers functional heterogeneity in skeletal muscle stem cell responses to environmental stress. Cell Stem Cell 2019, 24, 958–973.e9. [Google Scholar] [CrossRef]
  12. Brack, A.S.; Conboy, M.J.; Roy, S.; Lee, M.; Kuo, C.J.; Keller, C.; Rando, T.A. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science 2007, 317, 807–810. [Google Scholar] [CrossRef]
  13. Price, F.D.; Von Maltzahn, J.; Bentzinger, C.F.; Dumont, N.A.; Yin, H.; Chang, N.C.; Wilson, D.H.; Frenette, J.; Rudnicki, M.A. Inhibition of JAK-STAT signaling stimulates adult satellite cell function. Nat. Med. 2014, 20, 1174–1181. [Google Scholar] [CrossRef]
  14. Elabd, C.; Cousin, W.; Upadhyayula, P.; Chen, R.Y.; Chooljian, M.S.; Li, J.; Kung, S.; Jiang, K.P.; Conboy, I.M. Oxytocin is an age-specific circulating hormone that is necessary for muscle maintenance and regeneration. Nat. Commun. 2014, 5, 4082. [Google Scholar] [CrossRef]
  15. Brack, A.S.; Conboy, I.M.; Conboy, M.J.; Shen, J.; Rando, T.A. A temporal switch from notch to Wnt signaling in muscle stem cells is necessary for normal adult myogenesis. Cell Stem Cell 2008, 2, 50–59. [Google Scholar] [CrossRef]
  16. Carlson, M.E.; Hsu, M.; Conboy, I.M. Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells. Nature 2008, 454, 528–532. [Google Scholar] [CrossRef] [PubMed]
  17. Palacios, D.; Mozzetta, C.; Consalvi, S.; Caretti, G.; Saccone, V.; Proserpio, V.; Marquez, V.E.; Valente, S.; Mai, A.; Forcales, S.V. TNF/p38α/polycomb signaling to Pax7 locus in satellite cells links inflammation to the epigenetic control of muscle regeneration. Cell Stem Cell 2010, 7, 455–469. [Google Scholar] [CrossRef]
  18. Shams, A.S.; Arpke, R.W.; Gearhart, M.D.; Weiblen, J.; Mai, B.; Oyler, D.; Bosnakovski, D.; Mahmoud, O.M.; Hassan, G.M.; Kyba, M. The chemokine receptor CXCR4 regulates satellite cell activation, early expansion, and self-renewal, in response to skeletal muscle injury. Front. Cell Dev. Biol. 2022, 10, 949532. [Google Scholar] [CrossRef] [PubMed]
  19. Feldman, H.A.; Longcope, C.; Derby, C.A.; Johannes, C.B.; Araujo, A.B.; Coviello, A.D.; Bremner, W.J.; McKinlay, J.B. Age trends in the level of serum testosterone and other hormones in middle-aged men: Longitudinal results from the Massachusetts male aging study. J. Clin. Endocrinol. Metab. 2002, 87, 589–598. [Google Scholar] [CrossRef]
  20. Tepper, P.G.; Randolph, J.F., Jr.; McConnell, D.S.; Crawford, S.L.; El Khoudary, S.R.; Joffe, H.; Gold, E.B.; Zheng, H.; Bromberger, J.T.; Sutton-Tyrrell, K. Trajectory clustering of estradiol and follicle-stimulating hormone during the menopausal transition among women in the Study of Women’s Health across the Nation (SWAN). J. Clin. Endocrinol. Metab. 2012, 97, 2872–2880. [Google Scholar] [CrossRef] [PubMed]
  21. Collins, B.C.; Laakkonen, E.K.; Lowe, D.A. Aging of the musculoskeletal system: How the loss of estrogen impacts muscle strength. Bone 2019, 123, 137–144. [Google Scholar] [CrossRef]
  22. Phillips, S.; Rook, K.; Siddle, N.; Bruce, S.; Woledge, R. Muscle weakness in women occurs at an earlier age than in men, but strength is preserved by hormone replacement therapy. Clin. Sci. 1993, 84, 95–98. [Google Scholar] [CrossRef] [PubMed]
  23. Phillips, S.; Sanderson, A.; Birch, K.; Bruce, S.; Woledge, R. Changes in maximal voluntary force of human adductor pollicis muscle during the menstrual cycle. J. Physiol. 1996, 496, 551–557. [Google Scholar] [CrossRef]
  24. Greising, S.M.; Baltgalvis, K.A.; Lowe, D.A.; Warren, G.L. Hormone therapy and skeletal muscle strength: A meta-analysis. J. Gerontol. Ser. A: Biomed. Sci. Med. Sci. 2009, 64, 1071–1081. [Google Scholar] [CrossRef]
  25. Taaffe, D.R.; Sipilä, S.; Cheng, S.; Puolakka, J.; Toivanen, J.; Suominen, H. The effect of hormone replacement therapy and/or exercise on skeletal muscle attenuation in postmenopausal women: A yearlong intervention. Clin. Physiol. Funct. Imaging 2005, 25, 297–304. [Google Scholar] [CrossRef] [PubMed]
  26. Rolland, Y.M.; Perry Iii, H.M.; Patrick, P.; Banks, W.A.; Morley, J.E. Loss of appendicular muscle mass and loss of muscle strength in young postmenopausal women. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2007, 62, 330–335. [Google Scholar] [CrossRef]
  27. Ronkainen, P.H.; Kovanen, V.; Alén, M.; Pollanen, E.; Palonen, E.-M.; Ankarberg-Lindgren, C.; Hamalainen, E.; Turpeinen, U.; Kujala, U.M.; Puolakka, J. Postmenopausal hormone replacement therapy modifies skeletal muscle composition and function: A study with monozygotic twin pairs. J. Appl. Physiol. 2009, 107, 25–33. [Google Scholar] [CrossRef]
  28. Qaisar, R.; Renaud, G.; Hedstrom, Y.; Pöllänen, E.; Ronkainen, P.; Kaprio, J.; Alen, M.; Sipilä, S.; Artemenko, K.; Bergquist, J. Hormone replacement therapy improves contractile function and myonuclear organization of single muscle fibres from postmenopausal monozygotic female twin pairs. J. Physiol. 2013, 591, 2333–2344. [Google Scholar] [CrossRef] [PubMed]
  29. Sipilä, S.; Taaffe, D.R.; Cheng, S.; Puolakka, J.; Toivanen, J.; Suominen, H. Effects of hormone replacement therapy and high-impact physical exercise on skeletal muscle in post-menopausal women: A randomized placebo-controlled study. Clin. Sci. 2001, 101, 147–157. [Google Scholar] [CrossRef]
  30. McMillin, S.L.; Minchew, E.C.; Lowe, D.A.; Spangenburg, E.E. Skeletal muscle wasting: The estrogen side of sexual dimorphism. Am. J. Physiol.-Cell Physiol. 2022, 322, C24–C37. [Google Scholar] [CrossRef]
  31. Stephany, R.W. Hormonal growth promoting agents in food producing animals. In Doping in Sports: Biochemical Principles, Effects and Analysis; Springer: Berlin/Heidelberg, Germany, 2010; pp. 355–367. [Google Scholar]
  32. Collins, B.C.; Arpke, R.W.; Larson, A.A.; Baumann, C.W.; Xie, N.; Cabelka, C.A.; Nash, N.L.; Juppi, H.-K.; Laakkonen, E.K.; Sipilä, S. Estrogen regulates the satellite cell compartment in females. Cell Rep. 2019, 28, 368–381.e6. [Google Scholar] [CrossRef] [PubMed]
  33. Larson, A.A.; Baumann, C.W.; Kyba, M.; Lowe, D.A. Oestradiol affects skeletal muscle mass, strength and satellite cells following repeated injuries. Exp. Physiol. 2020, 105, 1700–1707. [Google Scholar] [CrossRef] [PubMed]
  34. Larson, A.A.; Shams, A.S.; McMillin, S.L.; Sullivan, B.P.; Vue, C.; Roloff, Z.A.; Batchelor, E.; Kyba, M.; Lowe, D.A. Estradiol deficiency reduces the satellite cell pool by impairing cell cycle progression. Am. J. Physiol.-Cell Physiol. 2022, 322, C1123–C1137. [Google Scholar] [CrossRef]
  35. Sousa-Victor, P.; Gutarra, S.; García-Prat, L.; Rodriguez-Ubreva, J.; Ortet, L.; Ruiz-Bonilla, V.; Jardí, M.; Ballestar, E.; González, S.; Serrano, A.L. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature 2014, 506, 316–321. [Google Scholar] [CrossRef]
  36. Brack, A.S.; Bildsoe, H.; Hughes, S.M. Evidence that satellite cell decrement contributes to preferential decline in nuclear number from large fibres during murine age-related muscle atrophy. J. Cell Sci. 2005, 118, 4813–4821. [Google Scholar] [CrossRef] [PubMed]
  37. Chakkalakal, J.V.; Jones, K.M.; Basson, M.A.; Brack, A.S. The aged niche disrupts muscle stem cell quiescence. Nature 2012, 490, 355–360. [Google Scholar] [CrossRef] [PubMed]
  38. Day, K.; Shefer, G.; Shearer, A.; Yablonka-Reuveni, Z. The depletion of skeletal muscle satellite cells with age is concomitant with reduced capacity of single progenitors to produce reserve progeny. Dev. Biol. 2010, 340, 330–343. [Google Scholar] [CrossRef]
  39. Collins, C.A.; Zammit, P.S.; Ruiz, A.P.; Morgan, J.E.; Partridge, T.A. A population of myogenic stem cells that survives skeletal muscle aging. Stem Cells 2007, 25, 885–894. [Google Scholar] [CrossRef] [PubMed]
  40. Sullivan, B.P.; Larson, A.A.; Shams, A.S.; McMillin, S.L.; Ebeling, M.C.; Peng, S.; Kyba, M.; Lowe, D.A. Estradiol deficiency as a consequence of aging contributes to the depletion of the satellite cell pool in female mice. Aging Cell 2025, 24, e14441. [Google Scholar] [CrossRef]
  41. Arpke, R.W.; Shams, A.S.; Collins, B.C.; Larson, A.A.; Lu, N.; Lowe, D.A.; Kyba, M. Preservation of satellite cell number and regenerative potential with age reveals locomotory muscle bias. Skelet. Muscle 2021, 11, 22. [Google Scholar] [CrossRef] [PubMed]
  42. Unger, C.A.; Aladhami, A.K.; Hope, M.C., III; Cotham, W.E.; Nettles, K.W.; Clegg, D.J.; Velázquez, K.T.; Enos, R.T. Skeletal muscle endogenous estrogen production ameliorates the metabolic consequences of a high-fat diet in male mice. Endocrinology 2023, 164, bqad105. [Google Scholar] [CrossRef]
  43. Neal, A.; Boldrin, L.; Morgan, J.E. The satellite cell in male and female, developing and adult mouse muscle: Distinct stem cells for growth and regeneration. PLoS ONE 2012, 7, e37950. [Google Scholar]
  44. Bosnakovski, D.; Xu, Z.; Li, W.; Thet, S.; Cleaver, O.; Perlingeiro, R.C.; Kyba, M. Prospective isolation of skeletal muscle stem cells with a Pax7 reporter. Stem Cells 2008, 26, 3194–3204. [Google Scholar] [CrossRef]
  45. Arpke, R.W.; Darabi, R.; Mader, T.L.; Zhang, Y.; Toyama, A.; Lonetree, C.-l.; Nash, N.; Lowe, D.A.; Perlingeiro, R.C.; Kyba, M. A new immuno-, dystrophin-deficient model, the NSG-mdx 4Cv mouse, provides evidence for functional improvement following allogeneic satellite cell transplantation. Stem Cells 2013, 31, 1611–1620. [Google Scholar] [CrossRef]
  46. Arpke, R.W.; Kyba, M. Flow Cytometry and Transplantation-Based Quantitative Assays for Satellite Cell Self-Renewal and Differentiation. In Skeletal Muscle Regeneration in the Mouse: Methods and Protocols; Springer: New York, NY, USA, 2016; pp. 163–179. [Google Scholar]
  47. Chan, S.S.; Shi, X.; Toyama, A.; Arpke, R.W.; Dandapat, A.; Iacovino, M.; Kang, J.; Le, G.; Hagen, H.R.; Garry, D.J.; et al. Mesp1 patterns mesoderm into cardiac, hematopoietic, or skeletal myogenic progenitors in a context-dependent manner. Cell Stem Cell 2013, 12, 587–601. [Google Scholar] [CrossRef]
  48. Deasy, B.M.; Lu, A.; Tebbets, J.C.; Feduska, J.M.; Schugar, R.C.; Pollett, J.B.; Sun, B.; Urish, K.L.; Gharaibeh, B.M.; Cao, B. A role for cell sex in stem cell–mediated skeletal muscle regeneration: Female cells have higher muscle regeneration efficiency. J. Cell Biol. 2007, 177, 73–86. [Google Scholar] [CrossRef]
  49. LaBarge, S.; McDonald, M.; Smith-Powell, L.; Auwerx, J.; Huss, J.M. Estrogen-related receptor-α (ERRα) deficiency in skeletal muscle impairs regeneration in response to injury. FASEB J. 2014, 28, 1082. [Google Scholar] [CrossRef]
  50. Jomard, C.; Gondin, J. Influence of sexual dimorphism on satellite cell regulation and inflammatory response during skeletal muscle regeneration. Physiol. Rep. 2023, 11, e15798. [Google Scholar] [CrossRef]
  51. Tseng, L.A.; Delmonico, M.J.; Visser, M.; Boudreau, R.M.; Goodpaster, B.H.; Schwartz, A.V.; Simonsick, E.M.; Satterfield, S.; Harris, T.; Newman, A.B. Body composition explains sex differential in physical performance among older adults. J. Gerontol. Ser. A Biomed. Sci. Med. Sci. 2014, 69, 93–100. [Google Scholar]
  52. You, J.S.; Barai, P.; Chen, J. Sex differences in skeletal muscle size, function, and myosin heavy chain isoform expression during post-injury regeneration in mice. Physiol. Rep. 2023, 11, e15791. [Google Scholar]
  53. MacLean, H.E.; Chiu, W.M.; Notini, A.J.; Axell, A.-M.; Davey, R.A.; McManus, J.F.; Ma, C.; Plant, D.R.; Lynch, G.S.; Zajac, J.D. Impaired skeletal muscle development and function in male, but not female, genomic androgen receptor knockout mice. FASEB J. 2008, 22, 2676–2689. [Google Scholar] [CrossRef]
  54. Klose, A.; Liu, W.; Paris, N.D.; Forman, S.; Krolewski, J.J.; Nastiuk, K.L.; Chakkalakal, J.V. Castration induces satellite cell activation that contributes to skeletal muscle maintenance. JCSM Rapid Commun. 2018, 1, 1–16. [Google Scholar] [CrossRef]
  55. Cooke, P.S.; Nanjappa, M.K.; Ko, C.; Prins, G.S.; Hess, R.A. Estrogens in male physiology. Physiol. Rev. 2017, 97, 995–1043. [Google Scholar] [CrossRef]
  56. Barakat, R.; Oakley, O.; Kim, H.; Jin, J.; Ko, C.J. Extra-gonadal sites of estrogen biosynthesis and function. BMB Rep. 2016, 49, 488. [Google Scholar] [CrossRef]
  57. Schneider, G.; Kirschner, M.A.; Berkowitz, R.; Ertel, N.H. Increased estrogen production in obese men. J. Clin. Endocrinol. Metab. 1979, 48, 633–638. [Google Scholar] [CrossRef]
  58. Sugiura, T.; Ito, N.; Goto, K.; Naito, H.; Yoshioka, T.; Powers, S.K. Estrogen administration attenuates immobilization-induced skeletal muscle atrophy in male rats. J. Physiol. Sci. 2006, 56, 393–399. [Google Scholar] [CrossRef]
  59. Tiidus, P.; Deller, M.; Liu, X.J. Oestrogen influence on myogenic satellite cells following downhill running in male rats: A preliminary study. Acta Physiol. Scand. 2005, 184, 67–72. [Google Scholar] [CrossRef]
  60. Sinha-Hikim, I.; Roth, S.M.; Lee, M.I.; Bhasin, S. Testosterone-induced muscle hypertrophy is associated with an increase in satellite cell number in healthy, young men. Am. J. Physiol.-Endocrinol. Metab. 2003, 285, E197–E205. [Google Scholar] [CrossRef]
Figure 1. Male and female SCs respond similarly to E2 in vitro. (A) Schematic for the in vitro experiment where isolated male and female satellite cells were cultured ± E2 for 48 and 72 h, and analyzed by MTT assay and quantifying EdU+ cells. (B) RTqPCR for Esr1, Gper, and AR in satellite cells presented as fold change of the level of expression in the female cells. (C) Western blots for ERα and GAPDH control in Lin–Intga7/VCAM1 freshly sorted satellite cells from C57BL/6 male and female mice. (D) MTT cell viability assay showing percentage of absorbance relative to the vehicle-treated cells. (E) Percentage of EdU incorporation in cells after 72 h in culture with E2, relative to vehicle-treated cells. Charcoal-stripped serum is used to eliminate any background steroid hormones. Data are presented as mean ± SEM, * p < 0.05, by t-test for (B,E) and two-way ANOVA for (D).
Figure 1. Male and female SCs respond similarly to E2 in vitro. (A) Schematic for the in vitro experiment where isolated male and female satellite cells were cultured ± E2 for 48 and 72 h, and analyzed by MTT assay and quantifying EdU+ cells. (B) RTqPCR for Esr1, Gper, and AR in satellite cells presented as fold change of the level of expression in the female cells. (C) Western blots for ERα and GAPDH control in Lin–Intga7/VCAM1 freshly sorted satellite cells from C57BL/6 male and female mice. (D) MTT cell viability assay showing percentage of absorbance relative to the vehicle-treated cells. (E) Percentage of EdU incorporation in cells after 72 h in culture with E2, relative to vehicle-treated cells. Charcoal-stripped serum is used to eliminate any background steroid hormones. Data are presented as mean ± SEM, * p < 0.05, by t-test for (B,E) and two-way ANOVA for (D).
Cells 14 01606 g001
Figure 2. Male and female SCs engraft similarly, regardless of the sex of the recipient. (A) Schematic for the transplantation experiment showing the timing of irradiation and TA injury in the recipient mice. (B) Total number of Pax7-ZsGreen+ cells in TA muscles of recipient C57 BL/6 mice. Males (n = 6). Female (n = 6). (C) Total number of Pax7-ZsGreen+ cells in TA muscles of recipient NSG-mdx4cv mice. Males (n = 6). Female (n = 6). Data are presented as mean ± SEM; p values were calculated by two-way ANOVA.
Figure 2. Male and female SCs engraft similarly, regardless of the sex of the recipient. (A) Schematic for the transplantation experiment showing the timing of irradiation and TA injury in the recipient mice. (B) Total number of Pax7-ZsGreen+ cells in TA muscles of recipient C57 BL/6 mice. Males (n = 6). Female (n = 6). (C) Total number of Pax7-ZsGreen+ cells in TA muscles of recipient NSG-mdx4cv mice. Males (n = 6). Female (n = 6). Data are presented as mean ± SEM; p values were calculated by two-way ANOVA.
Cells 14 01606 g002
Figure 3. SC-specific deletion of Esr1 does not deplete the satellite cell compartment in males. (A) RTqPCR for Esr1 gene in the VCAM1+/Intga7+ cells from Pax7-cre- or cre+ male mice treated with tamoxifen (n = 4). (B) Total number of CD45/CD31(Lineage)-negative, VCAM1+/Itga7+-positive cells in tibialis anterior (TA), gastrocnemius (Gastroc), and soleus muscles in Esr1FL/FL, Pax7-cre +/− males. (C) Density of CD45/CD31(Lineage)-negative, VCAM1+/Itga+ cells in TA, gastrocnemius and soleus in Esr1FL/FL, Pax7-cre +/− males. Data are presented as mean ± SEM, p values calculated by t-test.
Figure 3. SC-specific deletion of Esr1 does not deplete the satellite cell compartment in males. (A) RTqPCR for Esr1 gene in the VCAM1+/Intga7+ cells from Pax7-cre- or cre+ male mice treated with tamoxifen (n = 4). (B) Total number of CD45/CD31(Lineage)-negative, VCAM1+/Itga7+-positive cells in tibialis anterior (TA), gastrocnemius (Gastroc), and soleus muscles in Esr1FL/FL, Pax7-cre +/− males. (C) Density of CD45/CD31(Lineage)-negative, VCAM1+/Itga+ cells in TA, gastrocnemius and soleus in Esr1FL/FL, Pax7-cre +/− males. Data are presented as mean ± SEM, p values calculated by t-test.
Cells 14 01606 g003
Figure 4. Male scERαKO cells expand normally after muscle injury. (A) Total number and Pax7-ZsGreen+ cells in the BaCl2 injured and contralateral TA from both Esr1FL/FL, Pax7-cre +/− males (n = 4); (B) Density of Pax7-ZsGreen+ cells in (A). Data are presented as mean ± SEM; p-values calculated by two-way ANOVA.
Figure 4. Male scERαKO cells expand normally after muscle injury. (A) Total number and Pax7-ZsGreen+ cells in the BaCl2 injured and contralateral TA from both Esr1FL/FL, Pax7-cre +/− males (n = 4); (B) Density of Pax7-ZsGreen+ cells in (A). Data are presented as mean ± SEM; p-values calculated by two-way ANOVA.
Cells 14 01606 g004
Figure 5. ERα is required in male satellite cells for optimal engraftment into female but not male muscle. (A) Schematic for the transplantation experiment into female recipients. (B) Total number of Pax7-ZsGreen+ cells in the TA muscle of the female recipients treated with tamoxifen and maintained on tamoxifen diet for 1 month (n = 8). (C) Total number of Pax7-ZsGreen+ cells in the TA muscle of female recipients treated with sunflower oil vehicle (n = 8). Analysis was performed after 1 month. (D) Schematic for the transplantation experiment into male recipients. (E) Total number of Pax7-ZsGreen+ cells in the TA muscle of the male recipients treated with tamoxifen and maintained on tamoxifen diet for 1 month (n = 6). Data are presented as mean ± SEM; p-values calculated by t-test.
Figure 5. ERα is required in male satellite cells for optimal engraftment into female but not male muscle. (A) Schematic for the transplantation experiment into female recipients. (B) Total number of Pax7-ZsGreen+ cells in the TA muscle of the female recipients treated with tamoxifen and maintained on tamoxifen diet for 1 month (n = 8). (C) Total number of Pax7-ZsGreen+ cells in the TA muscle of female recipients treated with sunflower oil vehicle (n = 8). Analysis was performed after 1 month. (D) Schematic for the transplantation experiment into male recipients. (E) Total number of Pax7-ZsGreen+ cells in the TA muscle of the male recipients treated with tamoxifen and maintained on tamoxifen diet for 1 month (n = 6). Data are presented as mean ± SEM; p-values calculated by t-test.
Cells 14 01606 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shams, A.S.; Sullivan, B.P.; Toso, E.A.; Lowe, D.A.; Kyba, M. Estrogen Receptor Regulates Male Satellite Cells in a Female, but Not Male, Environment. Cells 2025, 14, 1606. https://doi.org/10.3390/cells14201606

AMA Style

Shams AS, Sullivan BP, Toso EA, Lowe DA, Kyba M. Estrogen Receptor Regulates Male Satellite Cells in a Female, but Not Male, Environment. Cells. 2025; 14(20):1606. https://doi.org/10.3390/cells14201606

Chicago/Turabian Style

Shams, Ahmed S., Brian P. Sullivan, Erik A. Toso, Dawn A. Lowe, and Michael Kyba. 2025. "Estrogen Receptor Regulates Male Satellite Cells in a Female, but Not Male, Environment" Cells 14, no. 20: 1606. https://doi.org/10.3390/cells14201606

APA Style

Shams, A. S., Sullivan, B. P., Toso, E. A., Lowe, D. A., & Kyba, M. (2025). Estrogen Receptor Regulates Male Satellite Cells in a Female, but Not Male, Environment. Cells, 14(20), 1606. https://doi.org/10.3390/cells14201606

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop