Abstract
A thin endometrium can lead to low clinical pregnancy rates, low live birth rates, high spontaneous abortion rates, and low birth weight. However, current methods of treating thin endometria do not achieve ideal results. This study explored the effect of Indian Hedgehog (IHH) on thin endometrium and its mechanism of action. A thin endometrial rat model was established by infusion of 95% ethanol. IHH was overexpressed in model rats using adeno-associated viruses. The endometrial thickness and number of glands and vessels were determined using H&E staining. Endometrial fibrosis was detected using Masson’s trichrome staining. Immunohistochemistry was performed to detect α-SMA, MUC-1, and CK19. After modeling, the rats were mated, and the number of gestational sacs was counted for fertility assessment. Western blotting was used to detect the angiogenesis markers vWF, PCNA, and vim and Hedgehog signaling-related proteins SMO, GLI1, and GLI3. IHH overexpression reduced ethanol-induced edema and bruising, repaired the appearance of damaged tissue, increased endometrial thickness, promoted glandular and vascular regeneration, and alleviated endometrial fibrosis. IHH overexpression inhibited the expression of fibroblast marker α-SMA while promoting the expression of vWF, PCNA, vim, CK19, and MUC-1. It also increased the number of gestational sacs and promoted the expression of SMO, GLI1, and GLI3. In conclusion, IHH ameliorates ethanol-induced thin endometrium and improves fertility by activating the Hedgehog signaling pathway.
Keywords: Fertility, Hedgehog signaling pathway, Indian Hedgehog (IHH), Thin endometrium
The endometrium (uterine lining) plays a pivotal role in reproduction as it prepares for embryo implantation and maintains pregnancy if implantation occurs [1]. In contrast, the endometrium sheds every month (menstruation) in the absence of pregnancy and is rapidly rebuilt without scarring, presenting a powerful regeneration model [2]. A thin endometrium is a condition where the endometrium measures ≤ 7 mm thick, which can arise from inappropriate endometrial repair after curettage, defecting endometrial progenitor cell function, medical interventions, and infections that damage the endometrial basal layer [3, 4]. A thin endometrium is associated with lower rates of implantation and higher rates of miscarriage [5]. It is one of the most common causes of implantation failure in in vitro fertilization cycles [6]. Multiple medications and immunomodulatory or pro-inflammatory methods have been investigated and employed to treat thin endometrium; however, an optimal therapy has not yet been established [7]. The challenge of treating a thin endometrium is partially due to the rudimentary understanding of the pathogenesis of this condition.
A thin endometrium is characterized by slow glandular epithelial growth, high uterine arterial resistance, and low vascular endothelial growth factor (VEGF) levels [8]. Hedgehog signaling, a fundamental regulator of embryonic organogenesis, tissue regeneration, and cell proliferation/differentiation [9], modulates various aspects of endometrial homeostasis, such as epithelial-stromal crosstalk, epithelial cell proliferation, and stem cell differentiation and migration [10,11,12]. Hedgehog signaling contributes to the formation of vascular vessels in the uterus [13]. Therefore, Hedgehog signaling may be manipulated to improve endometrial repair.
Hedgehog signaling is initiated upon binding of the extracellular ligand Sonic, Desert, or Indian Hedgehog (SHH, DHH, IHH) to the transmembrane receptor Patched (PTCH), followed by the translocation of Smoothened (SMO) into the primary cilium and activation of glioma-associated oncogene (GLI) transcription factors [14]. IHH signaling is suppressed in the endometrium of patients with endometriosis or adenomyosis [15]. Epithelial IHH stimulates the production of stromal SHH, which in turn activates the PTCH-SMO-GLI pathway to promote endometrial decidualization [10]. These findings suggest that IHH is essential for maintaining normal endometrial function. However, the relationship between IHH and endometrial thickness remains unclear.
We hypothesized that IHH facilitates endometrial repair through the Hedgehog signaling pathway, providing a novel target for the treatment of thin endometria.
Materials and Methods
Preparation of a thin endometrium rat model
Healthy female Sprague Dawley rats (200–250 g, 6–8 weeks old) were supplied by Hunan SJA Laboratory Animal Co., Ltd. (Changsha, Hunan, China). After 1 week of adaptation, rats undergoing an estrus cycle for 4 consecutive days were used.
All animal experiments were approved by the Animal Ethics Committee of Hunan Provincial Maternal and Child Health Care Hospital.
Forty female rats were randomly divided into the sham, model, model + adeno-associated virus (AAV)-negative control (NC), and model + AAV-IHH groups (the AAVs were from GeneChem, Shanghai, China). The rats were anesthetized by intraperitoneal injection of 1% sodium pentobarbital (40 mg/kg), followed by uterine exposure. The uterine horns were isolated, and the ovarian and vaginal ends of the uterus were ligated. Subsequently, 95% ethanol (C0691520092, Nanjing Reagent, Nanjing, Jiangsu, China) was slowly injected into each uterine horn until the uterus was full. After 5 min, ethanol was aspirated using a syringe, and the uterine cavity was flushed with saline. The abdomen was closed and the rats were left in a warm place to wake up. Rats in the model + AAV-NC and model + AAV-IHH groups were injected with 10 nmol of AAV-NC and AAV-IHH, respectively, into the bilateral uterine horns before suturing. Rats in the sham group were injected with saline (100017414749, Cofoe, Changsha, Hunan, China). Five rats in each group were euthanized with 70% vol/min CO2 for uterine tissue collection after three sexual cycles.
Hematoxylin-eosin (H&E) staining and Masson staining
The rats were sacrificed 14 days after modeling. Collected endometrial tissue was fixed with 4% paraformaldehyde for 24 h, dehydrated, embedded in paraffin, and then sliced (10 μm thick) for conventional H&E (C0105S, Beyotime, Shanghai, China) and Masson (C0189S, Beyotime) staining. Three parts of each uterine specimen were randomly selected. Vertical endometrial thickness was measured at 3, 6, 9, and 12 points in each section. The average of four values for each section was calculated as the normal endometrial layer thickness. The average of the three measurements was used as the final endometrial thickness. The average number of endometrial glands and vessels in the three samples was also calculated. Data analysis was performed using the ImageJ software.
Immunohistochemistry
Dewaxed sections were immersed in sodium citrate buffer, and heat-induced antigen retrieval was performed. Endogenous peroxidase activity was blocked with 0.3% H2O2 in methanol (322415, Sigma-Aldrich, St. Louis, MO, USA), followed by blocking of nonspecific binding by incubation with 5% bovine serum albumin at 37°C for 30 min. The sections were incubated at 4°C overnight with primary antibodies (anti-IHH, 5 µg/ml, PA5-42706, Invitrogen, Waltham, MA, USA; anti-α-smooth muscle actin [α-SMA], 5 µg/ml, ab5831, Abcam, Cambridge, UK; anti-mucin-1 [MUC-1], 1:500, ab109185, Abcam; anti-cytokeratin 19 [CK19], 1:1000, ab76539, Abcam) and then at 37°C for 1 h with secondary antibodies (1:500, ab6721, Abcam). After 2-s differentiation with hydrochloric acid and alcohol, the sections were dehydrated, cleared, and mounted for observation.
Fertility assessment
Five rats in each group were mated 14 days after modeling at a female-to-male ratio of 3:1. Vaginal plugs were observed on the morning of the second day after mating. If a vaginal plug was found, the day was recorded as GD0. If there was no vaginal plug, a vaginal smear was performed, and the day was recorded as GD0 when the sperms were detected. Fourteen days after mating, female rats were euthanized to examine the uterine horns. Successful pregnancy depended on the presence of embryos in one or both uterine horns. This was included in the fertility analysis. The number of gestational sacs on the right and left sides was averaged.
RT-qPCR
The tissue was lysed in 1 ml of TRIzol (Thermo Fisher Scientific, Waltham, MA, USA) to extract total RNA, which was then reverse-transcribed using M-MLV reverse transcriptase (2641A, Takara, Dalian, China) and random primers (3801, Takara) to obtain cDNA. PCR mixtures were prepared and reaction conditions were set according to the instructions of the Premix Ex Taq™ kit (RR390A, Takara). PCR was performed using an ABI7500 qPCR instrument (Applied Biosystems, Shanghai, China), with β-actin as the mRNA internal reference. Data were analyzed using the 2-ΔΔCt method: ΔΔCt = (Cttarget gene – Ctreference gene)experimental group – (Cttarget gene – Ctreference gene)control group. The primer sequences are listed in Table 1.
Table 1. Primer sequences used in the study.
Gene symbol | Primer sequence |
---|---|
IHH-F | TGTTGGTCATGGATGGGGTG |
IHH-R | ACTTGGGCTCTAGTGGTCTCA |
β-actin-F | GTGGCTGGCTCAGAAAAAGG |
β-actin-R | GGGGAGATTCAGTGTGGTGG |
F, forward primer; R, reverse primer.
Western blotting
The total tissue protein was quantified using a BCA kit (23227, Thermo Fisher Scientific). Membranes loaded with gel-separated proteins were first treated with 5% (w/v) powdered milk in PBS (10010023, Gibco, Waltham, MA, USA) for 1 h at 20–25°C and then incubated overnight at 4°C with primary antibodies (anti-IHH, 1.25 µg/ml, anti-GLI1, 1:1000, ab134906, Abcam; anti-GLI3, 1:400, ab181130, Abcam; anti-SMO, 1:1000, pa5-76145, Invitrogen; anti-proliferating cell nuclear antigen [PCNA], 1:3000, ab29, Abcam; anti-vimentin [vim], 1:20000, ab92547, Abcam; anti-von Willebrand factor [vWF], 1:1000, ab287962, Abcam; anti-β-actin, 1:1000, ab8226, Abcam). After the membranes were washed, they were incubated with secondary antibodies (1:5000, ab6721, Abcam) at room temperature for 1 h. Images were captured using a BioSpectrum imaging system (UVP, Upland, CA, USA).
Statistical analysis
GraphPad Prism 8.0 was applied to the statistical analysis. All quantitative data are presented as mean ± SEM. The Shapiro–Wilk test was adopted to test the normality of data. Parameters of ≥ 3 groups were compared using a one-way analysis of variance followed by the Bonferroni test, while differences between 2 groups were analyzed using Student’s t-test. Statistical significance was denoted by P < 0.05.
Results
IHH promotes morphological and histological changes in uterine repair
The expression of IHH in the uterine tissues of rats in each group was detected using RT-qPCR, western blotting, and immunohistochemistry. The model group showed reduced IHH expression relative to the sham group; compared with the model + AAV-NC group, the model + AAV-IHH group showed increased IHH expression, indicating that IHH was successfully overexpressed (Fig. 1A–C, P < 0.05). The uterine tissue in the sham group was normal in appearance, red, and elastic. However, the uterine tissue in the model group showed obstruction, stenosis, and fluid accumulation. The edema and bruises caused by ethanol in rats in the model + AAV-IHH group generally subsided, and their uteri turned rosy and shiny (Fig. 1D), indicating that IHH could repair the morphology and appearance of damaged uterine tissue. H&E staining was then performed to detect endometrial changes. The endometrium of the model rats was thinned. The model + AAV-IHH group showed increased endometrial thickness (Fig. 1E) and an increased number of glands and vessels (Fig. 1F–G, P < 0.05). Based on the above experimental results, IHH can increase endometrial thickness and promote glandular and vascular regeneration.
Fig. 1.
IHH promotes tissue repair in rats with thin endometrium. (A–B) RT-qPCR (A) and western blotting (B) detection of IHH mRNA and protein expression; (C) Immunohistochemical detection of IHH expression; (D) Observation of uterine morphology; (E) H&E staining to detect endometrial histological features and changes in endothelial thickness; (F) Number of endometrial glands; (G) Number of endometrial vessels. N = 5; ** P < 0.01, compared with the sham group; ## P < 0.01, compared with the model group.
IHH reduces fibrosis in thin endometrium
After Masson’s trichrome staining, the fibers in the endometrial tissue were stained blue. The model rats exhibited increased endometrial fibrosis. Endometrial fibrosis in the model rats was alleviated by AAV-IHH treatment (Fig. 2A, P < 0.05). α-SMA is a marker for myofibroblasts and an indicator of fibrosis. Using immunohistochemistry, we detected α-SMA expression in the endometrium of the model rats. However, no significant α-SMA expression was observed in the endometrium of rats in the model + AAV-IHH group (Fig. 2B). This indicates that IHH can slow the development of fibrosis in the thin endometrium.
Fig. 2.
IHH reduces fibrosis in thin endometrium. (A) Masson staining of endometrial tissue (40 ×, 200 ×); (B) Immunohistochemistry to detect endometrial α-SMA (100 ×, 200 ×). N = 5; ** P < 0.01, compared with sham group; ## P < 0.01, compared with model group.
IHH promotes endometrial cell proliferation and angiogenesis
This study found that the collagen content of damaged endometrial tissues increased. Moreover, H&E staining revealed fluctuations in the number of cells and vessels. Western blotting was used to detect the angiogenic protein marker vWF and the cell proliferation markers PCNA and vim to evaluate injury-induced cellular and vascular changes in the endometrium. The levels of vWF, PCNA, and vim in the endometrium of the model group were lower than those in the sham group. The model + AAV-IHH group exhibited increased expression of vWF, PCNA, and vim compared with the model + AAV-NC group (Fig. 3A, P < 0.05). Immunohistochemical staining of the endometrial tissue further showed that the model group expressed CK19 and MUC-1 at lower levels than the sham group. CK19 and MUC-1 levels were elevated by AAV-IHH treatment in model rats (Fig. 3B–C). These data demonstrate that IHH can promote endothelial cell proliferation and angiogenesis.
Fig. 3.
IHH promotes endothelial cell proliferation and angiogenic marker expression. (A) Western blotting to detect the expression of vWF, PCNA, and vim proteins; (B–C) Immunohistochemical staining to detect the expression of CK19 (B) and MUC-1 (C) in endometrial glandular epithelial cells. N = 5; * P < 0.05 and ** P < 0.01, compared with the sham group; # P < 0.05 and ## P < 0.01, compared with the model group.
IHH facilitates fertility recovery in rats with thin endometrium
To evaluate the therapeutic potential of IHH in recovering fertility in the thin endometrial rat model, rats were sacrificed 14 days after mating to examine their pregnancy results. The number of gestational sacs was lower in the model group than in the sham group. The model + AAV-IHH group showed an increase in the number of gestational sacs relative to the model + AAV-NC group (Fig. 4, P < 0.05; Table 2), indicating that IHH can enhance the fertility of rats with a thin endometrium.
Fig. 4.
Number of gestational sacs in rats.
Table 2. Reproductive results of experimental rats.
sham group (n = 5) |
Model group (n = 5) |
Model + AAV-NC group (n = 5) |
Model + AAV-IHH group (n = 5) |
|
---|---|---|---|---|
Pregnancy rate (%) | 100 | 40 ** | 40 ** | 80 ## |
Number of embryos | 10.6 ± 1.0 | 4.5 ± 0.5 ** | 5.0 ± 1.0 ** | 7.3 ± 0.8 *# |
* P < 0.05 and ** P < 0.01, compared with the sham group; # P < 0.05 and ## P < 0.01, compared with the model group.
IHH promotes endometrial repair through the Hedgehog signaling pathway
Hedgehog is a novel endogenous damage signal that can activate multiple beneficial functions in human endometrial stem cells [12]. Hedgehog signaling activation can promote endometrial hyperplasia [16]. Moreover, the PathCards database shows that IHH is involved in the regulation of Hedgehog signaling. Therefore, we speculated that IHH might promote endometrial repair by regulating Hedgehog signaling. Western blotting showed a decrease in the expression of the Hedgehog signaling-related proteins SMO, GLI1, and GLI3 in model rats. The expression of SMO, GLI1, and GLI3 in model rats increased following AAV-IHH treatment (Fig. 5, P < 0.05). Taken together, these data indicate that IHH increases endometrial thickness and reproductive capacity by regulating Hedgehog signaling activation.
Fig. 5.
IHH promotes Hedgehog signaling activation. Western blotting was used to detect the expression of the Hedgehog signaling-related proteins SMO, GLI1, and GLI3 in each group of rats. N = 5; * P < 0.05 and ** P < 0.01, compared with the sham group; # P < 0.05 and ## P < 0.01, compared with the model group.
Discussion
Endometrial thickness, an important clinical indicator, is closely related to pregnancy outcomes regardless of whether fertilization occurs in vivo or in vitro. An endometrial thickness of ≤ 7 mm (thin endometrium) can lead to low clinical pregnancy rates, high spontaneous abortion rates, and low live birth rates. Current methods for treating thin endometria have not yielded ideal results. Therefore, the pathogenic landscape of thin endometria should be explored to identify novel targets for therapeutic intervention.
This study found that AAV-IHH treatment repaired ethanol-injured uteri and enhanced fertility in rats. Notably, IHH increased endometrial thickness and the number of endometrial glands and vessels. Specifically, IHH upregulated the levels of PCNA, vWF, vim, MUC-1, and CK19 in the endometrium. PCNA is an essential factor in DNA replication and repair and is a marker of cell proliferation [17]. Given its crucial role in normal hemostasis, vWF has been widely used as a reliable marker of endothelial cell activation and injury [18]. Vim is expressed in mesenchymal cells [19], which support endothelial cells in the process of new vessel formation during tissue regeneration [20]. Therefore, IHH may stimulate vascular regeneration in the thin endometrium by promoting cell proliferation and endothelial angiogenesis. MUC-1 is a transmembrane glycoprotein expressed on the surface of all epithelial cells, forming a tight mesh that protects cells from environmental insults [21]. CK19, an epithelial cytoskeletal marker, maintains luminal epithelial integrity and guarantees the proliferation of glandular epithelial cells [22]. Thus, IHH protects epithelial cells and facilitates gland formation in the endometrium by upregulating MUC-1 and CK19. Collectively, our findings suggest that IHH improves endometrial repair by affecting various cell types.
Following injury, the endometrium develops fibrosis in cases of failed regeneration and forms scars that may interfere with normal endometrial function, leading to infertility [2]. A previous study shows that IHH released from TNF-activated renal epithelia drives local and remote organ fibrosis by activating canonical Hedgehog signaling in Gli1+ cells, a major source of activated fibroblasts in multiple organs [23]. However, this study found that AAV-IHH treatment reduced the number of fibers in the endometrium of rats with thin endometrium. The expression of α-SMA was repressed in AAV-IHH-treated model rats. α-SMA is a myofibroblast marker protein that is incorporated into cytoskeletal stress fibers during tissue healing [24]. As mentioned above, IHH restores the regenerative capacity of the injured endometrium. Consequently, the activation of α-SMA-expressing myofibroblasts and the production of fibers are prevented.
As a Hedgehog ligand, IHH promoted endometrial regeneration in rats with a thin endometrium by activating the expression of the Hedgehog signaling-related proteins SMO, GLI1, and GLI3 in this study. A previous study has shown that IHH signaling is required for cyclical uterine remodeling across the estrous cycle and controls muscle fibers in the uterus [25], which is consistent with our results. The IHH-SMO signaling pathway is a well-known downstream progesterone pathway that suppresses estrogen-dependent epithelial proliferation and stimulates stromal proliferation in the uterus [26]. IHH signaling is inhibited in progesterone resistance-related endometrial diseases such as endometriosis and adenomyosis, leading to the aberrant activation of endometrial stromal cell autophagy [15]. IHH signaling also promotes the expression of 11β-hydroxysteroid dehydrogenase 2 to control the level of uterine corticosterone in early pregnancy, providing a suitable environment for embryo implantation [27]. These findings indicate that IHH is vital for normal endometrial function and uterine homeostasis. Moreover, epithelial IHH stimulates the production of stromal SHH, another Hedgehog ligand, which in turn activates the canonical Hedgehog signaling pathway to promote endometrial decidualization, a remodeling process necessary for successful pregnancy [10]. Overall, IHH may regulate endometrial remodeling and uterine homeostasis alone, in cooperation with other Hedgehog ligands, or both.
In summary, IHH ameliorates ethanol-induced thinning of the endometrium and improves fertility by stimulating the activation of the Hedgehog signaling pathway. This study is the first to elucidate the role of IHH signaling in thin endometrium. IHH signaling may be augmented to improve endometrial repair and enhance fertility in patients with a thin endometrium. Future studies should investigate the mechanisms regulating IHH expression in the thin endometrium to reinforce these findings.
Conflict of interests
The authors declare that they have no competing interests.
Data availability
The datasets used or analyzed during the current study are available from the corresponding author on reasonable request.
References
- 1.Critchley HOD, Maybin JA, Armstrong GM, Williams ARW. Physiology of the endometrium and regulation of menstruation. Physiol Rev 2020; 100: 1149–1179. [DOI] [PubMed] [Google Scholar]
- 2.Ang CJ, Skokan TD, McKinley KL. Mechanisms of Regeneration and Fibrosis in the Endometrium. Annu Rev Cell Dev Biol 2023; 39: 197–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Lv H, Zhao G, Jiang P, Wang H, Wang Z, Yao S, Zhou Z, Wang L, Liu D, Deng W, Dai J, Hu Y. Deciphering the endometrial niche of human thin endometrium at single-cell resolution. Proc Natl Acad Sci USA 2022; 119: 119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lin J, Wang Z, Huang J, Tang S, Saiding Q, Zhu Q, Cui W. Microenvironment-protected exosome-hydrogel for facilitating endometrial regeneration, fertility restoration, and live birth of offspring. Small 2021; 17: e2007235. [DOI] [PubMed] [Google Scholar]
- 5.Gharibeh N, Aghebati-Maleki L, Madani J, Pourakbari R, Yousefi M, Ahmadian Heris J. Cell-based therapy in thin endometrium and Asherman syndrome. Stem Cell Res Ther 2022; 13: 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Vartanyan E, Tsaturova K, Devyatova E. Thin endometrium problem in IVF programs. Gynecol Endocrinol 2020; 36(sup1): 24–27. [DOI] [PubMed] [Google Scholar]
- 7.Saad-Naguib MH, Kenfack Y, Sherman LS, Chafitz OB, Morelli SS. Impaired receptivity of thin endometrium: therapeutic potential of mesenchymal stem cells. Front Endocrinol (Lausanne) 2024; 14: 1268990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Zhang X, Li Y, Chen X, Jin B, Shu C, Ni W, Jiang Y, Zhang J, Ma L, Shu J. Single-cell transcriptome analysis uncovers the molecular and cellular characteristics of thin endometrium. FASEB J 2022; 36: e22193. [DOI] [PubMed] [Google Scholar]
- 9.Chen J, Sun Y, Chi Z. Regulation of hematopoiesis by hedgehog signaling (Review). Mol Med Rep 2023; 27: 27. [DOI] [PubMed] [Google Scholar]
- 10.Li B, Yan YP, He YY, Liang C, Li MY, Wang Y, Yang ZM. IHH, SHH, and primary cilia mediate epithelial-stromal cross-talk during decidualization in mice. Sci Signal 2023; 16: eadd0645. [DOI] [PubMed] [Google Scholar]
- 11.Zhou X, Kang Y, Chang Y, Xia S, Wu M, Liu J, Dong D, Zhang W, Chen H, Li H. CRC therapy identifies indian hedgehog signaling in mouse endometrial epithelial cells and inhibition of Ihh-KLF9 as a novel strategy for treating IUA. Cells 2022; 11: 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Park SR, Kim SR, Park CH, Lim S, Ha SY, Hong IS, Lee HY. Sonic Hedgehog, a novel endogenous damage signal, activates multiple beneficial functions of human endometrial stem cells. Mol Ther 2020; 28: 452–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Haraguchi R, Yamada G, Murashima A, Matsumaru D, Kitazawa R, Kitazawa S. New Insights into Development of Female Reproductive Tract-Hedgehog-Signal Response in Wolffian Tissues Directly Contributes to Uterus Development. Int J Mol Sci 2021; 22: 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hanna J, Beke F, O’Brien LM, Kapeni C, Chen HC, Carbonaro V, Kim AB, Kishore K, Adolph TE, Skjoedt MO, Skjoedt K, de la Roche M, de la Roche M. Cell-autonomous Hedgehog signaling controls Th17 polarization and pathogenicity. Nat Commun 2022; 13: 4075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zhou Y, Peng Y, Xia Q, Yan D, Zhang H, Zhang L, Chen Y, Zhao X, Li J. Decreased Indian hedgehog signaling activates autophagy in endometriosis and adenomyosis. Reproduction 2021; 161: 99–109. [DOI] [PubMed] [Google Scholar]
- 16.Kim KH, Kim JM, Choi YL, Shin YK, Lee HC, Seong IO, Kim BK, Chae SW, Chung YS, Kim SH. Expression of sonic hedgehog signaling molecules in normal, hyperplastic and carcinomatous endometrium. Pathol Int 2009; 59: 279–287. [DOI] [PubMed] [Google Scholar]
- 17.González-Magaña A, Blanco FJ. Human PCNA structure, function and interactions. Biomolecules 2020; 10: 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zeineddin A, Dong JF, Wu F, Terse P, Kozar RA. Role of von Willebrand factor after injury: it may do more than we think. Shock 2021; 55: 717–722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kuburich NA, den Hollander P, Pietz JT, Mani SA. Vimentin and cytokeratin: Good alone, bad together. Semin Cancer Biol 2022; 86: 816–826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Summer S, Rossmanith E, Pasztorek M, Fiedler C, Gröger M, Rauscher S, Weber V, Fischer MB. Mesenchymal stem cells support human vascular endothelial cells to form vascular sprouts in human platelet lysate-based matrices. PLoS One 2022; 17: e0278895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Chen W, Zhang Z, Zhang S, Zhu P, Ko JK, Yung KK. MUC1: Structure, function, and clinic application in epithelial cancers. Int J Mol Sci 2021; 22: 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Li T, Chan RWS, Li RHW, Ng EHY, Zhang S, Yeung WSB. Endometrial mesenchymal stromal/stem cells improve regeneration of injured endometrium in mice. Biol Res 2024; 57: 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.O’Sullivan ED, Mylonas KJ, Xin C, Baird DP, Carvalho C, Docherty MH, Campbell R, Matchett KP, Waddell SH, Walker AD, Gallagher KM, Jia S, Leung S, Laird A, Wilflingseder J, Willi M, Reck M, Finnie S, Pisco A, Gordon-Keylock S, Medvinsky A, Boulter L, Henderson NC, Kirschner K, Chandra T, Conway BR, Hughes J, Denby L, Bonventre JV, Ferenbach DA. Indian Hedgehog release from TNF-activated renal epithelia drives local and remote organ fibrosis. Sci Transl Med 2023; 15: eabn0736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Venugopal H, Hanna A, Humeres C, Frangogiannis NG. Properties and functions of fibroblasts and myofibroblasts in myocardial infarction. Cells 2022; 11: 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Roberson EC, Tran NK, Godambe AN, Mark H, Nguimtsop M, Rust T, Ung E, Barker LJ, Fitch RD, Wallingford JB. Hedgehog signaling is required for endometrial remodeling and myometrial homeostasis in the cycling mouse uterus. iScience 2023; 26: 107993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yang SC, Park M, Hong KH, La H, Park C, Wang P, Li G, Chen Q, Choi Y, DeMayo FJ, Lydon JP, Skalnik DG, Lim HJ, Hong SH, Park SH, Kim YS, Kim HR, Song H. CFP1 governs uterine epigenetic landscapes to intervene in progesterone responses for uterine physiology and suppression of endometriosis. Nat Commun 2023; 14: 3220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zheng HT, Fu T, Zhang HY, Yang ZS, Zheng ZH, Yang ZM. Progesterone-regulated Hsd11b2 as a barrier to balance mouse uterine corticosterone. J Endocrinol 2020; 244: 177–187. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The datasets used or analyzed during the current study are available from the corresponding author on reasonable request.