Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters

Publication year range
1.
J Assist Reprod Genet ; 40(5): 1163-1171, 2023 May.
Article in English | MEDLINE | ID: mdl-36662355

ABSTRACT

PURPOSE: The purpose was to evaluate the effect of intrauterine injection of aBMNC on the endometrial function in patients with refractory Asherman's syndrome (AS) and/or thin and dysfunctional endometrium (TE). STUDY DESIGN: This is a prospective, experimental, non-controlled study MATERIAL AND METHODS: The study was carried out between December 2018 and December 2020 on 20 patients, who were of age < 45 years and had oligo/amenorrhea and primary infertility due to refractory AS and/or TE. One hundred ml BM was extracted. aBMNC cells were separated according to generic volume reduction protocol by using the Cell Separation System SEPAX S-100 table top centrifuge system. We have evaluated CD34+, mononuclear cell (MNC), and total nucleated cell (TNC) counts. The transplantation aBMNC was performed by two intrauterine injections at an interval of one week, transvaginally into the endometrial-myometrial junction by an ovum aspiration needle. Midcyclic endometrial thickness (ET) and gestations after transplantation were evaluated. RESULTS: The mean TNC, MNC, and CD34+ cells were 11.55 ± 4.7 × 108, 3.85 ± 2.01 × 108, and 7.00 ± 2.88 × 106 at first injection, respectively, and 6.85 ± 2.67 × 108, 2.04 ± 1.11 × 108, and 3.44 ± 1.31 × 106 at second injection, respectively. The maximum posttransplantation ET was significantly higher than the maximum pretransplantation ET: 2.97 ± 0.48 vs. 5.76 ± 1.19 (mean ± standard deviation, p < 0.01). Twelve patients had frozen-thaw embryo transfers after the study. In 42% (n = 5 of 12) of the patients, pregnancy was achieved. One of the five patients delivered a healthy baby at term. CONCLUSIONS: Autologous BMNC transplantation may contribute to endometrial function in patients with AS and/or TE.


Subject(s)
Gynatresia , Pregnancy , Female , Humans , Middle Aged , Prospective Studies , Gynatresia/therapy , Bone Marrow , Endometrium , Stem Cell Transplantation/methods
2.
Small ; 18(31): e2201225, 2022 08.
Article in English | MEDLINE | ID: mdl-35798313

ABSTRACT

Asherman's syndrome (AS) occurs as a consequence of severe damage to the endometrial basalis, usually leading to menstrual abnormalities, infertility, and recurrent miscarriage in women. Currently, human endometrium-derived adventitial cells (En-ADVs) are considered ideal seed cells with high pluripotency for regenerative medicine. However, critical issues such as noninvasive repair of tissues, targeting of native stem cells, and continuous action in the injured sites are not well resolved. Herein, En-ADV spheroid-loaded hierarchical microneedles (MN/En-ADV) for in situ intrauterine repair are developed. The flexible microneedles are fabricated with gelatin methacryloyl and lactoferrin, imparting the characteristics of rapid degradation and antimicrobial activity. Benefiting from an array of microwells on microneedles, En-ADVs can rapidly form 3D cell spheroids, which display higher potential for cell proliferation, differentiation, and migration than dissociated cells. With the application of MN/En-ADV, the repaired uteri show well-defined myometrial regeneration, angiogenesis, and an increase of endometrial receptivity in a rat AS model. Notably, embryos are able to implant in the reconstructed sites and remain viable, indicating that this system promotes the restoration of both normal morphology and reproductive function in the injured uterus. It is anticipated that multifunctional MN/En-ADV can be an ideal candidate for versatile in situ tissue regeneration.


Subject(s)
Anti-Infective Agents , Gynatresia , Regeneration , Uterus , Animals , Anti-Infective Agents/pharmacology , Endometrium/growth & development , Female , Gelatin , Gynatresia/therapy , Humans , Methacrylates , Rats , Uterus/growth & development
3.
J Clin Ultrasound ; 50(9): 1320-1321, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35657771

ABSTRACT

Although 10% of pregnancies following treatment of Asherman's syndrome are estimated to have abnormal placental adhesion, there is a paucity of reports describing imaging features in such cases. We describe ultrasound and MRI features in one of such cases, showing a peculiar pattern of shallow but diffuse abnormally adherent placenta.


Subject(s)
Gynatresia , Placenta Accreta , Placenta Diseases , Female , Humans , Pregnancy , Gynatresia/therapy , Hysteroscopy/methods , Retrospective Studies , Placenta/diagnostic imaging , Placenta Diseases/diagnostic imaging , Magnetic Resonance Imaging , Placenta Accreta/diagnostic imaging
4.
Mol Reprod Dev ; 88(8): 527-543, 2021 08.
Article in English | MEDLINE | ID: mdl-34293229

ABSTRACT

Endometrial function is essential for embryo implantation and pregnancy, but managing endometrial thickness that is too thin to support pregnancy or an endometrium of compromised functionality due to intrauterine adhesions is an ongoing challenge in reproductive medicine. Here, we review current and emerging therapeutic and experimental options for endometrial regeneration with a focus on animal models used to study solutions for Asherman's syndrome and endometrial atrophy, which both involve a damaged endometrium. A review of existing literature was performed that confirmed the lack of consensus on endometrial therapeutic options, though promising new alternatives have emerged in recent years (platelet-rich plasma, exosomes derived from stem cells, bioengineering-based techniques, endometrial organoids, among others). In the future, basic research using established experimental models of endometrial pathologies (combined with new high-tech solutions) and human clinical trials with large population sizes are needed to evaluate these emerging and new endometrial therapies.


Subject(s)
Endometrium/pathology , Gynatresia/therapy , Animals , Disease Models, Animal , Female , Gynatresia/pathology , Humans , Platelet-Rich Plasma , Stem Cell Transplantation
5.
Mol Reprod Dev ; 88(6): 379-394, 2021 06.
Article in English | MEDLINE | ID: mdl-34014590

ABSTRACT

Endometrial damage is an important cause of female reproductive problems, manifested as menstrual abnormalities, infertility, recurrent pregnancy loss, and other complications. These conditions are collectively termed "Asherman syndrome" (AS) and are typically associated with recurrent induced pregnancy terminations, repeated diagnostic curettage and intrauterine infections. Cancer treatment also has unexpected detrimental side effects on endometrial function in survivors independently of ovarian effects. Endometrial stem cells act in the regeneration of the endometrium and in repair through direct differentiation or paracrine effects. Nonendometrial adult stem cells, such as bone marrow-derived mesenchymal stem cells and umbilical cord-derived mesenchymal stem cells, with autologous and allogenic applications, can also repair injured endometrial tissue in animal models of AS and in human studies. However, there remains a lack of research on the repair of the damaged endometrium after the reversal of tumors, especially endometrial cancers. Here, we review the biological mechanisms of endometrial regeneration, and research progress and challenges for adult stem cell therapy for damaged endometrium, and discuss the potential applications of their use for endometrial repair after cancer remission, especially in endometrial cancers. Successful application of such cells will improve reproductive parameters in patients with AS or cancer. Significance: The endometrium is the fertile ground for embryos, but damage to the endometrium will greatly impair female fertility. Adult stem cells combined with tissue engineering scaffold materials or not have made great progress in repairing the injured endometrium due to benign lesions. However, due to the lack of research on the repair of the damaged endometrium caused by malignant tumors or tumor therapies, the safety and effectiveness of such stem cell-based therapies need to be further explored. This review focuses on the molecular insights and clinical application potential of adult stem cells in endometrial regeneration and discusses the possible challenges or difficulties that need to be overcome in stem cell-based therapies for tumor survivors. The development of adult stem cell-related new programs will help repair damaged endometrium safely and effectively and meet fertility needs in tumor survivors.


Subject(s)
Adult Stem Cells/physiology , Endometrium/physiology , Gynatresia/physiopathology , Regeneration/physiology , Abortion, Habitual/etiology , Abortion, Habitual/prevention & control , Adult Stem Cells/transplantation , Amnion/cytology , Animals , Antigens, Differentiation/analysis , Bone Marrow Cells , Cellular Senescence , Disease Models, Animal , Endometrial Neoplasms/physiopathology , Endometrial Neoplasms/therapy , Endometrium/blood supply , Endometrium/cytology , Endometrium/injuries , Female , Fetal Blood/cytology , Gynatresia/complications , Gynatresia/therapy , Humans , Hydrogels , Induced Pluripotent Stem Cells/transplantation , Infertility, Female/etiology , Infertility, Female/therapy , Menstruation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mouth Mucosa/cytology , Side-Population Cells/cytology , Stem Cell Niche , Tissue Engineering/methods , Tissue Scaffolds
6.
J Assist Reprod Genet ; 38(11): 2955-2963, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34613578

ABSTRACT

PURPOSE: Treatment of Asherman syndrome (AS) presents a significant clinical challenge. Based on our in vitro data showing that PRP could activate endometrial cell proliferation and migration, we hypothesized that intrauterine infusion of autologous platelet-rich plasma (PRP) may improve endometrial regeneration and fertility outcomes in patients with moderate-severe AS. MATERIALS AND METHODS: Subjects with moderate-severe AS were randomized to PRP or saline control administered following hysteroscopic adhesiolysis. Due to relative inability to randomize patients to the control group, after initial randomization of 10 subjects (6 in PRP and 4 in control groups), the remainder were prospectively enrolled in PRP group (n = 9), with 11 historic controls added to control group, for a total of 30 subjects (PRP n = 15; saline control n = 15). Right after hysteroscopy, 0.5-1 mL of PRP or saline was infused into the uterus via a Wallace catheter, followed by estrogen therapy. The primary outcomes were changes in endometrial thickness (EMT, checked in 3 weeks) and in menstrual flow; secondary outcomes were pregnancy and live birth rates. EMT and menstrual bleeding pattern were assessed before and after the intervention. Pregnancy was assessed over a 6-month period. RESULTS: There were no statistically significant differences in age, gravidity/parity, cause of AS, preoperative menses assessment, AS hysteroscopy score, and intrauterine balloon placement between the groups. There was no statistically significant difference (p = 0.79) in EMT pre-PRP infusion for control (5.7 mm, 4.0-6.0) and study arm (5.3 mm, 4.9-6.0). There was no statistically significant change (p = 0.78) in EMT after PRP infusion (1.4 mm, - 0.5-2.4) vs saline (1.0 mm, 0.0-2.5). Patients tolerated the procedure well, with no adverse effects. There was no difference in the predicted likelihood of pregnancy (p = 0.45) between the control (0.67, 0.41-0.85) and study arm (0.53, 0.29-0.76). CONCLUSIONS: PRP was well accepted and tolerated in AS patients. However, we did not observe any significant EMT increase or improved pregnancy rates after adding PRP infusion, compared to standard treatment only. The use of intrauterine PRP infusion may be a feasible option, and its potential use must be tested on a larger sample size of AS patients.


Subject(s)
Embryo Implantation , Embryo Transfer , Fertilization in Vitro/methods , Gynatresia/therapy , Live Birth/epidemiology , Platelet-Rich Plasma/cytology , Severity of Illness Index , Adult , Birth Rate , California/epidemiology , Case-Control Studies , Female , Follow-Up Studies , Gynatresia/pathology , Humans , Hysteroscopy , Menstruation , Pilot Projects , Pregnancy , Pregnancy Rate , Prognosis , Prospective Studies , Single-Blind Method , Transplantation, Autologous
7.
BJOG ; 127(5): 551-560, 2020 04.
Article in English | MEDLINE | ID: mdl-31876085

ABSTRACT

OBJECTIVE: Determining genetic and paracrine mechanisms behind endometrial regeneration in Asherman's syndrome and endometrial atrophy (AS/EA) patients after autologous CD133+ bone marrow-derived stem cell (CD133+ BMDSC) transplantation. DESIGN: Retrospective study using human endometrial biopsies and mouse models. SETTING: Fundación-IVI, IIS-La Fe, Valencia, Spain. SAMPLES: Endometrial biopsies collected before and after CD133+ BMDSC therapy, from eight women with AS/EA (NCT02144987) from the uterus of five mice with only left horns receiving CD133+ BMDSC therapy. METHODS: In human samples, haematoxylin and eosin (H&E) staining, RNA arrays, PCR validation, and neutrophil elastase (NE) immunohistochemistry (IHQ). In mouse samples, PCR validation and protein immunoarrays. MAIN OUTCOME MEASURES: H&E microscopic evaluation, RNA expression levels, PCR, and growth/angiogenic factors quantification, NE IHQ signal. RESULTS: Treatment improved endometrial morphology and thickness for all patients. In human samples, Jun, Serpine1, and Il4 were up-regulated whereas Ccnd1 and Cxcl8 were down-regulated after treatment. The significant decrease of NE signal corroborated Cxcl8 expression. Animal model analysis confirmed human results and revealed a higher expression of pro-angiogenic cytokines (IL18, HGF, MCP-1, MIP2) in treated uterine horns. CONCLUSIONS: CD133+ BMDSC seems to activate several factors through a paracrine mechanism to help tissue regeneration, modifying endometrial behaviour through an immunomodulatory milieu that precedes proliferation and angiogenic processes. Insight into these processes could bring us one step closer to a non-invasive treatment for AS/EA patients. TWEETABLE ABSTRACT: CD133+ BMDSC therapy regenerates endometrium, modifying the immunological milieu that precedes proliferation and angiogenesis.


Subject(s)
Atrophy/therapy , Endometrium/pathology , Endometrium/physiology , Gynatresia/therapy , Regeneration , Stem Cell Transplantation , AC133 Antigen/metabolism , Animals , Cyclin D1/metabolism , Cytokines/metabolism , Down-Regulation , Female , Humans , Interleukin-8/metabolism , Leukocyte Elastase/metabolism , Models, Animal , Plasminogen Activator Inhibitor 1/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Retrospective Studies , Transplantation, Autologous , Up-Regulation , Uterus/metabolism
8.
Mol Biol Rep ; 47(6): 4541-4552, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32444974

ABSTRACT

Asherman syndrome (AS) occurs due to fibrosis or uterine adhesions as a result of damage to the basal layer of the endometrium. The aim of this study is investigating the effects of adipose tissue-derived mesenchymal stem cell (ADMSC) application on the expression of vascular endothelial growth factor (VEGF), insulin-like growth factor (IGF-1), miRNA-98, miRNA199a in endometrial tissue in rats with AS. Study groups were designed as, control (C), Asherman syndrome (AS), AS + oral estrogen (ASO), AS + ADMSC (ASSC), AS + oral estrogen + ADMSC (ASSCO) with 7 samples in each group. Characterization and differentiation experiments were performed in ADMSC obtained. Two weeks after the development of the AS, ADMSC therapy was applied. BrdU (5-bromo-2'-deoxyuridine) labeling was performed to show the presence of ADMSC in the tissues. Rats were sacrificed after 8 weeks and bilateral uterine horn resection was performed. Tissues were fixed in formaldehyde. After routine tissue follow-up, sections were taken and evaluated with hematoxylin eosin staining. VEGF1 and IGF1 expressions were evaluated by immunohistochemical staining and western blot analysis. Expression changes of miR-98 and miR-199a were detected by RT-PCR. Our results showed that stem cells and estrogen giving together reduced inflammation and fibrosis in the endometrium. Immunohistochemistry and western blot results suggested that this effect was achieved especially through IGF-1. In our study, decreased miR-98 and miR-199a expressions were determined in Asherman syndrome. Furthermore, no changes of miRNA expressions were observed in treatment groups.


Subject(s)
Endometrium/metabolism , Gynatresia/therapy , Mesenchymal Stem Cells/metabolism , Adipose Tissue/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Endometrium/drug effects , Estrogens/pharmacology , Female , Fibrosis/metabolism , Gynatresia/metabolism , Insulin-Like Growth Factor I/metabolism , Mesenchymal Stem Cell Transplantation/methods , MicroRNAs/genetics , MicroRNAs/metabolism , Rats , Rats, Wistar , Vascular Endothelial Growth Factor A/metabolism
9.
J Assist Reprod Genet ; 37(8): 1861-1868, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32535814

ABSTRACT

PURPOSE: Cell therapy is a promising strategy for the treatment of Asherman's syndrome (AS), but the origin of these cells and injection route influence the therapeutic effect and complications of cell therapy. Herein, we compared the effects of systemic or local intrauterine injection of bone marrow or adipose-derived mesenchymal stem cells (BMSCs/AMSCs) on the endometrium in a rat model of AS. METHODS: After induction of AS in adult Wistar rats, the CM-Dil-positive BMSCs or AMSCs were injected either locally or intravenously. After 3 weeks, endometrial thickness, collagen deposition, cell migration, and VEGF expression were evaluated using histochemistry/immunofluorescence studies. RESULTS: In all stem cell-treated groups, an ameliorative effect on the damaged endometrium was noted. Collagen deposition diminished in both groups (IV and local injection) compared to the AS model. In rats injected locally with MSC, fibrosis decreased compared to the other groups. Moreover, endometrial thickness increased in the groups that received local injection of BMSCs and AMSCs more than the IV-transplanted AMSCs group. Immunofluorescent staining demonstrated that although the systemic transplantation of BMSCs was more effective than the other groups on VEGF expression, it led to the lowest number of CM-Dil+ stem cells in the damaged endometrium. CONCLUSION: Stem cell transplantation may reconstruct the damaged endometrium, but it is recommended to select the most effective stem cells and injection route. Because the removal of the fibrosis and the replacement of the epithelia cells is an effective therapeutic strategy for AS, in this study, we conclude that the local injection of AMSCs is more appropriate than BMSCs to treat AS.


Subject(s)
Cell- and Tissue-Based Therapy , Gynatresia/therapy , Mesenchymal Stem Cell Transplantation , Vascular Endothelial Growth Factor A/genetics , Adipose Tissue/cytology , Adipose Tissue/transplantation , Animals , Bone Marrow Cells/cytology , Disease Models, Animal , Female , Gene Expression Regulation, Developmental/genetics , Gynatresia/genetics , Gynatresia/pathology , Humans , Mesenchymal Stem Cells/cytology , Rats , Regenerative Medicine
10.
J Assist Reprod Genet ; 37(7): 1511-1529, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32445154

ABSTRACT

The current treatment for Asherman syndrome is limited and not very effective. The aim of this review is to summarize the most recent evidence for stem cells in the treatment of Asherman syndrome. The advent of stem cell therapy has propagated experimentation on mice and humans as a novel treatment. The consensus is that the regenerative capacity of stem cells has demonstrated improved outcomes in terms of fertility and fibrosis in both mice and humans with Asherman syndrome. Stem cells have effects on tissue repair by homing to the injured site, recruiting other cells by secreting chemokines, modulating the immune system, differentiating into other types of cells, proliferating into daughter cells, and potentially having antimicrobial activity. The studies reviewed examine different origins and administration modalities of stem cells. In preclinical models, therapeutic systemic injection of stem cells is more effective than direct intrauterine injection in regenerating the endometrium. In conjunction, bone marrow-derived stem cells have a stronger effect on uterine regeneration than uterine-derived stem cells, likely due to their broader differentiation potency. Clinical trials have demonstrated the initial safety and effectiveness profiles of menstrual, bone marrow, umbilical cord, and adipose tissue-derived stem cells in resumption of menstruation, fertility outcomes, and endometrial regeneration.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Gynatresia/therapy , Amniotic Fluid/cytology , Animals , Biomarkers/metabolism , Endometrium/pathology , Endometrium/physiology , Epithelial-Mesenchymal Transition , Female , Gynatresia/pathology , Humans , Menstruation/blood , Mesenchymal Stem Cells/physiology , Placenta/cytology , Pregnancy , Regeneration , Stem Cell Transplantation , Umbilical Cord/cytology , Uterus/cytology
11.
Acta Obstet Gynecol Scand ; 98(5): 672-677, 2019 05.
Article in English | MEDLINE | ID: mdl-30815850

ABSTRACT

An update on the current state of endometrial cell therapies in terms of cell types, mechanisms of action, delivery, safety, regulatory frameworks and future perspectives. This review focuses on clinical trials using angiogenesis-promoting therapies and stromal therapies piloted in the last 10 years for alleviating Asherman's syndrome and long-term infertility. All studies present promising preliminary results, indicating increased endometrial thickness and resumed menstruation. Further characterization of individual cell products, their mode of action and larger clinical trials will be essential to establishing cell therapy as a viable option for the treatment of infertility and fertility preservation.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Endometrium , Gynatresia , Infertility, Female , Endometrium/blood supply , Endometrium/pathology , Female , Gynatresia/complications , Gynatresia/pathology , Gynatresia/therapy , Humans , Infertility, Female/etiology , Infertility, Female/therapy , Neovascularization, Physiologic
12.
Clin Obstet Gynecol ; 62(2): 257-270, 2019 06.
Article in English | MEDLINE | ID: mdl-31021928

ABSTRACT

Uterine factor infertility (UFI) may affect up to 1 in 500 reproductive age women. The uterus is an essential component of achieving pregnancy and carrying a pregnancy to term successfully. There are many etiologies of UFI which may be categorized into either congenital or acquired causes. In this review, we discuss the different causes of UFI as well as the treatment options, which now includes uterine transplant.


Subject(s)
Infertility, Female/etiology , Adenomyosis/complications , Adenomyosis/therapy , Female , Gynatresia/complications , Gynatresia/therapy , Humans , Hysterectomy , Infertility, Female/therapy , Leiomyoma/complications , Leiomyoma/therapy , Polyps/complications , Polyps/therapy , Radiation Dosage , Surrogate Mothers , Uterine Diseases/complications , Uterine Diseases/therapy , Uterine Neoplasms/complications , Uterine Neoplasms/therapy , Uterus/abnormalities , Uterus/radiation effects , Uterus/transplantation
13.
Curr Opin Obstet Gynecol ; 29(4): 249-256, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28582327

ABSTRACT

PURPOSE OF REVIEW: Intrauterine adhesions, also known as Asherman's syndrome, can have an impact on both reproductive outcomes and gynaecologic symptoms. Understanding the cause of intrauterine adhesions and the common clinical presentation will increase awareness of the condition and guide the patient to appropriate therapy. Surgical management offers favourable fertility outcomes and is often successful in restoring menstruation. RECENT FINDINGS: Surgical management with hysteroscopic lysis of adhesions is the gold standard for treatment and adopting an office-based approach offers several advantages. Prevention of reformation of adhesions remains challenging and no single method for preventing recurrence has shown superiority. Cell-based therapies using endometrial stem/progenitor cells hold promise for future use in regenerating inadequate endometrium. SUMMARY: Increased awareness of the symptoms suggestive of intrauterine adhesive disease, as well as recognition of common causes and preceding events, is crucial for early diagnosis, patient counselling and treatment. VIDEO ABSTRACT: http://links.lww.com/COOG/A36.


Subject(s)
Gynatresia/etiology , Gynatresia/therapy , Hysteroscopy , Adult , Endometrium/cytology , Endometrium/pathology , Female , Fertility , Humans , Hysterosalpingography , Menstruation , Pregnancy , Prognosis , Recurrence , Regeneration , Stem Cells/cytology , Tissue Adhesions , Treatment Outcome , Ultrasonography , Uterine Diseases/etiology , Uterine Diseases/therapy
14.
Hum Reprod ; 31(5): 1087-96, 2016 May.
Article in English | MEDLINE | ID: mdl-27005892

ABSTRACT

STUDY QUESTION: Could cell therapy using autologous peripheral blood CD133+ bone marrow-derived stem cells (BMDSCs) offer a safe and efficient therapeutic approach for patients with refractory Asherman's syndrome (AS) and/or endometrial atrophy (EA) and a wish to conceive? SUMMARY ANSWER: In the first 3 months, autologous cell therapy, using CD133+ BMDSCs in conjunction with hormonal replacement therapy, increased the volume and duration of menses as well as the thickness and angiogenesis processes of the endometrium while decreasing intrauterine adhesion scores. WHAT IS KNOWN ALREADY: AS is characterized by the presence of intrauterine adhesions and EA prevents the endometrium from growing thicker than 5 mm, resulting in menstruation disorders and infertility. Many therapies have been attempted for these conditions, but none have proved effective. STUDY DESIGN, SIZE, DURATION: This was a prospective, experimental, non-controlled study. There were 18 patients aged 30-45 years with refractory AS or EA were recruited, and 16 of these completed the study. Medical history, physical examination, endometrial thickness, intrauterine adhesion score and neoangiogenesis were assessed before and 3 and 6 months after cell therapy. PARTICIPANTS/MATERIALS, SETTING, METHODS: After the initial hysteroscopic diagnosis, BMDSC mobilization was performed by granulocyte-CSF injection, then CD133+ cells were isolated through peripheral blood aphaeresis to obtain a mean of 124.39 million cells (range 42-236), which were immediately delivered into the spiral arterioles by catheterization. Subsequently, endometrial treatment after stem cell therapy was assessed in terms of restoration of menses, endometrial thickness (by vaginal ultrasound), adhesion score (by hysteroscopy), neoangiogenesis and ongoing pregnancy rate. The study was conducted at Hospital Clínico Universitario of Valencia and IVI Valencia (Spain). MAIN RESULTS AND THE ROLE OF CHANCE: All 11 AS patients exhibited an improved uterine cavity 2 months after stem cell therapy. Endometrial thickness increased from an average of 4.3 mm (range 2.7-5) to 6.7 mm (range 3.1-12) ( ITALIC! P = 0.004). Similarly, four of the five EA patients experienced an improved endometrial cavity, and endometrial thickness increased from 4.2 mm (range 2.7-5) to 5.7 mm (range 5-12) ( ITALIC! P = 0.03). The beneficial effects of the cell therapy increased the mature vessel density and the duration and intensity of menses in the first 3 months, with a return to the initial levels 6 months after the treatment. Three patients became pregnant spontaneously, resulting in one baby boy born, one ongoing pregnancy and a miscarriage. Furthermore, seven pregnancies were obtained after fourteen embryo transfers, resulting in three biochemical pregnancies, one miscarriage, one ectopic pregnancy, one baby born and one ongoing pregnancy. LIMITATIONS, REASONS FOR CAUTION: Limitations of this pilot study include the small sample size and the lack of control group. WIDER IMPLICATIONS OF THE FINDINGS: This novel autologous cell therapy is a promising therapeutic option for patients with these incurable pathologies and a wish to conceive. STUDY FUNDING/COMPETING INTERESTS: This study was funded by the Spanish Ministry of Science and Innovation (SAF 2012-31017, Principal Investigator C.S.), Spanish Ministry of Health (EC11-299, Principal Investigator C.S.) and Regional Valencian Ministry of Education (PROMETEOII/2013/018, Principal Investigator C.S.). Four authors (X.S., I.C., A.P. and C.S.) are co-inventors of the patent resulting from this work (Application number: 62/013,121). S.C., C.A., F.R., J.F., J.P. and J.R. have no conflict of interest in relation to this work. TRIAL REGISTRATION NUMBER: This study was registered with ClinicalTrials.gov (NCT02144987).


Subject(s)
AC133 Antigen/metabolism , Blood Transfusion, Autologous , Cell- and Tissue-Based Therapy/methods , Gynatresia/therapy , Hematopoietic Stem Cell Transplantation , Transplantation, Autologous , Adult , Atrophy/therapy , Cohort Studies , Endometrium/pathology , Female , Hematopoietic Stem Cells/metabolism , Humans , Middle Aged , Pilot Projects , Prospective Studies
15.
Hum Reprod ; 31(12): 2723-2729, 2016 12.
Article in English | MEDLINE | ID: mdl-27664218

ABSTRACT

STUDY QUESTION: Does autologous transplantation of menstrual blood-derived stromal cells (menSCs) regenerate endometrium to support pregnancy in patients with severe Asherman's syndrome (AS)? SUMMARY ANSWER: Autologous menSCs transplantation significantly increases endometrial thickness (ET) for women with severe AS. WHAT IS KNOWN ALREADY: AS is a major cause of secondary infertility in women. Cell transplantation has been tried in a few clinical cases with encouraging results. STUDY DESIGN, SIZE, DURATION: In this experimental, non-controlled and prospective 3-year clinical study involving seven patients with AS, autologous menSCs were isolated and cultured from menstrual blood of each patient within ~2 weeks and then transplanted back into their uterus. Endometrial growth and pregnancy were assessed after cell therapy. PARTICIPANTS/MATERIALS, SETTING, METHOD: Infertile women, aged 20-40 years, diagnosed with severe AS (Grade III-V) by hysteroscopy and with menstrual fluid were recruited at the Shengjing Hospital affiliated to China Medical University. Autologous menSCs transplantation was conducted followed by HRT. Endometrial thickness was monitored with frozen embryo transfer (FET) as needed. MAIN RESULTS AND THE ROLE OF CHANCE: We successfully cultured menSCs from seven patients and transferred the autologous cells back to their uterus. Our results showed that the ET was significantly (P = 0.0002) increased to 7 mm in five women, which ensured embryo implantation. Four patients underwent FET and two of them conceived successfully. One patient had spontaneous pregnancy after second menSCs transplantation. LIMITATIONS, REASONS FOR CAUTION: Limited sample size, lack of rigorous controls or knowledge of underlying mechanism. WIDER IMPLICATIONS OF THE FINDINGS: Autologous menSCs transplantation is a potential option for treating women with severe AS. STUDY FUNDING/COMPETING INTERESTS: This study was supported by Liaoning Provincial Science and Technology Program. The sponsor and authors declare no conflicts of interest. TRIAL REGISTRATION NUMBER: Registered in the Chinese Clinical Trial Registry (ChiCTR-ONB-15007464).


Subject(s)
Endometrium/cytology , Gynatresia/therapy , Infertility, Female/therapy , Menstruation/blood , Stromal Cells/transplantation , Adult , Endometrium/diagnostic imaging , Female , Humans , Infertility, Female/diagnostic imaging , Prospective Studies , Transplantation, Autologous , Treatment Outcome , Ultrasonography
16.
Hum Reprod ; 30(2): 406-16, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25475585

ABSTRACT

STUDY QUESTION: Can regenerative-medicine techniques using oral mucosal epithelial cell sheets (OMECS) provide a new treatment method for intrauterine adhesions (IUA) which cause female infertility? SUMMARY ANSWER: Transplantation of OMECS was confirmed to be effective in preventing IUA after endometrial damage in rats. WHAT IS KNOWN ALREADY: Uterine disorders such as IUA, commonly known as Asherman's syndrome, are one factor that can result in infertility. Clinical therapy for this kind of disease is targeted at the prevention of re-adhesion by surgical synechiotomy, administration of hormones after the operation, and the use of intrauterine devices. Recently, a new approach called 'cell-sheet engineering', which harvests confluent culture cells as a contiguous cell sheet having intact cell-cell junctions and an extracellular matrix, without having to use enzymatic treatment, has been developed for tissue regeneration. STUDY DESIGN, SIZE, DURATION: OMECS were prepared from rat oral mucosal tissues. An IUA model was made in rat uteri, and OMECS were transplanted into the model. Uteri transplanted with OMECS were compared with the non-transplanted control uteri by histological analysis at 1, 2 and 8 days after surgery (n = 3). PARTICIPANTS/MATERIALS, SETTING, METHODS: Oral mucosal tissues were resected from neonatal rats, and oral mucosal epithelial cells were collected with enzymatic treatment. An isolated cell suspension was seeded on a temperature-responsive cell culture-insert and incubated. After being detached from the insert, a cell sheet was transplanted onto the endometrium defect. At 1, 2 and 8 days after surgery, uteri were resected and examined. MAIN RESULTS AND THE ROLE OF CHANCE: Histological examination of the non-treated specimens at 1, 2 and 8 days after surgery did not show any uterine cavities typically caused by IUA. In contrast, the histology of uteri transplanted with OMECS immediately after endometrial damage showed the presence of uterine cavities, and furthermore, stratified squamous epithelial cells on the luminal surface (n = 3). LIMITATIONS, REASONS FOR CAUTION: The results of this study are difficult to apply directly to humans, because the structure and function of rat uteri are different from those of human. WIDER IMPLICATIONS OF THE FINDINGS: Transplantation of OMECS offers a reliable method not only to protect the woman's fertility from intrauterine re-adhesion after synechiotomy for IUA or uterine lumen adhesion but also to prevent adhesion after any intrauterine surgery in clinical cases.


Subject(s)
Disease Models, Animal , Gynatresia/therapy , Mouth Mucosa/cytology , Regeneration , Tissue Adhesions/therapy , Tissue Engineering , Uterus/physiology , Animals , Animals, Newborn , Biomarkers/metabolism , Cells, Cultured , Crosses, Genetic , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Gynatresia/surgery , Immunohistochemistry , Mouth Mucosa/immunology , Mouth Mucosa/metabolism , Neutrophil Infiltration , Rats, Inbred F344 , Rats, Nude , Rats, Transgenic , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tissue Adhesions/surgery , Uterus/cytology , Uterus/immunology , Uterus/metabolism
17.
Acta Obstet Gynecol Scand ; 93(4): 425-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24484072

ABSTRACT

Intrauterine adhesions (Asherman syndrome) are rare and mainly seen after delivery or abortion in the presence of retained placental tissue. This descriptive study aimed to identify common risk factors for intrauterine adhesions. In a 10-year period 61 women were identified with intrauterine adhesions. The pathology was suspected from symptoms, ultrasonography or on hysterosalpingography, but a final diagnosis could only be given after hysteroscopy. There was no definite evidence regarding methods for prevention and treatment of the disorder. It seems, however, that a conservative approach to curettage, hysteroscopic removal of retained tissue, and the use of distending media are important, together with gentle tissue handling when such procedures are required.


Subject(s)
Gynatresia/diagnosis , Gynatresia/epidemiology , Pregnancy Rate , Tissue Adhesions/complications , Uterus/pathology , Adult , Curettage/adverse effects , Denmark/epidemiology , Female , Gynatresia/etiology , Gynatresia/therapy , Humans , Incidence , Pregnancy , Prevalence , Retrospective Studies , Tissue Adhesions/epidemiology , Tissue Adhesions/etiology , Tissue Adhesions/surgery , Treatment Outcome , Uterus/surgery
18.
Isr Med Assoc J ; 16(8): 502-5, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25269342

ABSTRACT

BACKGROUND: Removal of retained placental tissue postpartum and retained products of conception (RPOC) abortion is done by uterine curettage or hysteroscopy. Trauma to the endometrium from surgical procedures, primarily curettage, can cause intrauterine adhesions (Asherman's syndrome) and subsequent infertility. The incidence of malpractice claims relating to intrauterine adhesions is rising, justifying reevaluation of the optimal way of handling these complications. OBJECTIVES: To review malpractice claims regarding intrauterine adhesions, and to explore the clinical approach that might reduce those claims or improve their medical and legal outcomes. METHODS: We examined 42 Asherman's syndrome claims handled by MCI, the largest professional liability insurer in Israel. The clinical chart of each case was reviewed and analyzed by the event preceding the adhesion formations, timing and mode of diagnosis, and outcome. We also assessed whether the adverse outcome was caused by substandard care and it it could have been avoided by different clinical practice. The legal outcome was also evaluated. RESULTS: Forty-seven percent of the cases occurred following vaginal delivery, 19% followed cesarean section, 28% were RPOC following a first-trimester pregnancy termination, and 2% followed a second-trimester pregnancy termination. CONCLUSIONS: It is apparent that due to the lack of an accepted management protocol for cases of RPOC, it is difficult to legally defend those cases when the complication of Asherman syndrome develops.


Subject(s)
Gynatresia , Malpractice/statistics & numerical data , Obstetric Surgical Procedures/adverse effects , Obstetrics , Placenta, Retained , Adult , Clinical Protocols , Female , Gynatresia/etiology , Gynatresia/therapy , Humans , Insurance Claim Review , Israel , Liability, Legal , Obstetric Surgical Procedures/methods , Obstetrics/legislation & jurisprudence , Obstetrics/methods , Outcome Assessment, Health Care , Placenta, Retained/diagnosis , Placenta, Retained/therapy , Pregnancy
19.
Cell Transplant ; 32: 9636897231218408, 2023.
Article in English | MEDLINE | ID: mdl-38097275

ABSTRACT

Asherman's syndrome is an endometrial regeneration disorder resulting from injury to the endometrial basal layer, causing the formation of scar tissue in the uterus and cervix. This usually leads to uterine infertility, menstrual disorders, and placental abnormalities. While stem cell therapy has shown extensive progress in repairing the damaged endometrium and preventing intrauterine adhesion, issues of low engraftment rates, rapid senescence, and the risk of tumorigenesis remain to be resolved for efficient and effective application of this technology in endometrial repair. This study addressed these challenges by developing a co-culture system to generate multi-lineage endometrial organoids (MLEOs) comprising endometrial epithelium organoids (EEOs) and endometrial mesenchymal stem cells (eMSCs). The efficacy of these MLEOs was investigated by seeding them on a biocompatible scaffold, the human acellular amniotic membrane (HAAM), to create a biological graft patch, which was subsequently transplanted into an injury model of the endometrium in rats. The results indicated that the MLEOs on the HAAM patch facilitated endometrial angiogenesis, regeneration, and improved pregnancy outcomes. The MLEOs on the HAAM patch could serve as a promising strategy for treating endometrial injury and preventing Asherman's syndrome.


Subject(s)
Gynatresia , Humans , Female , Rats , Animals , Pregnancy , Gynatresia/therapy , Amnion , Placenta , Endometrium , Uterus
20.
Stem Cell Res Ther ; 13(1): 33, 2022 01 28.
Article in English | MEDLINE | ID: mdl-35090547

ABSTRACT

Numerous treatment strategies have so far been proposed for treating refractory thin endometrium either without or with the Asherman syndrome. Inconsistency in the improvement of endometrial thickness is a common limitation of such therapies including tamoxifen citrate as an ovulation induction agent, acupuncture, long-term pentoxifylline and tocopherol or tocopherol only, low-dose human chorionic gonadotropin during endometrial preparation, aspirin, luteal gonadotropin-releasing hormone agonist supplementation, and extended estrogen therapy. Recently, cell therapy has been proposed as an ideal alternative for endometrium regeneration, including the employment of stem cells, platelet-rich plasma, and growth factors as therapeutic agents. The mechanisms of action of cell therapy include the cytokine induction, growth factor production, natural killer cell activity reduction, Th17 and Th1 decrease, and Treg cell and Th2 increase. Since cell therapy is personalized, dynamic, interactive, and specific and could be an effective strategy. Despite its promising nature, further research is required for improving the procedure and the safety of this strategy. These methods and their results are discussed in this article.


Subject(s)
Gynatresia , Platelet-Rich Plasma , Cell- and Tissue-Based Therapy , Chorionic Gonadotropin , Endometrium , Female , Gynatresia/therapy , Humans
SELECTION OF CITATIONS
SEARCH DETAIL