1
|
Rodríguez-Eguren A, Bueno-Fernandez C, Gómez-Álvarez M, Francés-Herrero E, Pellicer A, Bellver J, Seli E, Cervelló I. Evolution of biotechnological advances and regenerative therapies for endometrial disorders: a systematic review. Hum Reprod Update 2024; 30:584-613. [PMID: 38796750 PMCID: PMC11369227 DOI: 10.1093/humupd/dmae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/12/2024] [Indexed: 05/28/2024] Open
Abstract
BACKGROUND The establishment and maintenance of pregnancy depend on endometrial competence. Asherman syndrome (AS) and intrauterine adhesions (IUA), or endometrial atrophy (EA) and thin endometrium (TE), can either originate autonomously or arise as a result from conditions (i.e. endometritis or congenital hypoplasia), or medical interventions (e.g. surgeries, hormonal therapies, uterine curettage or radiotherapy). Affected patients may present an altered or inadequate endometrial lining that hinders embryo implantation and increases the risk of poor pregnancy outcomes and miscarriage. In humans, AS/IUA and EA/TE are mainly treated with surgeries or pharmacotherapy, however the reported efficacy of these therapeutic approaches remains unclear. Thus, novel regenerative techniques utilizing stem cells, growth factors, or tissue engineering have emerged to improve reproductive outcomes. OBJECTIVE AND RATIONALE This review comprehensively summarizes the methodologies and outcomes of emerging biotechnologies (cellular, acellular, and bioengineering approaches) to treat human endometrial pathologies. Regenerative therapies derived from human tissues or blood which were studied in preclinical models (in vitro and in vivo) and clinical trials are discussed. SEARCH METHODS A systematic search of full-text articles available in PubMed and Embase was conducted to identify original peer-reviewed studies published in English between January 2000 and September 2023. The search terms included: human, uterus, endometrium, Asherman syndrome, intrauterine adhesions, endometrial atrophy, thin endometrium, endometritis, congenital hypoplasia, curettage, radiotherapy, regenerative therapy, bioengineering, stem cells, vesicles, platelet-rich plasma, biomaterials, microfluidic, bioprinting, organoids, hydrogel, scaffold, sheet, miRNA, sildenafil, nitroglycerine, aspirin, growth hormone, progesterone, and estrogen. Preclinical and clinical studies on cellular, acellular, and bioengineering strategies to repair or regenerate the human endometrium were included. Additional studies were identified through manual searches. OUTCOMES From a total of 4366 records identified, 164 studies (3.8%) were included for systematic review. Due to heterogeneity in the study design and measured outcome parameters in both preclinical and clinical studies, the findings were evaluated qualitatively and quantitatively without meta-analysis. Groups using stem cell-based treatments for endometrial pathologies commonly employed mesenchymal stem cells (MSCs) derived from the human bone marrow or umbilical cord. Alternatively, acellular therapies based on platelet-rich plasma (PRP) or extracellular vesicles are gaining popularity. These are accompanied by the emergence of bioengineering strategies based on extracellular matrix (ECM)-derived hydrogels or synthetic biosimilars that sustain local delivery of cells and growth factors, reporting promising results. Combined therapies that target multiple aspects of tissue repair and regeneration remain in preclinical testing but have shown translational value. This review highlights the myriad of therapeutic material sources, administration methods, and carriers that have been tested. WIDER IMPLICATIONS Therapies that promote endometrial proliferation, vascular development, and tissue repair may help restore endometrial function and, ultimately, fertility. Based on the existing evidence, cost, accessibility, and availability of the therapies, we propose the development of triple-hit regenerative strategies, potentially combining high-yield MSCs (e.g. from bone marrow or umbilical cord) with acellular treatments (PRP), possibly integrated in ECM hydrogels. Advances in biotechnologies together with insights from preclinical models will pave the way for developing personalized treatment regimens for patients with infertility-causing endometrial disorders such as AS/IUA, EA/TE, and endometritis. REGISTRATION NUMBER https://osf.io/th8yf/.
Collapse
Affiliation(s)
- Adolfo Rodríguez-Eguren
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Clara Bueno-Fernandez
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
- Department of Paediatrics, Obstetrics and Gynecology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - María Gómez-Álvarez
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Emilio Francés-Herrero
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
- Department of Paediatrics, Obstetrics and Gynecology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Antonio Pellicer
- Department of Paediatrics, Obstetrics and Gynecology, Faculty of Medicine, University of Valencia, Valencia, Spain
- IVIRMA Global Research Alliance, IVI Rome, Rome, Italy
| | - José Bellver
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
- Department of Paediatrics, Obstetrics and Gynecology, Faculty of Medicine, University of Valencia, Valencia, Spain
- IVIRMA Global Research Alliance, IVI Valencia, Valencia, Spain
| | - Emre Seli
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
- IVIRMA Global Research Alliance, IVIRMA New Jersey, Basking Ridge, NJ, USA
| | - Irene Cervelló
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| |
Collapse
|
2
|
Amiri M, Kaviari MA, Rostaminasab G, Barimani A, Rezakhani L. A novel cell-free therapy using exosomes in the inner ear regeneration. Tissue Cell 2024; 88:102373. [PMID: 38640600 DOI: 10.1016/j.tice.2024.102373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/01/2024] [Accepted: 04/03/2024] [Indexed: 04/21/2024]
Abstract
Cellular and molecular alterations associated with hearing loss are now better understood with advances in molecular biology. These changes indicate the participation of distinct damage and stress pathways that are unlikely to be fully addressed by conventional pharmaceutical treatment. Sensorineural hearing loss is a common and debilitating condition for which comprehensive pharmacologic intervention is not available. The complex and diverse molecular pathology that underlies hearing loss currently limits our ability to intervene with small molecules. The present review focuses on the potential for the use of extracellular vesicles in otology. It examines a variety of inner ear diseases and hearing loss that may be treatable using exosomes (EXOs). The role of EXOs as carriers for the treatment of diseases related to the inner ear as well as EXOs as biomarkers for the recognition of diseases related to the ear is discussed.
Collapse
Affiliation(s)
- Masoumeh Amiri
- Faculty of Medicine, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Mohammad Amin Kaviari
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran; Universal Scientific Education and Research Network (USERN) Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Gelavizh Rostaminasab
- Clinical Research Development Center, Imam Khomeini and Mohammad Kermanshahi and Farabi Hospitals, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Barimani
- Clinical Research Development Center, Imam Khomeini and Mohammad Kermanshahi and Farabi Hospitals, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
3
|
Shin EY, Jeong S, Lee JE, Jeong DS, Han DK, Hong SH, Lee DR. Multiple treatments with human embryonic stem cell-derived mesenchymal progenitor cells preserved the fertility and ovarian function of perimenopausal mice undergoing natural aging. Stem Cell Res Ther 2024; 15:58. [PMID: 38433223 PMCID: PMC10910829 DOI: 10.1186/s13287-024-03684-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/23/2024] [Indexed: 03/05/2024] Open
Abstract
OBJECTIVES Currently, no approved stem cell-based therapies for preserving ovarian function during aging. To solve this problem, we developed a long-term treatment for human embryonic stem cell-derived mesenchymal progenitor cells (hESC-MPCs). We investigated whether the cells retained their ability to resist ovarian aging, which leads to delayed reproductive senescence. MATERIALS AND METHODS In a middle-aged female model undergoing natural aging, we analyzed whether hESC-MPCs benefit the long-term maintenance of reproductive fecundity and ovarian reservoirs and how their transplantation regulates ovarian function. RESULTS The number of primordial follicles and mice with regular estrous cycles were increased in perimenopausal mice who underwent multiple introductions of hESC-MPCs compared to age-matched controls. The estradiol levels in the hESC-MPCs group were restored to those in the young and adult groups. Embryonic development and live birth rates were higher in the hESC-MPC group than in the control group, suggesting that hESC-MPCs delayed ovarian senescence. In addition to their direct effects on the ovary, multiple-treatments with hESC-MPCs reduced ovarian fibrosis by downregulating inflammation and fibrosis-related genes via the suppression of myeloid-derived suppressor cells (MDSCs) produced in the bone marrow. CONCLUSIONS Multiple introductions of hESC-MPCs could be a useful approach to prevent female reproductive senescence and that these cells are promising sources for cell therapy to postpone the ovarian aging and retain fecundity in perimenopausal women.
Collapse
Affiliation(s)
- Eun-Young Shin
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam- si, 13488, Gyeonggi-do, Republic of Korea
| | - Suji Jeong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, 24431, Gangwon-do, Republic of Korea
| | - Jeoung Eun Lee
- CHA Advanced Research Institute, Bundang CHA Medical Center, 335 Pangyo-ro, Bundang- gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Dong Seok Jeong
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam- si, 13488, Gyeonggi-do, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam- si, 13488, Gyeonggi-do, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, 24431, Gangwon-do, Republic of Korea.
| | - Dong Ryul Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam- si, 13488, Gyeonggi-do, Republic of Korea.
- CHA Advanced Research Institute, Bundang CHA Medical Center, 335 Pangyo-ro, Bundang- gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
4
|
Khan T, Waseem R, Shahid M, Ansari J, Ahanger IA, Hassan I, Islam A. Recent advancement in therapeutic strategies for Alzheimer's disease: Insights from clinical trials. Ageing Res Rev 2023; 92:102113. [PMID: 37918760 DOI: 10.1016/j.arr.2023.102113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, characterized by the presence of plaques of amyloid beta and Tau proteins. There is currently no permanent cure for AD; the only medications approved by the FDA for mild to moderate AD are cholinesterase inhibitors, NMDA receptor antagonists, and immunotherapies against core pathophysiology, that provide temporary relief only. Researchers worldwide have made significant attempts to find new targets and develop innovative therapeutic molecules to treat AD. The FDA-approved drugs are palliative and couldn't restore the damaged neuron cells of AD. Stem cells have self-differentiation properties, making them prospective therapeutics to treat AD. The promising results in pre-clinical studies of stem cell therapy for AD seek attention worldwide. Various stem cells, mainly mesenchymal stem cells, are currently in different phases of clinical trials and need more advancements to take this therapy to the translational level. Here, we review research from the past decade that has identified several hypotheses related to AD pathology. Moreover, this article also focuses on the recent advancement in therapeutic strategies for AD treatment including immunotherapy and stem cell therapy detailing the clinical trials that are currently undergoing development.
Collapse
Affiliation(s)
- Tanzeel Khan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Rashid Waseem
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohammad Shahid
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Jaoud Ansari
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ishfaq Ahmad Ahanger
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; Department of Clinical Biochemistry, University of Kashmir,190006, India
| | - Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
5
|
Rodrigues LLV, Moura YBF, Viana JVDS, de Oliveira LRM, Praxedes ÉA, Vieira JDB, Sales SLA, Silva HVR, Luciano MCDS, Pessoa C, Pereira AF. Full confluency, serum starvation, and roscovitine for inducing arrest in the G 0/G 1 phase of the cell cycle in puma skin-derived fibroblast lines. Anim Reprod 2023; 20:e20230017. [PMID: 37101424 PMCID: PMC10124155 DOI: 10.1590/1984-3143-ar2023-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/22/2023] [Indexed: 04/28/2023] Open
Abstract
The puma population is constantly decreasing, and cloning by somatic cell nuclear transfer can be used to conserve the species. One of the factors determining the success of the development of cloned embryos is the cell cycle stage of the donor cells. We evaluated the effects of full confluency (~100%), serum starvation (0.5% serum), and roscovitine (15 µM) treatments on the cell cycle synchronization in G0/G1 of puma skin-derived fibroblasts by flow cytometric analysis. Also, we assessed the effects of these synchronization methods on morphology, viability, and apoptosis levels using microscopy tools. The results showed that culturing the cells to confluence for 24 h (84.0%), 48 h (84.6%), and 72 h (84.2%) and serum starvation for 96 h (85.4%) yielded a significantly higher percentage of cells arrested in the G0/G1 (P 0.05) phase than cells not subjected to any cell cycle synchronization method (73.9%). Nevertheless, while serum starvation reduced the percentage of viable cells, no difference was observed for the full confluence and roscovitine treatments (P 0.05). Moreover, roscovitine for 12 h (78.6%) and 24 h (82.1%) was unable to synchronize cells in G0/G1 (P 0.05). In summary, full confluency induces puma fibroblast cell cycle synchronization at the G0/G1 stage without affecting cell viability. These outcomes may be valuable for planning donor cells for somatic cell nuclear transfer in pumas.
Collapse
Affiliation(s)
| | | | | | | | - Érika Almeida Praxedes
- Laboratório de Biotecnologia Animal, Universidade Federal Rural do Semi-Árido, Mossoró, RN, Brasil
| | - José de Brito Vieira
- Laboratório de Oncologia Experimental, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | | | | | | | - Claudia Pessoa
- Laboratório de Oncologia Experimental, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - Alexsandra Fernandes Pereira
- Laboratório de Biotecnologia Animal, Universidade Federal Rural do Semi-Árido, Mossoró, RN, Brasil
- Corresponding author:
| |
Collapse
|
6
|
Kim SY, Lee JE, Kang SH, Lee SM, Jeon J, Lee DR. The Protective Effects of Human Embryonic Stem Cell-Derived Mesenchymal Stem Cells in Noise-Induced Hearing Loss of Rats. Cells 2022; 11:3524. [PMID: 36359920 PMCID: PMC9654588 DOI: 10.3390/cells11213524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/24/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2024] Open
Abstract
A few prior animal studies have suggested the transplantation or protective effects of mesenchymal stem cells (MSCs) in noise-induced hearing loss. This study intended to evaluate the fates of administered MSCs in the inner ears and the otoprotective effects of MSCs in the noise-induced hearing loss of rats. Human embryonic stem cell-derived MSCs (ES-MSCs) were systematically administered via the tail vein in adult rats. Eight-week-old Sprague-Dawley rats were randomly allocated to the control (n = 8), ES-MSC (n = 4), noise (n = 8), and ES-MSC+noise (n = 10) groups. In ES-MSC and ES-MSC+noise rats, 5 × 105 ES-MSCs were injected via the tail vein. In noise and ES-MSC+noise rats, broadband noise with 115 dB SPL was exposed for 3 h daily for 5 days. The hearing levels were measured using auditory brainstem response (ABR) at 4, 8, 16, and 32 kHz. Cochlear histology was examined using H&E staining and cochlear whole mount immunofluorescence. The presence of human DNA was examined using Sry PCR, and the presence of human cytoplasmic protein was examined using STEM121 immunofluorescence staining. The protein expression levels of heat shock protein 70 (HSP70), apoptosis-inducing factor (AIF), poly (ADP-ribose) (PAR), PAR polymerase (PARP), caspase 3, and cleaved caspase 3 were estimated. The ES-MSC rats did not show changes in ABR thresholds following the administration of ES-MSCs. The ES-MSC+ noise rats demonstrated lower ABR thresholds at 4, 8, and 16 kHz than the noise rats. Cochlear spiral ganglial cells and outer hair cells were more preserved in the ES-MSC+ noise rats than in the noise rats. The Sry PCR bands were highly detected in lung tissue and less in cochlear tissue of ES-MSC+noise rats. Only a few STEM121-positivities were observed in the spiral ganglial cell area of ES-MSC and ES-MSC+noise rats. The protein levels of AIF, PAR, PARP, caspase 3, and cleaved caspase 3 were lower in the ES-MSC+noise rats than in the noise rats. The systemic injection of ES-MSCs preserved hearing levels and attenuated parthanatos and apoptosis in rats with noise-induced hearing loss. In addition, a tiny number of transplanted ES-MSCs were observed in the spiral ganglial areas.
Collapse
Affiliation(s)
- So Young Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, CHA University, Seongnam-si 13496, Korea
| | - Jeoung Eun Lee
- CHA Advanced Research Institute, Seongnam-si 13488, Korea
| | - Sung Hun Kang
- School of Medicine, CHA University, Seongnam-si 13488, Korea
| | - So Min Lee
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, CHA University, Seongnam-si 13496, Korea
| | - Jiwon Jeon
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, CHA University, Seongnam-si 13496, Korea
| | - Dong Ryul Lee
- CHA Advanced Research Institute, Seongnam-si 13488, Korea
- Department of Biomedical Science, CHA University, Seongnam-si 13488, Korea
| |
Collapse
|
7
|
Kim MK, Yoon JA, Yoon SY, Park M, Lee WS, Lyu SW, Song H. Human Platelet-Rich Plasma Facilitates Angiogenesis to Restore Impaired Uterine Environments with Asherman’s Syndrome for Embryo Implantation and Following Pregnancy in Mice. Cells 2022; 11:cells11091549. [PMID: 35563855 PMCID: PMC9101537 DOI: 10.3390/cells11091549] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 02/05/2023] Open
Abstract
Asherman’s syndrome (AS) is caused by intrauterine adhesions and inactive endometrium from repeated curettage of the uterine endometrium. AS is a major cause of recurrent implantation failure and miscarriage and is very difficult to treat because of the poor recovery of endometrial basal cells. Platelet-rich plasma (PRP) has abundant growth factors that may induce angiogenesis and cell proliferation. Here, we demonstrate that human PRP (hPRP) significantly enhances angiogenesis to restore embryo implantation, leading to successful pregnancy in mice with AS. In mice with AS, hPRP treatment considerably reduced the expression of fibrosis markers and alleviated oligo/amenorrhea phenotypes. Mice with AS did not produce any pups, but the hPRP therapy restored their infertility. AS-induced abnormalities, such as aberrantly delayed embryo implantation and intrauterine growth retardation, were considerably eliminated by hPRP. Furthermore, hPRP significantly promoted not only the elevation of various angiogenic factors, but also the migration of endometrial stromal cells. It also increased the phosphorylation of STAT3, a critical mediator of wound healing, and the expression of tissue remodeling genes in a fibrotic uterus. PRP could be a promising therapeutic strategy to promote angiogenesis and reduce fibrosis in impaired uterine environments, leading to successful embryo implantation for better clinical outcomes in patients with AS.
Collapse
Affiliation(s)
- Min Kyoung Kim
- Department of Obstetrics and Gynecology, CHA Fertility Center Gangnam, CHA University School of Medicine, 569 Nonhyun-ro, Gangnam-gu, Seoul 06125, Korea; (M.K.K.); (J.A.Y.); (S.Y.Y.); (W.S.L.); (S.W.L.)
| | - Jung Ah Yoon
- Department of Obstetrics and Gynecology, CHA Fertility Center Gangnam, CHA University School of Medicine, 569 Nonhyun-ro, Gangnam-gu, Seoul 06125, Korea; (M.K.K.); (J.A.Y.); (S.Y.Y.); (W.S.L.); (S.W.L.)
| | - Sook Young Yoon
- Department of Obstetrics and Gynecology, CHA Fertility Center Gangnam, CHA University School of Medicine, 569 Nonhyun-ro, Gangnam-gu, Seoul 06125, Korea; (M.K.K.); (J.A.Y.); (S.Y.Y.); (W.S.L.); (S.W.L.)
| | - Mira Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam 13488, Korea;
| | - Woo Sik Lee
- Department of Obstetrics and Gynecology, CHA Fertility Center Gangnam, CHA University School of Medicine, 569 Nonhyun-ro, Gangnam-gu, Seoul 06125, Korea; (M.K.K.); (J.A.Y.); (S.Y.Y.); (W.S.L.); (S.W.L.)
| | - Sang Woo Lyu
- Department of Obstetrics and Gynecology, CHA Fertility Center Gangnam, CHA University School of Medicine, 569 Nonhyun-ro, Gangnam-gu, Seoul 06125, Korea; (M.K.K.); (J.A.Y.); (S.Y.Y.); (W.S.L.); (S.W.L.)
| | - Haengseok Song
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam 13488, Korea;
- Correspondence: ; Tel.: +82-031-881-7150
| |
Collapse
|
8
|
Kirkwood PM, Shaw IW, Saunders PTK. Mechanisms of Scarless Repair at Time of Menstruation: Insights From Mouse Models. FRONTIERS IN REPRODUCTIVE HEALTH 2022; 3:801843. [PMID: 36304046 PMCID: PMC9580659 DOI: 10.3389/frph.2021.801843] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/08/2021] [Indexed: 11/18/2022] Open
Abstract
The human endometrium is a remarkable tissue which may experience up to 400 cycles of hormone-driven proliferation, differentiation and breakdown during a woman's reproductive lifetime. During menstruation, when the luminal portion of tissue breaks down, it resembles a bloody wound with piecemeal shedding, exposure of underlying stroma and a strong inflammatory reaction. In the absence of pathology within a few days the integrity of the tissue is restored without formation of a scar and the endometrium is able to respond appropriately to subsequent endocrine signals in preparation for establishment of pregnancy if fertilization occurs. Understanding mechanisms regulating scarless repair of the endometrium is important both for design of therapies which can treat conditions where this is aberrant (heavy menstrual bleeding, fibroids, endometriosis, Asherman's syndrome) as well as to provide new information that might allow us to reduce fibrosis and scar formation in other tissues. Menstruation only occurs naturally in species that exhibit spontaneous stromal cell decidualization during the fertile cycle such as primates (including women) and the Spiny mouse. To take advantage of genetic models and detailed time course analysis, mouse models of endometrial shedding/repair involving hormonal manipulation, artificial induction of decidualization and hormone withdrawal have been developed and refined. These models are useful in modeling dynamic changes across the time course of repair and have recapitulated key features of endometrial repair in women including local hypoxia and immune cell recruitment. In this review we will consider the evidence that scarless repair of endometrial tissue involves changes in stromal cell function including mesenchyme to epithelial transition, epithelial cell proliferation and multiple populations of immune cells. Processes contributing to endometrial fibrosis (Asherman's syndrome) as well as scarless repair of other tissues including skin and oral mucosa are compared to that of menstrual repair.
Collapse
|
9
|
López-Martínez S, Rodríguez-Eguren A, de Miguel-Gómez L, Francés-Herrero E, Faus A, Díaz A, Pellicer A, Ferrero H, Cervelló I. Bioengineered endometrial hydrogels with growth factors promote tissue regeneration and restore fertility in murine models. Acta Biomater 2021; 135:113-125. [PMID: 34428563 DOI: 10.1016/j.actbio.2021.08.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
Extracellular matrix (ECM) hydrogels obtained from decellularized tissues are promising biocompatible materials for tissue regeneration. These biomaterials may provide important options for endometrial pathologies such as Asherman's syndrome and endometrial atrophy, which lack effective therapies thus far. First, we performed a proteomic analysis of a decellularized endometrial porcine hydrogel (EndoECM) to describe the specific role of ECM proteins related to regenerative processes. Furthermore, we investigated the ability of a bioengineered system-EndoECM alone or supplemented with growth factors (GFs)-to repair the endometrium in a murine model of endometrial damage. For this model, the uterine horns of female C57BL/6 mice were first injected with 70% ethanol, then four days later, they were treated with: saline (negative control); biotin-labeled EndoECM; or biotin-labeled EndoECM plus platelet-derived GF, basic fibroblast GF, and insulin-like GF 1 (EndoECM+GF). Endometrial regeneration and fertility restoration were evaluated by assessing the number of glands, endometrial area, cell proliferation, neaoangiogenesis, reduction of collagen deposition, and fertility restoration. Interestingly, regenerative effects such as an increased number of endometrial glands, increased area, high cell proliferative index, development of new blood vessels, reduction of collagen deposition, and higher pregnancy rate occurred in mice treated with EndoECM+GF. Thus, a bioengineered system based on EndoECM hydrogel supplemented with GFs may be promising for the clinical treatment of endometrial conditions such as Asherman's syndrome and endometrial atrophy. STATEMENT OF SIGNIFICANCE: In the last years, the bioengineering field has developed new and promising approaches to regenerate tissues or replace damaged and diseased tissues. Bioengineered hydrogels offer an ideal option because these materials can be used not only as treatments but also as carriers of drugs and other therapeutics. The present work demonstrates for the first time how hydrogels derived from pig endometrium loaded with growth factors could treat uterine pathologies in a mouse model of endometrial damage. These findings provide scientific evidence about bioengineered hydrogels based on tissue-specific extracellular matrix offering new options to treat human infertility from endometrial causes such as Asherman's syndrome or endometrial atrophy.
Collapse
Affiliation(s)
- Sara López-Martínez
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106, Hospital La Fe, Torre A, Planta 1ª, Valencia 46026, Spain
| | - Adolfo Rodríguez-Eguren
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106, Hospital La Fe, Torre A, Planta 1ª, Valencia 46026, Spain
| | - Lucía de Miguel-Gómez
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106, Hospital La Fe, Torre A, Planta 1ª, Valencia 46026, Spain; University of Valencia, Avenida de Blasco Ibáñez, 13, Valencia 46010, Spain
| | - Emilio Francés-Herrero
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106, Hospital La Fe, Torre A, Planta 1ª, Valencia 46026, Spain; University of Valencia, Avenida de Blasco Ibáñez, 13, Valencia 46010, Spain
| | - Amparo Faus
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106, Hospital La Fe, Torre A, Planta 1ª, Valencia 46026, Spain
| | - Ana Díaz
- University of Valencia, Avenida de Blasco Ibáñez, 13, Valencia 46010, Spain
| | - Antonio Pellicer
- University of Valencia, Avenida de Blasco Ibáñez, 13, Valencia 46010, Spain; IVIRMA Roma, Largo Ildebrando Pizzetti, 1, Roma 00197, Italy
| | - Hortensia Ferrero
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106, Hospital La Fe, Torre A, Planta 1ª, Valencia 46026, Spain
| | - Irene Cervelló
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106, Hospital La Fe, Torre A, Planta 1ª, Valencia 46026, Spain.
| |
Collapse
|
10
|
Rapid Production and Genetic Stability of Human Mesenchymal Progenitor Cells Derived from Human Somatic Cell Nuclear Transfer-Derived Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms22179238. [PMID: 34502145 PMCID: PMC8431643 DOI: 10.3390/ijms22179238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022] Open
Abstract
Pluripotent stem cell-derived mesenchymal progenitor cells (PSC-MPCs) are primarily derived through two main methods: three-dimensional (3D) embryoid body-platform (EB formation) and the 2D direct differentiation method. We recently established somatic cell nuclear transfer (SCNT)-PSC lines and showed their stemness. In the present study, we produced SCNT-PSC-MPCs using a novel direct differentiation method, and the characteristics, gene expression, and genetic stability of these MPCs were compared with those derived through EB formation. The recovery and purification of SCNT-PSC-Direct-MPCs were significantly accelerated compared to those of the SCNT-PSC-EB-MPCs, but both types of MPCs expressed typical surface markers and exhibited similar proliferation and differentiation potentials. Additionally, the analysis of gene expression patterns using microarrays showed very similar patterns. Moreover, array CGH analysis showed that both SCNT-PSC-Direct-MPCs and SCNT-PSC-EB-MPCs exhibited no significant differences in copy number variation (CNV) or single-nucleotide polymorphism (SNP) frequency. These results indicate that SCNT-PSC-Direct-MPCs exhibited high genetic stability even after rapid differentiation into MPCs, and the rate at which directly derived MPCs reached a sufficient number was higher than that of MPCs derived through the EB method. Therefore, we suggest that the direct method of differentiating MPCs from SCNT-PSCs can improve the efficacy of SCNT-PSCs applied to allogeneic transplantation.
Collapse
|
11
|
Shin EY, Kim DS, Lee MJ, Lee AR, Shim SH, Baek SW, Han DK, Lee DR. Prevention of chemotherapy-induced premature ovarian insufficiency in mice by scaffold-based local delivery of human embryonic stem cell-derived mesenchymal progenitor cells. Stem Cell Res Ther 2021; 12:431. [PMID: 34332643 PMCID: PMC8325282 DOI: 10.1186/s13287-021-02479-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/27/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is one of the most serious side effects of chemotherapy in young cancer survivors. It may not only reduce fecundity but also affect lifelong health. There is no standard therapy for preserving ovarian health after chemotherapy. Recently, administration of embryonic stem cell-derived mesenchymal progenitor cells (ESC-MPCs) has been considered a new therapeutic option for preventing POI. However, the previous method of directly injecting cells into the veins of patients exhibits low efficacy and safety. This study aimed to develop safe and effective local delivery methods for the prevention of POI using two types of bioinspired scaffolds. METHODS Female mice received intraperitoneal cisplatin for 10 days. On day 11, human ESC-MPCs were delivered through systemic administration using intravenous injection or local administration using intradermal injection and intradermal transplantation with a PLGA/MH sponge or hyaluronic acid (HA) gel (GEL) type of scaffold. PBS was injected intravenously as a negative control. Ovarian function and fertility were evaluated 4 weeks after transplantation. Follicle development was observed using hematoxylin and eosin staining. The plasma levels of sex hormones were measured using ELISA. Expression levels of anti-Müllerian hormone (AMH) and ki-67 were detected using immunostaining, and the quality of oocytes and embryos was evaluated after in vitro fertilization. The estrous cycles were observed at 2 months after transplantation. RESULTS The local administration of human ESC-MPCs using the bioinspired scaffold to the backs of mice effectively prolonged the cell survival rate in vivo. The HA GEL group exhibited the best recovered ovarian functions, including a significantly increased number of ovarian reserves, estrogen levels, and AMH levels and decreased apoptotic levels. Furthermore, the HA GEL group showed improved quality of oocytes and embryos and estrous cycle regularity. CONCLUSIONS HA GEL scaffolds can be used as new delivery platforms for ESC-MPC therapy, and this method may provide a novel option for the clinical treatment of chemotherapy-induced POI.
Collapse
Affiliation(s)
- Eun-Young Shin
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea
| | - Da-Seul Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Min Ji Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea
| | - Ah Reum Lee
- CHA Advanced Research Institute, CHA Medical Center, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea
| | - Sung Han Shim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea
| | - Seung Woon Baek
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea.
| | - Dong Ryul Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea.
| |
Collapse
|
12
|
Shin EY, Yoon YJ, Lee JE, Shim SH, Park GH, Lee DR. Identification of Putative Markers That Predict the In Vitro Senescence of Mesenchymal Progenitor Cells. Cells 2021; 10:cells10061301. [PMID: 34073789 PMCID: PMC8225148 DOI: 10.3390/cells10061301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal progenitor cells (MPCs) are a promising cell source for regenerative medicine because of their immunomodulatory properties, anti-inflammatory molecule secretion, and replacement of damaged cells. Despite these advantages, heterogeneity in functional potential and limited proliferation capacity of MPCs, as well as the lack of suitable markers for product potency, hamper the development of large-scale manufacturing processes of MPCs. Therefore, there is a sustained need to develop highly proliferative and standardized MPCs in vitro and find suitable functional markers for measuring product potency. In this study, three lines of pluripotent stem cell (PSC)-derived MPCs with high proliferative ability were established and compared with bone-marrow-derived MPCs using proliferation assays and microarrays. A total of six genes were significantly overexpressed (>10-fold) in the highest proliferative MPC line (CHA-hNT5-MPCs) and validated by qRT-PCR. However, only two of the genes (MYOCD and ODZ2) demonstrated a significant correlation with MPC senescence in vitro. Our study provides new gene markers for predicting replicative senescence and the available quantity of MPCs but may also help to guide the development of new standard criteria for manufacturing.
Collapse
Affiliation(s)
- Eun-Young Shin
- Department of Biomedical Science, CHA University, Seongnam 13488, Gyunggi-do, Korea; (E.-Y.S.); (Y.-J.Y.); (S.H.S.); (G.H.P.)
| | - Yeo-Joon Yoon
- Department of Biomedical Science, CHA University, Seongnam 13488, Gyunggi-do, Korea; (E.-Y.S.); (Y.-J.Y.); (S.H.S.); (G.H.P.)
| | - Jeoung Eun Lee
- CHA Advanced Research Institute, CHA University, Seongnam 13488, Gyunggi-do, Korea;
| | - Sung Han Shim
- Department of Biomedical Science, CHA University, Seongnam 13488, Gyunggi-do, Korea; (E.-Y.S.); (Y.-J.Y.); (S.H.S.); (G.H.P.)
| | - Gene Hong Park
- Department of Biomedical Science, CHA University, Seongnam 13488, Gyunggi-do, Korea; (E.-Y.S.); (Y.-J.Y.); (S.H.S.); (G.H.P.)
| | - Dong Ryul Lee
- Department of Biomedical Science, CHA University, Seongnam 13488, Gyunggi-do, Korea; (E.-Y.S.); (Y.-J.Y.); (S.H.S.); (G.H.P.)
- Correspondence:
| |
Collapse
|
13
|
Kim HJ, Hong SJ, Lee S, Park JM, Park J, Park JS, Shim SH, Park K. Induction of Bone Formation by 3D Biologically Active Scaffolds Containing RGD‐NPs, BMP2, and NtMPCs. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Hye Jin Kim
- Laboratory of Nano‐regenerative Medical Engineering Department of Biomedical Science College of Life Science CHA University 618, CHA Biocomplex, Sampyeong‐Dong Bundang‐gu Seongnam‐si 13488 Republic of Korea
| | - Suk Jun Hong
- Laboratory of Nano‐regenerative Medical Engineering Department of Biomedical Science College of Life Science CHA University 618, CHA Biocomplex, Sampyeong‐Dong Bundang‐gu Seongnam‐si 13488 Republic of Korea
| | - Sujin Lee
- Laboratory of Nano‐regenerative Medical Engineering Department of Biomedical Science College of Life Science CHA University 618, CHA Biocomplex, Sampyeong‐Dong Bundang‐gu Seongnam‐si 13488 Republic of Korea
| | - Jong Min Park
- Laboratory of Nano‐regenerative Medical Engineering Department of Biomedical Science College of Life Science CHA University 618, CHA Biocomplex, Sampyeong‐Dong Bundang‐gu Seongnam‐si 13488 Republic of Korea
| | - Ji‐In Park
- Laboratory of Nano‐regenerative Medical Engineering Department of Biomedical Science College of Life Science CHA University 618, CHA Biocomplex, Sampyeong‐Dong Bundang‐gu Seongnam‐si 13488 Republic of Korea
| | - Ji Sun Park
- Laboratory of Nano‐regenerative Medical Engineering Department of Biomedical Science College of Life Science CHA University 618, CHA Biocomplex, Sampyeong‐Dong Bundang‐gu Seongnam‐si 13488 Republic of Korea
| | - Sung Han Shim
- Laboratory of Molecular Genetics Department of Biomedical Science College of Life Science CHA University 629, CHA Biocomplex, Sampyeong‐Dong Bundang‐gu Seongnam‐si 13488 Republic of Korea
| | - Keun‐Hong Park
- Laboratory of Nano‐regenerative Medical Engineering Department of Biomedical Science College of Life Science CHA University 618, CHA Biocomplex, Sampyeong‐Dong Bundang‐gu Seongnam‐si 13488 Republic of Korea
| |
Collapse
|
14
|
Bagheri-Mohammadi S. Stem cell-based therapy as a promising approach in Alzheimer's disease: current perspectives on novel treatment. Cell Tissue Bank 2021; 22:339-353. [PMID: 33398492 DOI: 10.1007/s10561-020-09896-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a neuronal disorder with insidious onset and slow progression, leading to growing global concern with huge implications for individuals and society. The occurrence of AD has been increased and has become an important health issue throughout the world. In recent years, the care of more than 35 million patients with AD costs over $ 600 billion per year, it is approximately 1 percent of the global Gross Domestic Product. Currently, the therapeutic approach is not effective for neurological deficits especially after the development of these major neurological disorders. The discovery of the technique called cell-based therapy has shown promising results and made important conclusions beyond AD using the stem cells approach. Here we review recent progress on stem cell-based therapy in the context of AD.
Collapse
Affiliation(s)
- Saeid Bagheri-Mohammadi
- Department of Physiology and Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Department of Physiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran. .,Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
15
|
Yoon SY, Yoon JA, Park M, Shin EY, Jung S, Lee JE, Eum JH, Song H, Lee DR, Lee WS, Lyu SW. Recovery of ovarian function by human embryonic stem cell-derived mesenchymal stem cells in cisplatin-induced premature ovarian failure in mice. Stem Cell Res Ther 2020; 11:255. [PMID: 32586410 PMCID: PMC7318510 DOI: 10.1186/s13287-020-01769-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/07/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Clinical use of mesenchymal stem cells (MSCs) requires a uniform cell population, and their harvesting is invasive and produces a limited number of cells. Human embryonic stem cell-derived MSCs (hESC-MSCs) can differentiate into three germ layers and possess immunosuppressive effects in vitro. Anticancer treatment is a well-known risk factor for premature ovarian failure (POF). In this study, we investigated the effect of hESC-MSC on recovery of ovarian function in cisplatin-induced POF in mice. METHODS Female mice received intraperitoneal cisplatin for 10 days. On day 12, CHA15-derived hESC-MSCs were transplanted into the mice by tail vein injection. An injection of PBS served as the negative control. Ovaries were removed 28 days after transplantation for assessment of ovarian histology, immunostaining, and fertility testing by superovulation and in vitro fertilization. hESC-MSC transplantation into mice with cisplatin-induced damage restored body weight and ovary size. RESULTS Mean primary and primordial follicle counts in the hESC-MSC group were significantly improved compared to the PBS group (P < 0.05), and counts of zona pellucida remnants, an apoptotic sign in ovarian follicles, were significantly reduced (P < 0.05). TUNEL assays and cleaved PARP immunostaining indicated apoptosis, which led to loss of ovarian stromal cells in negative control mice, while Ki-67 was higher in the hESC-MSC group and in non-cisplatin-treated controls than in the PBS group. Ovulation was reduced in the PBS group but recovered significantly in the hESC-MSC group. Rates of blastocyst formation from ovulated eggs and live births per mouse also recovered significantly in the hESC-MSC group. CONCLUSIONS hESC-MSC restored structure and function in the cisplatin-damaged ovary. Our study provides new insights into the great clinical potential of human hESC-MSC in treating POF.
Collapse
Affiliation(s)
- Sook Young Yoon
- Fertility Center of CHA Gangnam Medical Center, CHA University, 569 Nonhyun-ro, Gangnam-Gu, Seoul, 06125, South Korea
| | - Jung Ah Yoon
- Fertility Center of CHA Gangnam Medical Center, CHA University, 569 Nonhyun-ro, Gangnam-Gu, Seoul, 06125, South Korea
| | - Mira Park
- Department of Biomedical Science, CHA University, Seongnam-si, South Korea
| | - Eun-Young Shin
- Department of Biomedical Science, CHA University, Seongnam-si, South Korea
| | - Sookyung Jung
- CHA Advanced Research Institute, Seongnam-si, South Korea
| | - Jeoung Eun Lee
- CHA Advanced Research Institute, Seongnam-si, South Korea
| | - Jin Hee Eum
- Fertility Center of CHA Gangnam Medical Center, CHA University, 569 Nonhyun-ro, Gangnam-Gu, Seoul, 06125, South Korea
| | - Haengseok Song
- Department of Biomedical Science, CHA University, Seongnam-si, South Korea
| | - Dong Ryul Lee
- CHA Advanced Research Institute, Seongnam-si, South Korea.,Department of Biomedical Science, CHA University, Seongnam-si, South Korea
| | - Woo Sik Lee
- Fertility Center of CHA Gangnam Medical Center, CHA University, 569 Nonhyun-ro, Gangnam-Gu, Seoul, 06125, South Korea
| | - Sang Woo Lyu
- Fertility Center of CHA Gangnam Medical Center, CHA University, 569 Nonhyun-ro, Gangnam-Gu, Seoul, 06125, South Korea.
| |
Collapse
|
16
|
Lee JE, Lee JY, Park CH, Eum JH, Jung SK, Han AR, Seol DW, Lee JS, Shin HS, Im JH, Chun T, Ha K, Heo DR, Yoon TK, Lee DR. Cryopreserved Human Oocytes and Cord Blood Cells Can Produce Somatic Cell Nuclear Transfer-Derived Pluripotent Stem Cells with a Homozygous HLA Type. Stem Cell Reports 2020; 15:171-184. [PMID: 32502464 PMCID: PMC7363744 DOI: 10.1016/j.stemcr.2020.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/05/2020] [Accepted: 05/05/2020] [Indexed: 02/08/2023] Open
Abstract
Human pluripotent stem cells (PSCs) through somatic cell nuclear transfer (SCNT) may be an important source for regenerative medicine. The low derivation efficiency of stem cells and the accessibility of human oocytes are the main obstacles to their application. We previously reported that the efficiency of SCNT was increased by overexpression of H3K9me3 demethylase. Here, we applied a modified derivation method to the PSC line and first obtained human SCNT-PSC lines derived from both donated cryopreserved oocytes and cord blood cells with a homozygous human leukocyte antigen (HLA) type. The SCNT-PSCs have very similar characteristics with embryonic stem cells (ESCs) and additionally have shown immunocompatibility in an in vitro and in vivo humanized mouse with a matching HLA type. Our study demonstrates that SCNT technology using donated cryopreserved oocytes and cord blood cells with a known HLA type provides a promising method for establishing a human HLA-matched SCNT-PSC bank for regenerative medicine. Human normal SCNT-PSC line with homozygous HLA type is derived from both donated cryopreserved oocytes and cord blood cells SCNT-PSC derivation is improved by modified method using ESC-conditioned medium Differentiated functional cells from SCNT-PSC with homozygous HLA type have shown immunocompatibility in humanized mouse with a matching HLA type This SCNT technology facilitates the establishment of the human HLA-matched SCNT-PSC bank
Collapse
Affiliation(s)
- Jeoung Eun Lee
- CHA Advanced Research Institute, CHA University, Seongnam, Gyunggi-do 13488, Korea
| | - Ji Yoon Lee
- CHA Advanced Research Institute, CHA University, Seongnam, Gyunggi-do 13488, Korea
| | - Chang-Hwan Park
- Graduated School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Jin Hee Eum
- Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul 06135, Korea
| | - Soo Kyung Jung
- CHA Advanced Research Institute, CHA University, Seongnam, Gyunggi-do 13488, Korea
| | - A-Reum Han
- Department of Biomedical Science, CHA University, Seongnam, Gyunggi-do 13488, Korea
| | - Dong-Won Seol
- Department of Biomedical Science, CHA University, Seongnam, Gyunggi-do 13488, Korea
| | - Jin Saem Lee
- Graduated School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Hyun Soo Shin
- Department of Radiation Oncology, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea
| | - Jung Ho Im
- Department of Radiation Oncology, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea
| | - Taehoon Chun
- Department of Biotechnology, College of Life and Biotechnology, Korea University, Seoul 02841, Korea
| | - Kyungsoo Ha
- New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Korea
| | - Deok Rim Heo
- New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Korea
| | - Tae Ki Yoon
- Department of Biomedical Science, CHA University, Seongnam, Gyunggi-do 13488, Korea
| | - Dong Ryul Lee
- CHA Advanced Research Institute, CHA University, Seongnam, Gyunggi-do 13488, Korea; Department of Biomedical Science, CHA University, Seongnam, Gyunggi-do 13488, Korea.
| |
Collapse
|
17
|
An update on stem cell therapy for Asherman syndrome. J Assist Reprod Genet 2020; 37:1511-1529. [PMID: 32445154 DOI: 10.1007/s10815-020-01801-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/28/2020] [Indexed: 02/06/2023] Open
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.
Collapse
|
18
|
Perivascular Stem Cell-Derived Cyclophilin A Improves Uterine Environment with Asherman's Syndrome via HIF1α-Dependent Angiogenesis. Mol Ther 2020; 28:1818-1832. [PMID: 32534604 DOI: 10.1016/j.ymthe.2020.05.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/22/2020] [Accepted: 05/14/2020] [Indexed: 12/18/2022] Open
Abstract
Asherman's syndrome (AS) is characterized by intrauterine adhesions or fibrosis resulting from scarring inside the endometrium. AS is associated with infertility, recurrent miscarriage, and placental abnormalities. Although mesenchymal stem cells show therapeutic promise for the treatment of AS, the molecular mechanisms underlying its pathophysiology remain unclear. We ascertained that mice with AS, like human patients with AS, suffer from extensive fibrosis, oligo/amenorrhea, and infertility. Human perivascular stem cells (hPVSCs) from umbilical cords repaired uterine damage in mice with AS, regardless of their delivery routes. In mice with AS, embryo implantation is aberrantly deferred, which leads to intrauterine growth restriction followed by no delivery at term. hPVSC administration significantly improved implantation defects and subsequent poor pregnancy outcomes via hypoxia inducible factor 1α (HIF1α)-dependent angiogenesis in a dose-dependent manner. Pharmacologic inhibition of HIF1α activity hindered hPVSC actions on pregnancy outcomes, whereas stabilization of HIF1α activity facilitated such actions. Furthermore, therapeutic effects of hPVSCs were not observed in uterine-specific HIF1α-knockout mice with AS. Secretome analyses of hPVSCs identified cyclophilin-A as the major paracrine factor for hPVSC therapy via HIF1α-dependent angiogenesis. Collectively, we demonstrate that hPVSCs-derived cyclophilin-A facilitates HIF1α-dependent angiogenesis to ameliorate compromised uterine environments in mice with AS, representing the major pathophysiologic features of humans with AS.
Collapse
|
19
|
Kim JH, Park M, Paek JY, Lee WS, Song H, Lyu SW. Intrauterine Infusion of Human Platelet-Rich Plasma Improves Endometrial Regeneration and Pregnancy Outcomes in a Murine Model of Asherman's Syndrome. Front Physiol 2020; 11:105. [PMID: 32116803 PMCID: PMC7033504 DOI: 10.3389/fphys.2020.00105] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022] Open
Abstract
Asherman’s syndrome (AS) is characterized by intrauterine adhesion or fibrosis resulting from damage to the endometrium, often leading to amenorrhea, infertility, or recurrent pregnancy loss. Although various therapeutic strategies for AS have been proposed, the options remain limited. New strategies such as bone marrow-derived mesenchymal stem cell (BM-MSC) therapy aim to potentiate the intrinsic capacity of endometrial regeneration. However, BM-MSC therapy has not been widely adopted mainly because it involves invasive and expensive procedures such as bone marrow biopsy and cell storing. On the other hand, platelet-rich plasma (PRP) is considered safe and affordable because it involves the less invasive procedure of blood collection from peripheral veins to produce PRP. To assess the effectiveness of human PRP infusion for endometrial regeneration, we established a murine model of injury-induced AS and evaluated endometrial morphology, expression of fibrosis-related factors, implantation sites (IS), and pregnancy outcomes associated with human PRP treatment. We found that treatment with human PRP was associated with improved endometrial morphology, reduced degree of fibrosis, and down-regulated expression of fibrosis-related factors in murine model of AS. Furthermore, human PRP treatment was associated with a higher number of IS and live-births. Our results suggest that human PRP treatment may become a valuable strategy to promote the regeneration of damaged endometrium and thus improve fertility and pregnancy outcomes in clinical practice.
Collapse
Affiliation(s)
- Ji Hye Kim
- Department of Obstetrics and Gynecology, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, South Korea
| | - Mira Park
- Department of Biomedical Science, CHA University, Seongnam, South Korea
| | - Jin Young Paek
- Department of Laboratory Medicine, CHA Gangnam Medical Center, CHA University, Seoul, South Korea
| | - Woo-Sik Lee
- Department of Obstetrics and Gynecology, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, South Korea
| | - Haengseok Song
- Department of Biomedical Science, CHA University, Seongnam, South Korea
| | - Sang Woo Lyu
- Department of Obstetrics and Gynecology, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, South Korea
| |
Collapse
|
20
|
Jun SM, Park M, Lee JY, Jung S, Lee JE, Shim SH, Song H, Lee DR. Single cell-derived clonally expanded mesenchymal progenitor cells from somatic cell nuclear transfer-derived pluripotent stem cells ameliorate the endometrial function in the uterus of a murine model with Asherman's syndrome. Cell Prolif 2019; 52:e12597. [PMID: 30896075 PMCID: PMC6536448 DOI: 10.1111/cpr.12597] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/20/2019] [Accepted: 02/12/2019] [Indexed: 12/24/2022] Open
Abstract
Objectives Because primary mesenchymal progenitor cells (adult‐MPCs) have various functions that depend on the tissue origin and donor, de novo MPCs from human pluripotent stem cells (hPSCs) would be required in regenerative medicine. However, the characteristics and function of MPCs derived from reprogrammed hPSCs have not been well studied. Thus, we show that functional MPCs can be successfully established from a single cell‐derived clonal expansion following MPC derivation from somatic cell nuclear transfer‐derived (SCNT)‐hPSCs, and these cells can serve as therapeutic contributors in an animal model of Asherman's syndrome (AS). Materials and methods We developed single cell‐derived clonal expansion following MPC derivation from SCNT‐hPSCs to offer a pure population and a higher biological activity. Additionally, we investigated the therapeutic effects of SCNT‐hPSC‐MPCs in model mice of Asherman's syndrome (AS), which is characterized by synechiae or fibrosis with endometrial injury. Results Their humoral effects in proliferating host cells encouraged angiogenesis and decreased pro‐inflammatory factors via a host‐dependent mechanism, resulting in reduction in AS. We also addressed that cellular activities such as the cell proliferation and population doubling of SCNT‐hPSC‐MPCs resemble those of human embryonic stem cell‐derived MPCs (hESC‐MPCs) and are much higher than those of adult‐MPCs. Conclusions Somatic cell nuclear transfer‐derived‐hPSCs‐MPCs could be an advanced therapeutic strategy for specific diseases in the field of regenerative medicine.
Collapse
Affiliation(s)
- Sung-Min Jun
- CHA Advanced Research Institute, Seongnam, Korea
| | - Mira Park
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Ji Yoon Lee
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | | | | | - Sung Han Shim
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Haengseok Song
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Dong Ryul Lee
- CHA Advanced Research Institute, Seongnam, Korea.,Department of Biomedical Science, CHA University, Seongnam, Korea
| |
Collapse
|