1
|
Izawa H, Xiang C, Ogawa S, Hisanaga I, Yoshimoto T. Amelioration of female menopausal syndrome by intravenous administration of autologous menstrual blood-derived stem cells. Regen Ther 2025; 29:192-201. [PMID: 40225052 PMCID: PMC11992397 DOI: 10.1016/j.reth.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/29/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction Menopausal syndrome is characterized by a wide range of physical and psychological symptoms in women aged 40s-50s as a result of hormonal fluctuations and age-related decline. Various treatments have been used to manage the symptoms, including hormone replacement therapy, but no effective causal therapies have yet been identified. Regenerative medicine has gained considerable attention as a promising approach to age-related problems, and mesenchymal stem cell therapies have been extensively studied. Recently, menstrual blood has emerged as a novel cell source of stem cells, called menstrual blood-derived stem cells (MenSCs), due to its non-invasive, regular and consistent collection from women. In this study, we have investigated the therapeutic potential of intravenous administration of autologous MenSCs on female menopausal syndromes. Methods Menstrual blood was collected from 15 patients aged 30s-60s with ovarian dysfunction using a menstrual cup, and MenSCs were isolated, cultured and expanded. Patients received either 3 × 107 cells or 1 × 108 cells intravenously 1 to 5 times at intervals of more than 1 month. Patient-reported symptoms were assessed using the Simplified Menopausal Index at pre-treatment and after 1, 3, 6, and 12 months, and safety assessments were performed. Serum estradiol and follicle-stimulating hormone levels were also measured by immunoassay. Results Almost all patients who received MenSCs experienced a sharp reduction in menopausal symptoms, including vasomotor, neuropsychiatric, and motor symptoms, one month after the first administration, and these symptoms remained low for 6 months. The Simplified Menopausal Index score was significantly reduced after treatment. The reducing potency of 1 × 108 MenSCs was greater than that of 3 × 107 MenSCs. Patients who received a higher number of MenSCs showed an increasing trend in estradiol levels and a decreasing trend in follicle-stimulating hormone levels. When MenSCs were administered to postmenopausal patients, this trend was more pronounced. Overall, no apparent serious adverse events were observed during these treatments. Conclusions The present results suggest that the administration of MenSCs improved menopausal symptoms and regulated hormonal balance without any serious adverse events. This is the first report on the promising therapeutic potential of cell-based therapy using autologous MenSCs for female menopausal syndrome.
Collapse
Affiliation(s)
- Hiromi Izawa
- Jingu-Gaien Woman Life Clinic, Shibuya-ku, Tokyo, Japan
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Hangzhou, Beijing, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Seiji Ogawa
- Jingu-Gaien Woman Life Clinic, Shibuya-ku, Tokyo, Japan
- Fujita Medical Innovation Center Tokyo, Reproduction Center, Ota-ku, Tokyo, Japan
| | - Ichiro Hisanaga
- Jingu-Gaien Woman Life Clinic, Shibuya-ku, Tokyo, Japan
- Dai Nippon Printing Co., Ltd., Human Engineering Laboratory, Shinjuku-ku, Tokyo, Japan
- Ritsumeikan University, Art Research Center, Kyoto-shi, Kyoto, Japan
| | - Takayuki Yoshimoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
2
|
Sadiasa A, Werkmeister JA, Gurung S, Gargett CE. Steps towards the clinical application of endometrial and menstrual fluid mesenchymal stem cells for the treatment of gynecological disorders. Expert Opin Biol Ther 2025; 25:285-307. [PMID: 39925343 DOI: 10.1080/14712598.2025.2465826] [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: 10/29/2024] [Revised: 01/28/2025] [Accepted: 02/07/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION The human endometrium is a highly regenerative tissue that contains mesenchymal stem/stromal cells (MSCs). These MSCs are sourced via office-based biopsies and menstrual fluid, providing a less invasive and readily available option for cell-based therapies. This review provides an update on endometrial-derived MSCs as a treatment option for gynecological diseases. AREAS COVERED This narrative review covers the characterization and therapeutic mechanisms of endometrium biopsy-derived MSCs (eMSCs) and menstrual fluid-derived mesenchymal stromal cells (MenSCs), highlighting similarities and differences. It also covers studies of their application in preclinical animal models and in clinical trials as potential cell-based therapies for gynecological diseases. EXPERT OPINION eMSCs and MenSCs from a homologous tissue source have the potential to promote regenerative activity as a treatment for gynecological diseases. Both eMSCs and MenSCs demonstrate therapeutic benefits through their paracrine activity in tissue regeneration, immunomodulation, angiogenesis, and mitigating fibrosis. Further research is essential to establish standardized isolation and characterization protocols, particularly for heterogeneous MenSCs, and to fully understand their mechanisms of action. Implementing SUSD2 magnetic bead sorting for purifying eMSCs from endometrial tissues and menstrual fluid is crucial for their use in future cell-based therapies. Optimization of production, storage, and delivery methods will maximize their therapeutic effectiveness.
Collapse
Affiliation(s)
- Alexander Sadiasa
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Jerome A Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Shanti Gurung
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
3
|
Mitranovici MI, Costachescu D, Voidazan S, Munteanu M, Buicu CF, Oală IE, Ivan V, Apostol A, Melinte IM, Crisan A, Pușcașiu L, Micu R. Exploring the Shared Pathogenesis Mechanisms of Endometriosis and Cancer: Stemness and Targeted Treatments of Its Molecular Pathways-A Narrative Review. Int J Mol Sci 2024; 25:12749. [PMID: 39684461 PMCID: PMC11640855 DOI: 10.3390/ijms252312749] [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: 10/08/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Endometriosis is a benign disease but with malignant behavior, sharing numerous features with cancers. Endometriosis is the development of endometrial tissue outside the uterus, with the presence of both glands and stroma. Approximately 10% of women of reproductive age suffer from endometriosis; it involves high social costs and affects the patient's quality of life. In this review, we attempt to capture the pathogenesis mechanisms that are common to endometriosis and cancer based on molecular biology, focusing more on the principle of immunological changes and stemness. Clinical applicability will consist of targeted treatments that represent future directions in these diseases, which impose a burden on the healthcare system. Unlike endometriosis, cancer is a disease with fatal evolution, with conventional treatment based on chemo/radiotherapy. Here, we focus on the niche of personalized treatments that target molecular pathways. Our findings show that, in both pathologies, the resistance to treatments is due to the stemness of the stem cells, which might play a role in the appearance and evolution of both diseases. More research is needed before we can draw firm conclusions.
Collapse
Affiliation(s)
- Melinda-Ildiko Mitranovici
- Department of Obstetrics and Gynecology, Emergency County Hospital Hunedoara, 14 Victoriei Street, 331057 Hunedoara, Romania;
| | - Dan Costachescu
- Department of Orthopedics-Traumatology, Urology, Radiology and Medical Imaging, University of Medicine and Pharmacy Victor Babes, 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Septimiu Voidazan
- Department of Epidemiology, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania; (S.V.); (C.-F.B.); (I.M.M.); (A.C.); (L.P.)
| | - Mihai Munteanu
- Faculty of Electrical Engineering, Technical University, George Baritiu Street, 400394 Cluj-Napoca, Romania;
| | - Corneliu-Florin Buicu
- Department of Epidemiology, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania; (S.V.); (C.-F.B.); (I.M.M.); (A.C.); (L.P.)
| | - Ioan Emilian Oală
- Department of Obstetrics and Gynecology, Emergency County Hospital Hunedoara, 14 Victoriei Street, 331057 Hunedoara, Romania;
| | - Viviana Ivan
- Department VII, Internal Medicine II, Discipline of Cardiology, University of Medicine and Pharmacy Victor Babes, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (V.I.); (A.A.)
| | - Adrian Apostol
- Department VII, Internal Medicine II, Discipline of Cardiology, University of Medicine and Pharmacy Victor Babes, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (V.I.); (A.A.)
| | - Ioana M. Melinte
- Department of Epidemiology, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania; (S.V.); (C.-F.B.); (I.M.M.); (A.C.); (L.P.)
| | - Andrada Crisan
- Department of Epidemiology, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania; (S.V.); (C.-F.B.); (I.M.M.); (A.C.); (L.P.)
| | - Lucian Pușcașiu
- Department of Epidemiology, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania; (S.V.); (C.-F.B.); (I.M.M.); (A.C.); (L.P.)
| | - Romeo Micu
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy Iuliu Hatieganu, 400347 Cluj-Napoca, Romania;
| |
Collapse
|
4
|
Yuan L, Yao L, Ren X, Chen X, Li X, Xu Y, Jin T. Cartilage defect repair in a rat model via a nanocomposite hydrogel loaded with melatonin-loaded gelatin nanofibers and menstrual blood stem cells: an in vitro and in vivo study. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2024; 35:55. [PMID: 39347832 PMCID: PMC11442572 DOI: 10.1007/s10856-024-06820-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 07/26/2024] [Indexed: 10/01/2024]
Abstract
Cartilage damage caused by injuries or degenerative diseases remains a major challenge in the field of regenerative medicine. In this study, we developed a composite hydrogel system for the delivery of melatonin and menstrual blood stem cells (MenSCs) to treat a rat model of cartilage defect. The composite delivery system was produced by incorporation of melatonin into the gelatin fibers and dispersing these fibers into calcium alginate hydrogels. Various characterization methods including cell viability assay, microstructure studies, degradation rate measurement, drug release, anti-inflammatory assay, and radical scavenging assay were used to characterize the hydrogel system. MenSCs were encapsulated within the nanocomposite hydrogel and implanted into a rat model of full-thickness cartilage defect. A 1.3 mm diameter drilled in the femoral trochlea and used for the in vivo study. Results showed that the healing potential of nanocomposite hydrogels containing melatonin and MenSCs was significantly higher than polymer-only hydrogels. Our study introduces a novel composite hydrogel system, combining melatonin and MenSCs, demonstrating enhanced cartilage repair efficacy, offering a promising avenue for regenerative medicine.
Collapse
Affiliation(s)
- Libo Yuan
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China
| | - Ling Yao
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China
| | - Xianzhen Ren
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China
| | - Xusheng Chen
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China
| | - Xu Li
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China
| | - Yongqing Xu
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China.
| | - Tao Jin
- Department of Orthopedics, 920 Hospital of Joint Logistic Support Force, Kunming, China.
| |
Collapse
|
5
|
Chen L, Huang Y, Zhang N, Qu J, Fang Y, Fu J, Yuan Y, Zhang Q, Li H, Wen Z, Yuan L, Chen L, Xu Z, Li Y, Yan H, Izawa H, Li L, Xiang C. Single-cell RNA sequencing reveals reduced intercellular adhesion molecule crosstalk between activated hepatic stellate cells and neutrophils alleviating liver fibrosis in hepatitis B virus transgenic mice post menstrual blood-derived mesenchymal stem cell transplantation. MedComm (Beijing) 2024; 5:e654. [PMID: 39040848 PMCID: PMC11261812 DOI: 10.1002/mco2.654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 07/24/2024] Open
Abstract
Liver fibrosis can cause hepatitis B virus (HBV)-associated hepatocellular carcinoma. Menstrual blood-derived mesenchymal stem cells (MenSCs) can ameliorate liver fibrosis through paracrine. Single-cell RNA sequencing (scRNA-seq) may be used to explore the roadmap of activated hepatic stellate cell (aHSC) inactivation to target liver fibrosis. This study established HBV transgenic (HBV-Tg) mouse model of carbon tetrachloride (CCl4)-induced liver fibrosis and demonstrated that MenSCs migrated to the injured liver to improve serological indices and reduce fibrotic accumulation. RNA-bulk analysis revealed that MenSCs mediated extracellular matrix accumulation and cell adhesion. Liver parenchymal cells and nonparenchymal cells were identified by scRNA-seq in the control, CCl4, and MenSC groups, revealing the heterogeneity of fibroblasts/HSCs. A CellChat analysis revealed that diminished intercellular adhesion molecule (ICAM) signaling is vital for MenSC therapy. Specifically, Icam1 in aHSCs acted on Itgal/Itgb2 and Itgam/Itgb2 in neutrophils, causing decreased adhesion. The expression of Itgal, Itgam, and Itgb2 was higher in CCl4 group than in the control group and decreased after MenSC therapy in neutrophil clusters. The Lcn2, Pglyrp1, Wfdc21, and Mmp8 had high expression and may be potential targets in neutrophils. This study highlights interacting cells, corresponding molecules, and underlying targets for MenSCs in treating HBV-associated liver fibrosis.
Collapse
Affiliation(s)
- Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Research Units of Infectious Disease and MicroecologyChinese Academy of Medical SciencesBeijingChina
| | - Yuqi Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Ning Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jingjing Qu
- Department of Respiratory DiseaseThoracic Disease CentreThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yangxin Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiamin Fu
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Qi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hang Li
- Innovative Precision Medicine (IPM) GroupHangzhouChina
| | - Zuoshi Wen
- Department of CardiologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Li Yuan
- Innovative Precision Medicine (IPM) GroupHangzhouChina
| | - Lu Chen
- Innovative Precision Medicine (IPM) GroupHangzhouChina
| | - Zhenyu Xu
- Innovative Precision Medicine (IPM) GroupHangzhouChina
| | - Yifei Li
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Research Units of Infectious Disease and MicroecologyChinese Academy of Medical SciencesBeijingChina
| | - Huadong Yan
- Infectious Disease DepartmentShulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical CollegeHangzhouChina
| | | | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Research Units of Infectious Disease and MicroecologyChinese Academy of Medical SciencesBeijingChina
- Jinan Microecological Biomedicine Shandong LaboratoryJinanChina
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesNational Medical Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Research Units of Infectious Disease and MicroecologyChinese Academy of Medical SciencesBeijingChina
- Jinan Microecological Biomedicine Shandong LaboratoryJinanChina
| |
Collapse
|
6
|
Bouzid K, Bourdon M, Bartkowski R, Verbanck M, Chapron C, Marcellin L, Batteux F, Santulli P, Doridot L. Menstrual Blood Donation for Endometriosis Research: A Cross-Sectional Survey on Women's Willingness and Potential Barriers. Reprod Sci 2024; 31:1617-1625. [PMID: 38418666 PMCID: PMC11111534 DOI: 10.1007/s43032-024-01481-3] [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: 11/02/2023] [Accepted: 02/07/2024] [Indexed: 03/02/2024]
Abstract
An anonymous online survey in French was used to assess if endometriosis patients would be as ready as unaffected women to donate their menstrual blood for biological research on endometriosis and evaluate potential barriers to such donation. It was distributed in September 2022 by social media and two mailing lists, including a French patient organization. The questionnaire assessed participant age and brief medical history (hormonal contraception, endometriosis diagnosis, type of endometriosis), menstrual experience (menstrual blood abundance, dysmenorrhea), and whether participants would donate menstrual blood. Women who self-declared with an established endometriosis diagnosis versus no endometriosis were compared. Seven hundred seventy-eight women answered the survey. Among women with menstruation (n = 568), 78% are willing to donate menstrual blood for research. Importantly, this proportion was higher in women who declared having an established endometriosis diagnosis (83%, n = 299) compared to self-declared unaffected women (68%, n = 134, p < 0.001). The previous use of a menstrual cup and dysmenorrhea were significantly associated with the willingness to donate menstrual blood, while the use of hormonal contraception was significantly associated with an unwillingness to donate. Only the previous use of the menstrual cup had a predictive value for menstrual blood donation. No significant relationship was observed between menstrual blood donation and age, heavy menstrual bleeding and in endometriosis patients, endometriosis subtypes. In conclusion, women affected or not by endometriosis are largely willing to donate their menstrual blood for research on endometriosis, dysmenorrhea is not a barrier for donation, and women who use a menstrual cup are the more likely to donate.
Collapse
Affiliation(s)
- Kheira Bouzid
- Université Paris Cité, Institut Cochin, Inserm, CNRS, 75014, Paris, France
| | - Mathilde Bourdon
- Université Paris Cité, Institut Cochin, Inserm, CNRS, 75014, Paris, France
- Département de Gynécologie, Obstétrique Et Médecine de La Reproduction, AP-HP, Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France
| | - Roxane Bartkowski
- Université Paris Cité, Institut Cochin, Inserm, CNRS, 75014, Paris, France
| | - Marie Verbanck
- UR 7537 - BioSTM Biostatistique, Traitement et Modélisation des données Biologiques, Faculté de Pharmacie de Paris, Université Paris Cité, F-75270, Paris, France
| | - Charles Chapron
- Université Paris Cité, Institut Cochin, Inserm, CNRS, 75014, Paris, France
- Département de Gynécologie, Obstétrique Et Médecine de La Reproduction, AP-HP, Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France
| | - Louis Marcellin
- Université Paris Cité, Institut Cochin, Inserm, CNRS, 75014, Paris, France
- Département de Gynécologie, Obstétrique Et Médecine de La Reproduction, AP-HP, Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France
| | - Frederic Batteux
- Université Paris Cité, Institut Cochin, Inserm, CNRS, 75014, Paris, France
- Service d'Immunologie Biologique, AP-HP, Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France
| | - Pietro Santulli
- Université Paris Cité, Institut Cochin, Inserm, CNRS, 75014, Paris, France
- Département de Gynécologie, Obstétrique Et Médecine de La Reproduction, AP-HP, Centre Hospitalier Universitaire (CHU) Cochin, 75014, Paris, France
| | - Ludivine Doridot
- Université Paris Cité, Institut Cochin, Inserm, CNRS, 75014, Paris, France.
| |
Collapse
|
7
|
Tsang HF, Cheung YS, Yu CSA, Chan CSS, Wong CBT, Yim KYA, Pei X, Wong SCC. Menstrual Blood as a Diagnostic Specimen for Human Papillomavirus Genotyping and Genital Tract Infection Using Next-Generation Sequencing as a Novel Diagnostic Tool. Diagnostics (Basel) 2024; 14:686. [PMID: 38611599 PMCID: PMC11012019 DOI: 10.3390/diagnostics14070686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/04/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Menstrual blood (MB) is a convenient specimen type that can be self-collected easily and non-invasively by women. This study assessed the potential application of MB as a diagnostic specimen to detect genital tract infections (GTIs) and human papillomavirus (HPV) infections in women. METHOD Genomic DNA was extracted from MB samples. Pacific Bioscience (Pacbio) 16S ribosomal DNA (rDNA) high-fidelity (HiFi) long-read sequencing and HPV PCR were performed. RESULTS MB samples were collected from women with a pathological diagnosis of CIN1, CIN2, CIN3 or HPV infection. The sensitivity and positive predictive value (PPV) of high-risk HPV detection using MB were found to be 66.7%. A shift in vaginal flora and a significant depletion in Lactobacillus spp. in the vaginal microbiota communities were observed in the MB samples using 16S rDNA sequencing. CONCLUSIONS In this study, we demonstrated that MB is a proper diagnostic specimen of consideration for non-invasive detection of HPV DNA and genotyping using PCR and the diagnosis of GTIs using metagenomic next-generation sequencing (mNGS). MB testing is suitable for all women who menstruate and this study has opened up the possibility of the use of MB as a diagnostic specimen to maintain women's health.
Collapse
Affiliation(s)
- Hin-Fung Tsang
- Department of Clinical Laboratory and Pathology, Hong Kong Adventist Hospital, Hong Kong SAR, China
| | - Yui-Shing Cheung
- Department of Obstetrics and Gynaecology, Queen Elizabeth Hospital, Hong Kong SAR, China; (Y.-S.C.); (C.-S.S.C.)
| | - Chi-Shing Allen Yu
- Codex Genetics Limited, Hong Kong SAR, China; (C.-S.A.Y.); (C.-B.T.W.); (K.-Y.A.Y.)
| | - Chung-Sum Sammy Chan
- Department of Obstetrics and Gynaecology, Queen Elizabeth Hospital, Hong Kong SAR, China; (Y.-S.C.); (C.-S.S.C.)
| | - Chi-Bun Thomas Wong
- Codex Genetics Limited, Hong Kong SAR, China; (C.-S.A.Y.); (C.-B.T.W.); (K.-Y.A.Y.)
| | - Kay-Yuen Aldrin Yim
- Codex Genetics Limited, Hong Kong SAR, China; (C.-S.A.Y.); (C.-B.T.W.); (K.-Y.A.Y.)
| | - Xiaomeng Pei
- Department of Applied Biology & Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China;
| | - Sze-Chuen Cesar Wong
- Department of Applied Biology & Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China;
| |
Collapse
|
8
|
Yu L, Wu H, Zeng S, Hu X, Wu Y, Zhou J, Yuan L, Zhang Q, Xiang C, Feng Z. Menstrual blood-derived mesenchymal stem cells combined with collagen I gel as a regenerative therapeutic strategy for degenerated disc after discectomy in rats. Stem Cell Res Ther 2024; 15:75. [PMID: 38475906 DOI: 10.1186/s13287-024-03680-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Annulus fibrosis (AF) defects have been identified as the primary cause of disc herniation relapse and subsequent disc degeneration following discectomy. Stem cell-based tissue engineering offers a promising approach for structural repair. Menstrual blood-derived mesenchymal stem cells (MenSCs), a type of adult stem cell, have gained attention as an appealing source for clinical applications due to their potential for structure regeneration, with ease of acquisition and regardless of ethical issues. METHODS The differential potential of MenSCs cocultured with AF cells was examined by the expression of collagen I, SCX, and CD146 using immunofluorescence. Western blot and ELISA were used to examine the expression of TGF-β and IGF-I in coculture system. An AF defect animal model was established in tail disc of Sprague-Dawley rats (males, 8 weeks old). An injectable gel containing MenSCs (about 1*106/ml) was fabricated and transplanted into the AF defects immediately after the animal model establishment, to evaluate its repairment properties. Disc degeneration was assessed via magnetic resonance (MR) imaging and histological staining. Immunohistochemical analysis was performed to assess the expression of aggrecan, MMP13, TGF-β and IGF-I in discs with different treatments. Apoptosis in the discs was evaluated using TUNEL, caspase3, and caspase 8 immunofluorescence staining. RESULTS Coculturing MenSCs with AF cells demonstrated ability to express collagen I and biomarkers of AF cells. Moreover, the coculture system presented upregulation of the growth factors TGF-β and IGF-I. After 12 weeks, discs treated with MenSCs gel exhibited significantly lower Pffirrmann scores (2.29 ± 0.18), compared to discs treated with MenSCs (3.43 ± 0.37, p < 0.05) or gel (3.71 ± 0.29, p < 0.01) alone. There is significant higher MR index in disc treated with MenSCs gel than that treated with MenSCs (0.51 ± 0.05 vs. 0.24 ± 0.04, p < 0.01) or gel (0.51 ± 0.05 vs. 0.26 ± 0.06, p < 0.01) alone. Additionally, MenSCs gel demonstrated preservation of the structure of degenerated discs, as indicated by histological scoring (5.43 ± 0.43 vs. 9.71 ± 1.04 in MenSCs group and 10.86 ± 0.63 in gel group, both p < 0.01), increased aggrecan expression, and decreased MMP13 expression in vivo. Furthermore, the percentage of TUNEL and caspase 3-positive cells in the disc treated with MenSCs Gel was significantly lower than those treated with gel alone and MenSCs alone. The expression of TGF-β and IGF-I was higher in discs treated with MenSCs gel or MenSCs alone than in those treated with gel alone. CONCLUSION MenSCs embedded in collagen I gel has the potential to preserve the disc structure and prevent disc degeneration after discectomy, which was probably attributed to the paracrine of growth factors of MenSCs.
Collapse
Affiliation(s)
- Li Yu
- Department of Operating room, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Honghao Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shumei Zeng
- Department of gynaecology, Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojian Hu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuxu Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jinhong Zhou
- Department of gynaecology, Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Yuan
- Innovative Precision Medicine (IPM) Group, Hangzhou, Zhejiang, China
| | - Qingqing Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang, China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhiyun Feng
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- , Building 8-2, 58#, Chengzhan Road, Hangzhou, 310003, China.
| |
Collapse
|
9
|
Soto-Mercado V, Mendivil-Perez M, Velez-Pardo C, Jimenez-Del-Rio M. Neuroprotective Effect of Combined Treatment with Epigallocatechin 3-Gallate and Melatonin on Familial Alzheimer's Disease PSEN1 E280A Cerebral Spheroids Derived from Menstrual Mesenchymal Stromal Cells. J Alzheimers Dis 2024; 99:S51-S66. [PMID: 36846998 DOI: 10.3233/jad-220903] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Background Familial Alzheimer's disease (FAD) is caused by mutations in one or more of 3 genes known as AβPP, PSEN1, and PSEN2. There are currently no effective therapies for FAD. Hence, novel therapeutics are needed. Objective To analyze the effect of treatment with a combination of epigallocatechin-3-gallate (EGCG) and Melatonin (N-acetyl-5-methoxytryptamine, aMT) in a cerebral spheroid (CS) 3D in vitro model of PSEN 1 E280A FAD. Methods We developed a CS in vitro model based on menstrual stromal cells derived from wild-type (WT) and mutant PSEN1 E280A menstrual blood cultured in Fast-N-Spheres V2 medium. Results Beta-tubulin III, choline acetyltransferase, and GFAP in both WT and mutant CSs spontaneously expressed neuronal and astroglia markers when grown in Fast-N-Spheres V2 medium for 4 or 11 days. Mutant PSEN1 CSs had significantly increased levels of intracellular AβPP fragment peptides and concomitant appearance of oxidized DJ-1 as early as 4 days, and phosphorylated tau, decreased ΔΨm, and increased caspase-3 activity were observed on Day 11. Moreover, mutant CSs were unresponsive to acetylcholine. Treatment with a combination of EGCG and aMT decreased the levels of all typical pathological markers of FAD more efficiently than did EGCG or aMT alone, but aMT failed to restore Ca2+ influx in mutant CSs and decreased the beneficial effect of EGCG on Ca2+ influx in mutant CSs. Conclusion Treatment with a combination of EGCG and aMT can be of high therapeutic value due to the high antioxidant capacity and anti-amyloidogenic effect of both compounds.
Collapse
Affiliation(s)
- Viviana Soto-Mercado
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia, SIU Medellin, Colombia
| | - Miguel Mendivil-Perez
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia, SIU Medellin, Colombia
| | - Carlos Velez-Pardo
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia, SIU Medellin, Colombia
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia, SIU Medellin, Colombia
| |
Collapse
|
10
|
Zhang S, Yahaya BH, Pan Y, Liu Y, Lin J. Menstrual blood-derived endometrial stem cell, a unique and promising alternative in the stem cell-based therapy for chemotherapy-induced premature ovarian insufficiency. Stem Cell Res Ther 2023; 14:327. [PMID: 37957675 PMCID: PMC10644549 DOI: 10.1186/s13287-023-03551-w] [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: 12/16/2022] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Chemotherapy can cause ovarian dysfunction and infertility since the ovary is extremely sensitive to chemotherapeutic drugs. Apart from the indispensable role of the ovary in the overall hormonal milieu, ovarian dysfunction also affects many other organ systems and functions including sexuality, bones, the cardiovascular system, and neurocognitive function. Although conventional hormone replacement therapy can partly relieve the adverse symptoms of premature ovarian insufficiency (POI), the treatment cannot fundamentally prevent deterioration of POI. Therefore, effective treatments to improve chemotherapy-induced POI are urgently needed, especially for patients desiring fertility preservation. Recently, mesenchymal stem cell (MSC)-based therapies have resulted in promising improvements in chemotherapy-induced ovary dysfunction by enhancing the anti-apoptotic capacity of ovarian cells, preventing ovarian follicular atresia, promoting angiogenesis and improving injured ovarian structure and the pregnancy rate. These improvements are mainly attributed to MSC-derived biological factors, functional RNAs, and even mitochondria, which are directly secreted or indirectly translocated with extracellular vesicles (microvesicles and exosomes) to repair ovarian dysfunction. Additionally, as a novel source of MSCs, menstrual blood-derived endometrial stem cells (MenSCs) have exhibited promising therapeutic effects in various diseases due to their comprehensive advantages, such as periodic and non-invasive sample collection, abundant sources, regular donation and autologous transplantation. Therefore, this review summarizes the efficacy of MSCs transplantation in improving chemotherapy-induced POI and analyzes the underlying mechanism, and further discusses the benefit and existing challenges in promoting the clinical application of MenSCs in chemotherapy-induced POI.
Collapse
Affiliation(s)
- Shenghui Zhang
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Penang, Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Penang, Malaysia
| | - Ying Pan
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, , China
| | - Yanli Liu
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China.
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China.
| |
Collapse
|
11
|
de Pedro MÁ, López E, González-Nuño FM, Pulido M, Álvarez V, Marchena AM, Preußer C, Szymański W, Pogge von Strandmann E, Graumann J, Sánchez-Margallo FM, Casado JG, Gómez-Serrano M. Menstrual blood-derived mesenchymal stromal cells: impact of preconditioning on the cargo of extracellular vesicles as potential therapeutics. Stem Cell Res Ther 2023; 14:187. [PMID: 37507751 PMCID: PMC10386225 DOI: 10.1186/s13287-023-03413-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) have been shown to exert their therapeutic effects through the secretion of broad spectrum of paracrine factors, including extracellular vesicles (EVs). Accordingly, EVs are being pursued as a promising alternative to cell-based therapies. Menstrual blood-derived stromal cells (MenSCs) are a type of MSC that, due to their immunomodulatory and regenerative properties, have emerged as an innovative source. Additionally, new strategies of cell priming may potentially alter the concentration and cargo of released EVs, leading to modification of their biological properties. In this study, we aimed to characterize the EVs released by MenSCs and compare their therapeutic potential under three different preconditioning conditions (proinflammatory stimuli, physioxia, and acute hypoxia). METHODS MenSCs were isolated from five healthy women. Following culturing to 80% confluence, MenSCs were exposed to different priming conditions: basal (21% O2), proinflammatory stimuli (IFNγ and TNFα, 21% O2), physioxia (1-2% O2), and acute hypoxia (< 1% O2) for 48-72 h. Conditioned media from MenSCs was collected after 48 h and EVs were isolated by a combination of ultra-filtration and differential centrifugation. An extensive characterization ranging from nano-flow cytometry (nFC) to quantitative high-throughput shotgun proteomics was performed. Bioinformatics analyses were used to derive hypotheses on their biological properties. RESULTS No differences in the morphology, size, or number of EVs released were detected between priming conditions. The proteome analysis associated with basal MenSC-EVs prominently revealed their immunomodulatory and regenerative capabilities. Furthermore, quantitative proteomic analysis of differentially produced MenSC-EVs provided sufficient evidence for the utility of the differential preconditioning in purpose-tailoring EVs for their therapeutic application: proinflammatory priming enhanced the anti-inflammatory, regenerative and immunomodulatory capacity in the innate response of EVs, physioxia priming also improves tissue regeneration, angiogenesis and their immunomodulatory capacity targeting on the adaptive response, while acute hypoxia priming, increased hemostasis and apoptotic processes regulation in MenSC-EVs, also by stimulating immunomodulation mainly through the adaptive response. CONCLUSIONS Priming of MenSCs under proinflammatory and hypoxic conditions affected the cargo proteome of EVs released, resulting in different therapeutic potential, and thus warrants experimental exploration with the aim to generate better-defined MSC-derived bioproducts.
Collapse
Affiliation(s)
- María Ángeles de Pedro
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
| | - Esther López
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain.
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain.
| | | | - María Pulido
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
| | - Verónica Álvarez
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
| | - Ana María Marchena
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
| | - Christian Preußer
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany
- Core Facility Extracellular Vesicles, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany
| | - Witold Szymański
- Institute of Translational Proteomics, Biochemical/Pharmacological Center, Philipps University, 35043, Marburg, Germany
| | - Elke Pogge von Strandmann
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany
- Core Facility Extracellular Vesicles, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany
| | - Johannes Graumann
- Institute of Translational Proteomics, Biochemical/Pharmacological Center, Philipps University, 35043, Marburg, Germany
| | - Francisco Miguel Sánchez-Margallo
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, 10071, Cáceres, Spain
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
| | - Javier G Casado
- RICORS-TERAV Network, ISCIII, 28029, Madrid, Spain
- Immunology Unit, University of Extremadura, 10003, Cáceres, Spain
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003, Cáceres, Spain
| | - María Gómez-Serrano
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, 35043, Marburg, Germany.
| |
Collapse
|
12
|
Mendivil-Perez M, Velez-Pardo C, Lopera F, Kosik KS, Jimenez-Del-Rio M. PSEN1 E280A Cholinergic-like Neurons and Cerebral Spheroids Derived from Mesenchymal Stromal Cells and from Induced Pluripotent Stem Cells Are Neuropathologically Equivalent. Int J Mol Sci 2023; 24:8957. [PMID: 37240306 PMCID: PMC10218810 DOI: 10.3390/ijms24108957] [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: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurological condition characterized by the severe loss of cholinergic neurons. Currently, the incomplete understanding of the loss of neurons has prevented curative treatments for familial AD (FAD). Therefore, modeling FAD in vitro is essential for studying cholinergic vulnerability. Moreover, to expedite the discovery of disease-modifying therapies that delay the onset and slow the progression of AD, we depend on trustworthy disease models. Although highly informative, induced pluripotent stem cell (iPSCs)-derived cholinergic neurons (ChNs) are time-consuming, not cost-effective, and labor-intensive. Other sources for AD modeling are urgently needed. Wild-type and presenilin (PSEN)1 p.E280A fibroblast-derived iPSCs, menstrual blood-derived menstrual stromal cells (MenSCs), and umbilical cord-derived Wharton Jelly's mesenchymal stromal cells (WJ-MSCs) were cultured in Cholinergic-N-Run and Fast-N-Spheres V2 medium to obtain WT and PSEN 1 E280A cholinergic-like neurons (ChLNs, 2D) and cerebroid spheroids (CSs, 3D), respectively, and to evaluate whether ChLNs/CSs can reproduce FAD pathology. We found that irrespective of tissue source, ChLNs/CSs successfully recapitulated the AD phenotype. PSEN 1 E280A ChLNs/CSs show accumulation of iAPPβ fragments, produce eAβ42, present TAU phosphorylation, display OS markers (e.g., oxDJ-1, p-JUN), show loss of ΔΨm, exhibit cell death markers (e.g., TP53, PUMA, CASP3), and demonstrate dysfunctional Ca2+ influx response to ACh stimuli. However, PSEN 1 E280A 2D and 3D cells derived from MenSCs and WJ-MSCs can reproduce FAD neuropathology more efficiently and faster (11 days) than ChLNs derived from mutant iPSCs (35 days). Mechanistically, MenSCs and WJ-MSCs are equivalent cell types to iPSCs for reproducing FAD in vitro.
Collapse
Affiliation(s)
- Miguel Mendivil-Perez
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, Calle 62#52-59, Building 1, Room 412, SIU, Medellin 050010, Colombia; (M.M.-P.); (C.V.-P.); (F.L.)
| | - Carlos Velez-Pardo
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, Calle 62#52-59, Building 1, Room 412, SIU, Medellin 050010, Colombia; (M.M.-P.); (C.V.-P.); (F.L.)
| | - Francisco Lopera
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, Calle 62#52-59, Building 1, Room 412, SIU, Medellin 050010, Colombia; (M.M.-P.); (C.V.-P.); (F.L.)
| | - Kenneth S. Kosik
- Neuroscience Research Institute, Department of Molecular Cellular Developmental Biology, University of California, Santa Barbara, CA 93106, USA;
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, Calle 62#52-59, Building 1, Room 412, SIU, Medellin 050010, Colombia; (M.M.-P.); (C.V.-P.); (F.L.)
| |
Collapse
|
13
|
Song Y, Li P, Xu Y, Lin Z, Deng Z, Chen C. Menstrual Blood-Derived Mesenchymal Stem Cells Encapsulated in Autologous Platelet-Rich Gel Facilitate Rotator Cuff Healing in a Rabbit Model of Chronic Tears. Am J Sports Med 2023:3635465231168104. [PMID: 37184028 DOI: 10.1177/03635465231168104] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Successful management of chronic rotator cuff (RC) tears remains a challenge owing to its limited intrinsic healing capacity and unsatisfactory failure rate. Menstrual blood-derived mesenchymal stem cells (MenSCs) have the potential to differentiate into the chondrogenic or osteogenic lineage. Autologous platelet-rich gel (APG), a gel material derived from platelet-rich plasma (PRP), can be applied as a carrier system for cell delivery and also as a releasing system for endogenous growth factors. PURPOSE To investigate the effect of human MenSCs encapsulated in APG (MenSCs@APG) on the healing of chronic RC tears in a rabbit model. STUDY DESIGN Controlled laboratory study. METHODS After evaluation of the effect of PRP on MenSC proliferation or differentiation, the stem cells were encapsulated in APG for in vivo injection. Supraspinatus tenotomy from the right greater tuberosity was performed on 45 New Zealand White rabbits. After 6 weeks, these rabbits were randomly allocated to 3 supplemental treatments during supraspinatus repair: saline injection (control [CTL] group), APG injection (APG group), and MenSCs@APG injection (MenSCs@APG group). At week 18, these rabbits were sacrificed to harvest the humerus-supraspinatus tendon complexes for micro-computed tomography (CT), histological evaluation, tensile test, and MenSC tracking. RESULTS In vitro results showed that APG can stimulate MenSC proliferation and enhance chondrogenic or osteogenic differentiation. In vivo results showed that APG can act as a carrier for delivering MenSCs into the healing site, and also as a stimulator for enhancing the in vivo performance of MenSCs. Micro-CT showed that bone volume/total volume and trabecular thickness of the new bone in the MenSCs@APG group presented significantly larger values than those of the APG or CTL group (P < .05 for all). Histologically, compared with the CTL or APG group, significantly more mature fibrocartilage regenerated at the healing site in the MenSCs@APG group. A large number of human nuclei-stained cells were observed in the MenSCs@APG group, presenting a similar appearance to fibrochondrocytes or osteocytes. Biomechanically, the MenSCs@APG group showed significantly higher failure load and stiffness than the APG or CTL group (P < .05 for all). CONCLUSION Human MenSCs@APG facilitated RC healing in a rabbit model of chronic tears. CLINICAL RELEVANCE Autogenous MenSCs@APG may be a new stem cell-based therapy for augmenting RC healing in the clinic.
Collapse
Affiliation(s)
- Ya Song
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ping Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Xu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Zhangyuan Lin
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenhan Deng
- Department of Sports Medicine, First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
| | - Can Chen
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Department of Sports Medicine, First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
| |
Collapse
|
14
|
Zhou C, Zhang B, Yang Y, Jiang Q, Li T, Gong J, Tang H, Zhang Q. Stem cell-derived exosomes: emerging therapeutic opportunities for wound healing. Stem Cell Res Ther 2023; 14:107. [PMID: 37101197 PMCID: PMC10134577 DOI: 10.1186/s13287-023-03345-0] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/14/2023] [Indexed: 04/28/2023] Open
Abstract
Wound healing is a dynamic and highly sequential process involving a series of overlapping spatial and temporal phases, including hemostasis, inflammation, proliferation, and tissue remodeling. Mesenchymal stem cells (MSCs) are multipotent stem cells with self-renewal, multidirectional differentiation potential, and paracrine regulation. Exosomes are subcellular vesicular components 30-150 nm in size and are novel carriers of intercellular communication in regulating the biological behaviors of skin cells. Compared to MSCs, MSC-derived exosomes (MSC-exos) possess lower immunogenicity, easy storage, and highly effective biological activity. MSC-exos, mainly derived from adipose-derived stem cells (ADSCs), bone marrow-derived MSCs (BMSCs), human umbilical cord MSCs (hUC-MSCs), and other stem cell types, play a role in shaping the activity of fibroblasts, keratinocytes, immune cells, and endothelial cells in diabetic wounds, inflammatory wound repair, and even wound-related keloid formation. Therefore, this study focuses on the specific roles and mechanisms of different MSC-exos in wound healing, as well as the current limitations and various perspectives. Deciphering the biological properties of MSC-exos is crucial to providing a promising cell-free therapeutic tool for wound healing and cutaneous regeneration.
Collapse
Affiliation(s)
- Chuchao Zhou
- Department of Plastic Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, 430060, China
| | - Boyu Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Yanqing Yang
- Department of Plastic Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, 430060, China
| | - Qiong Jiang
- Department of Pharmacy, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Tianyu Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Gong
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| | - Hongbo Tang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| |
Collapse
|
15
|
Hojjat A, Mansour RN, Enderami SE, Hassannia H, Mahdavi M, Mellati A, Mehdipour Chari K, Salarinia R, Saburi E. The differentiation and generation of glucose-sensitive beta like-cells from menstrual blood-derived stem cells using an optimized differentiation medium with platelet-rich plasma (PRP). Acta Histochem 2023; 125:152025. [PMID: 37058856 DOI: 10.1016/j.acthis.2023.152025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/16/2023]
Abstract
Regarding their reversible damage of insulin-producing cells (IPCs) and the inefficiency of treatment methods for type 1 diabetes mellitus (T1DM), scientists decided to produce IPCs from an unlimited source of cells. But the production of these cells is constantly faced with problems such as low differentiation efficiency in cell therapy and regenerative medicine. This study provided an ideal differentiation medium enriched with plasma-rich platelet (PRP) delivery to produce IPCs from menstrual blood-derived stem cells (MenSCs). We compared them with and without PRP differentiation medium. MenSCs were then cultured in two experimental groups: with/without PRP differentiation medium and a control group (undifferentiated MenSCs). After 18 days, differentiated cells were analyzed for expression of pancreatic gene markers by real-time PCR. Immunocytochemical staining was used to detect the presence of insulin and Pdx-1 in the differentiated cells, and insulin and C-peptide secretion response to glucose were tested by ELISA. Finally, the morphology of differentiated cells was examined by an inverted microscope. In vitro studies showed that MenSCs differentiated in the PRP differentiation medium had strong properties of IPCs such as pancreatic islet-like structure. The expression of pancreatic markers at both RNA and protein levels showed that the differentiation efficiency was higher in the PRP differentiation medium. In both experimental groups, the differentiated cells were functional and secreted C-peptide and insulin on glucose stimulation, but the secretion of C-peptide and insulin in the PRP group was higher than those cultured in the without PRP differentiation medium. Our findings showed that using of PRP enriched differentiation medium can promote the differentiation of MenSCs into IPCs compared to the without PRP culture group. Therefore, the use of PRP into differentiation media can be proposed as a new approach to producing IPCs from MenSCs and used in cell-based therapies for T1DM.
Collapse
Affiliation(s)
- Atefeh Hojjat
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Islamic Republic of Iran
| | - Reyhaneh Nassiri Mansour
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Islamic Republic of Iran
| | - Seyed Ehsan Enderami
- Immunogenetics Research Center, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Islamic Republic of Iran.
| | - Hadi Hassannia
- Immunogenetics Research Center, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Islamic Republic of Iran
| | - Mohammadreza Mahdavi
- Thalassemia Research Center (TRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Mazandaran, Islamic Republic of Iran
| | - Amir Mellati
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Islamic Republic of Iran
| | - Kayvan Mehdipour Chari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Islamic Republic of Iran
| | - Reza Salarinia
- Department of Advanced Sciences and Technologies, School of Medicine, North Khorasan University of Sciences, Bojnurd, Islamic Republic of Iran
| | - Ehsan Saburi
- Medical Genetics and Molecular Medicine Department, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Islamic Republic of Iran
| |
Collapse
|
16
|
Zhou S, Liu Y, Zhang Q, Xu H, Fang Y, Chen X, Fu J, Yuan Y, Li Y, Yuan L, Xiang C. Human menstrual blood-derived stem cells reverse sorafenib resistance in hepatocellular carcinoma cells through the hyperactivation of mitophagy. Stem Cell Res Ther 2023; 14:58. [PMID: 37005657 PMCID: PMC10068152 DOI: 10.1186/s13287-023-03278-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 03/13/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND Sorafenib is a first-line drug targeting the RTK-MAPK signalling pathway used to treat advanced hepatocellular carcinoma (HCC). However, tumour cells readily develop sorafenib resistance, limiting long-term therapy with this drug. In our previous study, we found that human menstrual blood-derived stem cells (MenSCs) altered the expression of some sorafenib resistance-associated genes in HCC cells. Therefore, we wanted to further explore the feasibility of MenSC-based combination therapy in treating sorafenib-resistant HCC (HCC-SR) cells. METHODS The therapeutic efficiency of sorafenib was determined using CCK-8 (Cell Counting Kit-8), Annexin V/PI and clone formation assays in vitro and a xenograft mouse model in vivo. DNA methylation was determined using RT‒PCR and methylated DNA immunoprecipitation (MeDIP). Autophagy was detected by measuring LC3-II degradation and autophagosome maturation. Transmission electron microscopy identified autophagosomes and mitochondria. Physiological functions of mitochondria were assessed by measuring the ATP content, reactive oxygen species (ROS) generation, and mitochondrial membrane potential (MMP). RESULTS The tumour suppressor genes BCL2 interacting protein 3 (BNIP3) and BCL2 interacting protein 3 like (BNIP3L) were silenced by promoter methylation and that BNIP3 and BNIP3L levels correlated negatively with sorafenib resistance in HCC-SR cells. Strikingly, MenSCs reversed sorafenib resistance. MenSCs upregulated BNIP3 and BNIP3L expression in HCC-SR cells via tet methylcytosine dioxygenase 2 (TET2)-mediated active demethylation. In HCC-SR cells receiving sorafenib and MenSC combination therapy, pressure from sorafenib and elevated BNIP3 and BNIP3L levels disrupted balanced autophagy. Hyperactivation of mitophagy significantly caused severe mitochondrial dysfunction and eventually led to the autophagic death of HCC-SR cells. CONCLUSIONS Our research suggests that combining sorafenib and MenSCs may be a potentially new strategy to reverse sorafenib resistance in HCC-SR cells.
Collapse
Affiliation(s)
- Sining Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yiming Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Laboratory of Cancer Biology, Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Qi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Huikang Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yangxin Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xin Chen
- Department of Haematology, Affiliated Hangzhou First People's Hospital, Zhejiang University, School of Medicine, Hangzhou, 310027, China
| | - Jiamin Fu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yifei Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Li Yuan
- Innovative Precision Medicine (IPM) Group, Hangzhou, 311215, China
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
17
|
Chen K, Zheng S, Fang F. Endometrial Stem Cells and Their Applications in Intrauterine Adhesion. Cell Transplant 2023; 32:9636897231159561. [PMID: 36891869 PMCID: PMC9998408 DOI: 10.1177/09636897231159561] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023] Open
Abstract
Intrauterine adhesion (IUA), resulting from pregnancy or nonpregnant uterine trauma, is one of the major causes of abnormal menstruation, infertility, or repeated pregnancy loss. Although a few methods, including hysteroscopy and hormone therapy, are routinely used for its diagnosis and treatment, they cannot restore tissue regeneration. Stem cells, which have self-renewal and tissue regeneration abilities, have been proposed as a promising therapy for patients with severe IUAs. In this review, we summarize the origin and features of endometrium-associated stem cells and their applications in the treatment of IUAs based on animal models and human clinical trials. We expect that this information will help to elucidate the underlying mechanism for tissue regeneration and to improve the design of stem cell-based therapies for IUAs.
Collapse
Affiliation(s)
- Kai Chen
- Reproductive Medicine Center & Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Wannan Medical College, Wuhu, China
| | - Shengxia Zheng
- Reproductive Medicine Center & Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Fang Fang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
18
|
Mitochondrial Transfer from Menstrual Blood Stromal/Stem Cells Promotes Survival of Cardiomyocytes Following Myocardial Infarction. Avicenna J Med Biotechnol 2022; 14:321-322. [PMID: 36504566 PMCID: PMC9706244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/20/2022] [Indexed: 12/15/2022] Open
|
19
|
Penariol LBC, Thomé CH, Tozetti PA, Paier CRK, Buono FO, Peronni KC, Orellana MD, Covas DT, Moraes MEA, Silva WA, Rosa-e-Silva JC, Ferriani RA, Faça VM, Poli-Neto OB, Tiezzi DG, Meola J. What Do the Transcriptome and Proteome of Menstrual Blood-Derived Mesenchymal Stem Cells Tell Us about Endometriosis? Int J Mol Sci 2022; 23:11515. [PMID: 36232817 PMCID: PMC9570451 DOI: 10.3390/ijms231911515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/19/2022] [Accepted: 09/22/2022] [Indexed: 11/21/2022] Open
Abstract
Given the importance of menstrual blood in the pathogenesis of endometriosis and the multifunctional roles of menstrual mesenchymal stem cells (MenSCs) in regenerative medicine, this issue has gained prominence in the scientific community. Moreover, recent reviews highlight how robust the integrated assessment of omics data are for endometriosis. To our knowledge, no study has applied the multi-omics approaches to endometriosis MenSCs. This is a case-control study at a university-affiliated hospital. MenSCs transcriptome and proteome data were obtained by RNA-seq and UHPLC-MS/MS detection. Among the differentially expressed proteins and genes, we emphasize ATF3, ID1, ID3, FOSB, SNAI1, NR4A1, EGR1, LAMC3, and ZFP36 genes and MT2A, TYMP, COL1A1, COL6A2, and NID2 proteins that were already reported in the endometriosis. Our functional enrichment analysis reveals integrated modulating signaling pathways such as epithelial-mesenchymal transition (↑) and PI3K signaling via AKT to mTORC1 (↓ in proteome), mTORC1 signaling, TGF beta signaling, TNFA signaling via NFkB, IL6 STAT3 signaling, and response to hypoxia via HIF1A targets (↑ in transcriptome). Our findings highlight primary changes in the endometriosis MenSCs, suggesting that the chronic inflammatory endometrial microenvironment can modulate these cells, providing opportunities for endometriosis etiopathogenesis. Moreover, they identify challenges for future research leveraging knowledge for regenerative and precision medicine in endometriosis.
Collapse
Affiliation(s)
- Letícia B. C. Penariol
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Carolina H. Thomé
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- Regional Blood Center, Medical School of Hemocenter Foundation of Ribeirão Preto, University of São Paulo, São Paulo 14051-140, Brazil
| | - Patrícia A. Tozetti
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Carlos R. K. Paier
- Drug Research and Development Center, Federal University of Ceara, Ceará 60430-275, Brazil
| | - Fabiana O. Buono
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Kamila C. Peronni
- Department of Genetics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Maristela D. Orellana
- Regional Blood Center, Medical School of Hemocenter Foundation of Ribeirão Preto, University of São Paulo, São Paulo 14051-140, Brazil
| | - Dimas T. Covas
- Regional Blood Center, Medical School of Hemocenter Foundation of Ribeirão Preto, University of São Paulo, São Paulo 14051-140, Brazil
| | - Maria E. A. Moraes
- Drug Research and Development Center, Federal University of Ceara, Ceará 60430-275, Brazil
| | - Wilson A. Silva
- Department of Genetics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Júlio C. Rosa-e-Silva
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- Laboratory for Translational Data Science, Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Rui A. Ferriani
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- National Institute of Hormones and Women’s Health (Hormona), CNPq, Porto Alegre 90035-003, Brazil
| | - Vitor M. Faça
- Regional Blood Center, Medical School of Hemocenter Foundation of Ribeirão Preto, University of São Paulo, São Paulo 14051-140, Brazil
- Department Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Omero B. Poli-Neto
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- Laboratory for Translational Data Science, Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Daniel G. Tiezzi
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- Laboratory for Translational Data Science, Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Juliana Meola
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- Laboratory for Translational Data Science, Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- National Institute of Hormones and Women’s Health (Hormona), CNPq, Porto Alegre 90035-003, Brazil
| |
Collapse
|
20
|
The Double Engines and Single Checkpoint Theory of Endometriosis. Biomedicines 2022; 10:biomedicines10061403. [PMID: 35740424 PMCID: PMC9219825 DOI: 10.3390/biomedicines10061403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/27/2022] [Accepted: 06/07/2022] [Indexed: 12/12/2022] Open
Abstract
Endometriosis is a chronic disease characterized by the ectopic localization of the endometrial tissue in the peritoneal cavity. Consequently, it causes local pathological changes and systemic symptoms, affecting at least one in every ten women. This disease is difficult to diagnose early, it is prone to dissemination, is difficult to eradicate, tends to recur, and is regarded as “a cancer of no kill”. Indeed, the development of endometriosis closely resembles that of cancer in the way of mutagenesis, pelvic spreading, and immunological adaptation. While retrograde menstruation has been regarded as the primary cause of endometriosis, the role of ovulation and menstrual stimuli in the development of endometriosis has long been overlooked. The development of ovarian and peritoneal endometrioses, similar to the development of high-grade serous carcinoma in the fallopian tube fimbriae with intraperitoneal metastasis, depends highly on the carcinogens released during ovulation. Moreover, endometriosis carries an extremely hypermutated genome, which is non-inferior to the ultra-mutated endometrial cancer. The hypermutation would lead to an overproduction of new proteins or neoantigens. Because of this, the developing endometriosis may have to turn on the PD-1/PDL-1 “self-tolerance” checkpoint to evade immune surveillance, leaving an Achilles tendon for an immune checkpoint blockade. In this review, we present the double engines and single checkpoint theory of the genesis of endometriosis, provide the current pieces of evidence supporting the hypothesis, and discuss the new directions of prevention and treatment.
Collapse
|
21
|
Sahraei SS, Kowsari A, Asl FD, Sheykhhasan M, Naserpoor L, Sheikholeslami A. Evaluating the effect of conditioned medium from endometrial stem cells on endometriosis-derived endometrial stem cells. Anat Cell Biol 2022; 55:100-108. [PMID: 35082175 PMCID: PMC8968229 DOI: 10.5115/acb.21.169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 02/08/2023] Open
Abstract
Endometriosis is a common, benign gynecological disease which is determined as an overspreading of endometrial tissue in exterior region of the uterine cavity. Evidence suggests that retrograde menstrual blood which contains mesenchymal stem cells with differential gene expression compared to healthy women may play a role in endometriosis creation. We aimed to identify whether the conditioned medium (CM) from menstrual blood-derived mesenchymal stem cells (MenSCs) of healthy women can affect the expression level of inflammatory and stemness genes of MenSCs from endometriosis women. Endometriosis-derived MenSCs (E-MenSCs) were treated with CM derived from healthy women's MenSCs (non-endometriosis derived MenSCs [NE-MenSCs]). Some CD markers were analyzed by flow cytometer before and after treatment compared with NE-MenSCs, and the expression level of inflammatory and stemness genes was evaluated by real-time PCR. E-MenSCs show different morphology in vitro culture in comparison with NE-MenSCs, which were changed in the presence of CM, into a morphology more similar to normal cells and showed significant decrease expression of CD10 after CM treatment. In our results, the interleukin-1, cyclooxygenase-2, and hypoxia-inducible factor 1α as inflamaturay genes and octamer-binding transcription factor 4, NANOG, and sex determining region Y-box 2 as stemness genes showed significantly different expression level in E-MenSCs after treating with CM. Our study indicates that the expression level of some inflammatory- and stemness-related genes which have differential expression in E-MenSCs compared with NEMenSCs, could be changed to normal status by using CM derived from NE-MenSCs.
Collapse
Affiliation(s)
- Seyedeh Saeideh Sahraei
- Department of Mesenchymal Stem Cells, The Academic Centre for Education, Culture and Research, Qom, Iran
- Department of Reproductive Biology, The Academic Centre for Education, Culture and Research, Qom, Iran
| | - Ali Kowsari
- Department of Mesenchymal Stem Cells, The Academic Centre for Education, Culture and Research, Qom, Iran
| | - Faezeh Davoodi Asl
- Department of Mesenchymal Stem Cells, The Academic Centre for Education, Culture and Research, Qom, Iran
| | - Mohsen Sheykhhasan
- Department of Mesenchymal Stem Cells, The Academic Centre for Education, Culture and Research, Qom, Iran
| | - Leila Naserpoor
- Department of Reproductive Biology, The Academic Centre for Education, Culture and Research, Qom, Iran
| | - Azar Sheikholeslami
- Department of Mesenchymal Stem Cells, The Academic Centre for Education, Culture and Research, Qom, Iran
| |
Collapse
|
22
|
Yan Y, Wang X, Zhu G. Endometrium Derived Stem Cells as Potential Candidates in Nervous System Repair. Ann Biomed Eng 2022; 50:485-498. [PMID: 35235077 DOI: 10.1007/s10439-022-02909-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/01/2022] [Indexed: 11/24/2022]
Abstract
Limited cell division and lack of endogenous repair mechanisms in the central nervous system, hampers tissue repair following neurodegenerative diseases or tissue injuries. Unlike central nervous system; peripheral nervous system has some capacity to repair after injury, but in case of critical sized defects the use of supporting cells in the neural guidance channels seems inevitable to obtain a satisfactory functional recovery. Stem cell therapies have provided new frontiers in the repair of nervous system largely through paracrine secretion mechanisms. The therapeutic potential of stem cells differs according to their tissue of origin, mode of isolation, administration route, and passage number. During the past decades, studies have been focused on stem cells harvested from disposable tissues such as menstrual blood or biopsies from endometrium. These cells are characterized by their high differentiation and proliferation potential, ease of harvest, and lack of ethical concerns. In the current review, we will discuss the prospects and challenges of endometrial stem cells' application in nervous system repair.
Collapse
Affiliation(s)
- Yifen Yan
- Department of Gynecology, Renmin Hospital, Hubei University of Medicine, Maojian District, No. 39, Chaoyang Zhong Road, Shiyan City, 442000, Hubei Province, China
| | - Xiaoli Wang
- Department of Gynecology, Renmin Hospital, Hubei University of Medicine, Maojian District, No. 39, Chaoyang Zhong Road, Shiyan City, 442000, Hubei Province, China
| | - Guijuan Zhu
- Department of Gynecology, Renmin Hospital, Hubei University of Medicine, Maojian District, No. 39, Chaoyang Zhong Road, Shiyan City, 442000, Hubei Province, China.
| |
Collapse
|
23
|
Gao M, Yu Z, Yao D, Qian Y, Wang Q, Jia R. Mesenchymal stem cells therapy: A promising method for the treatment of uterine scars and premature ovarian failure. Tissue Cell 2021; 74:101676. [PMID: 34798583 DOI: 10.1016/j.tice.2021.101676] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/12/2022]
Abstract
Both intrauterine adhesions (IUA) and premature ovarian failure (POF) have plagued women all over the world for a long time. It is well known that all invasive operations involving the uterus can disrupt its structural and functional integrity to a varying degree, which inevitably lead to abnormal scar formation, such as IUA, also known as Asherman's syndrome with symptoms like hypomenorrhea or infertility. Another reproductive disorder that causes infertility is primary ovarian insufficiency (POI) or POF, which is a degenerative phenomenon in the ovary among women under the age of 40. In recent years, various types of stem cells, especially mesenchymal stem cells (MSCs) have been widely used in reproductive medicine due to their properties, such as immunoregulation, anti-inflammation, angiogenesis, anti-apoptosis, and trophicity. However, the extensive clinical application of cell therapy is impeded by their safety, cost, and manufacturing. In this review, we sought to summarize the recent advances in using different types of MSCs in treating uterine scars and POF. We also describe several biological pathways and molecules involved in animal studies and clinical application; extracellular vesicles secreted by MSCs may be a promising attractive tool to ensure the treatment of infertility by restoring normal reproductive function.
Collapse
Affiliation(s)
- Mingming Gao
- Fourth Clinical Medicine College, Nanjing Medical University, Nanjing, Jiangsu Province, China; Department of Obstetrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China
| | - Zhaoer Yu
- Fourth Clinical Medicine College, Nanjing Medical University, Nanjing, Jiangsu Province, China; Department of Obstetrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China
| | - Dan Yao
- Fourth Clinical Medicine College, Nanjing Medical University, Nanjing, Jiangsu Province, China; Department of Obstetrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China
| | - Yating Qian
- Department of Obstetrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China
| | - Qi Wang
- Department of Obstetrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China
| | - Ruizhe Jia
- Department of Obstetrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China.
| |
Collapse
|
24
|
Liu Y, Zhang Z, Yang F, Wang H, Liang S, Wang H, Yang J, Lin J. The role of endometrial stem cells in the pathogenesis of endometriosis and their application to its early diagnosis†. Biol Reprod 2021; 102:1153-1159. [PMID: 31965165 DOI: 10.1093/biolre/ioaa011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/21/2019] [Accepted: 01/16/2020] [Indexed: 12/14/2022] Open
Abstract
Pelvic pain, infertility, and a high postoperative recurrence rate are associated with endometriosis and adversely affect the physical and mental health of patients. Moreover, these factors place a heavy burden on families and society. The identification of endometrial stem cells (EnSCs) in the eutopic endometrium, menstrual blood, and ectopic lesions of women with endometriosis not only provides new research objects in the context of endometriosis but also promotes and improves our understanding of its pathogenesis. Furthermore, based on previous studies, we reasonably suppose that dysfunctions of eutopic EnSCs play a critical role in the onset of endometriosis and directly cause abnormalities in the endometrium; subsequently, retrograde menstruation facilitates the delivery of abnormal endometrial tissues to the ovaries and pelvic cavity, where they ectopically implant, grow, and form ectopic lesions. Additionally, as a chronically progressive disease, there is a delay (3-11 years) from the first onset of symptoms to the diagnosis of endometriosis. Therefore, the development of a method for early diagnosis with high sensitivity and specificity is essential for endometriosis patients and has the potential to enable early treatment, prevent endometriosis progression, and relieve pain in patients. Thus, focusing on EnSCs will contribute to clarifying the potential pathogenesis of endometriosis and provide support for the application of EnSCs as therapeutic and early diagnostic targets in endometriosis treatment. SUMMARY SENTENCE Focusing on endometrial stem cells (EnSCs) will contribute to clarifying the potential pathogenesis of endometriosis and provide support for the application of EnSCs as therapeutic and early diagnostic targets in endometriosis treatment.
Collapse
Affiliation(s)
- Yanli Liu
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Zhiqin Zhang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Fen Yang
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China.,College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Hongmei Wang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Shengying Liang
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Huiling Wang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jun Yang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
25
|
Chen L, Qu J, Mei Q, Chen X, Fang Y, Chen L, Li Y, Xiang C. Small extracellular vesicles from menstrual blood-derived mesenchymal stem cells (MenSCs) as a novel therapeutic impetus in regenerative medicine. Stem Cell Res Ther 2021; 12:433. [PMID: 34344458 PMCID: PMC8330084 DOI: 10.1186/s13287-021-02511-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/11/2021] [Indexed: 02/07/2023] Open
Abstract
Menstrual blood-derived mesenchymal stem cells (MenSCs) have great potential in regenerative medicine. MenSC has received increasing attention owing to its impressive therapeutic effects in both preclinical and clinical trials. However, the study of MenSC-derived small extracellular vesicles (EVs) is still in its initial stages, in contrast to some common MSC sources (e.g., bone marrow, umbilical cord, and adipose tissue). We describe the basic characteristics and biological functions of MenSC-derived small EVs. We also demonstrate the therapeutic potential of small EVs in fulminant hepatic failure, myocardial infarction, pulmonary fibrosis, prostate cancer, cutaneous wound, type-1 diabetes mellitus, aged fertility, and potential diseases. Subsequently, novel hotspots with respect to MenSC EV-based therapy are proposed to overcome current challenges. While complexities regarding the therapeutic potential of MenSC EVs continue to be unraveled, advances are rapidly emerging in both basic science and clinical medicine. MenSC EV-based treatment has great potential for treating a series of diseases as a novel therapeutic strategy in regenerative medicine.
Collapse
Affiliation(s)
- Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China
| | - Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China
| | - Quanhui Mei
- Department of Intensive Care Unit, The First People's Hospital of Changde City, Changde, Hunan, 415000, People's Republic of China
| | - Xin Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China
| | - Yangxin Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China
| | - Lu Chen
- Innovative Precision Medicine (IPM) Group, Hangzhou, Zhejiang, 311215, People's Republic of China
| | - Yifei Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China.
| |
Collapse
|
26
|
Lv Q, Wang L, Luo X, Chen X. Adult stem cells in endometrial regeneration: Molecular insights and clinical applications. Mol Reprod Dev 2021; 88:379-394. [PMID: 34014590 PMCID: PMC8362170 DOI: 10.1002/mrd.23476] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 03/23/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
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.
Collapse
Affiliation(s)
- Qiaoying Lv
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan UniversityShanghaiChina
| | - Lulu Wang
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan UniversityShanghaiChina
| | - Xuezhen Luo
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan UniversityShanghaiChina
| | - Xiaojun Chen
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan UniversityShanghaiChina
| |
Collapse
|
27
|
Uzieliene I, Bagdonas E, Hoshi K, Sakamoto T, Hikita A, Tachtamisevaite Z, Rakauskiene G, Kvederas G, Mobasheri A, Bernotiene E. Different phenotypes and chondrogenic responses of human menstrual blood and bone marrow mesenchymal stem cells to activin A and TGF-β3. Stem Cell Res Ther 2021; 12:251. [PMID: 33926568 PMCID: PMC8082646 DOI: 10.1186/s13287-021-02286-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Due to its low capacity for self-repair, articular cartilage is highly susceptible to damage and deterioration, which leads to the development of degenerative joint diseases such as osteoarthritis (OA). Menstrual blood-derived mesenchymal stem/stromal cells (MenSCs) are much less characterized, as compared to bone marrow mesenchymal stem/stromal cells (BMMSCs). However, MenSCs seem an attractive alternative to classical BMMSCs due to ease of access and broader differentiation capacity. The aim of this study was to evaluate chondrogenic differentiation potential of MenSCs and BMMSCs stimulated with transforming growth factor β (TGF-β3) and activin A. METHODS MenSCs (n = 6) and BMMSCs (n = 5) were isolated from different healthy donors. Expression of cell surface markers CD90, CD73, CD105, CD44, CD45, CD14, CD36, CD55, CD54, CD63, CD106, CD34, CD10, and Notch1 was analyzed by flow cytometry. Cell proliferation capacity was determined using CCK-8 proliferation kit and cell migration ability was evaluated by scratch assay. Adipogenic differentiation capacity was evaluated according to Oil-Red staining and osteogenic differentiation according to Alizarin Red staining. Chondrogenic differentiation (activin A and TGF-β3 stimulation) was investigated in vitro and in vivo (subcutaneous scaffolds in nude BALB/c mice) by expression of chondrogenic genes (collagen type II, aggrecan), GAG assay and histologically. Activin A protein production was evaluated by ELISA during chondrogenic differentiation in monolayer culture. RESULTS MenSCs exhibited a higher proliferation rate, as compared to BMMSCs, and a different expression profile of several cell surface markers. Activin A stimulated collagen type II gene expression and glycosaminoglycan synthesis in TGF-β3 treated MenSCs but not in BMMSCs, both in vitro and in vivo, although the effects of TGF-β3 alone were more pronounced in BMMSCs in vitro. CONCLUSION These data suggest that activin A exerts differential effects on the induction of chondrogenic differentiation in MenSCs vs. BMMSCs, which implies that different mechanisms of chondrogenic regulation are activated in these cells. Following further optimization of differentiation protocols and the choice of growth factors, potentially including activin A, MenSCs may turn out to be a promising population of stem cells for the development of cell-based therapies with the capacity to stimulate cartilage repair and regeneration in OA and related osteoarticular disorders.
Collapse
Affiliation(s)
- Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406, Vilnius, Lithuania
| | - Edvardas Bagdonas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406, Vilnius, Lithuania
| | - Kazuto Hoshi
- Department of Sensory and Motor System Medicine, Department of Oral-maxillofacial Surgery, Dentistry and Orthodontics, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan.,Department of Tissue Engineering, the University of Tokyo Hospital, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tomoaki Sakamoto
- Department of Sensory and Motor System Medicine, Department of Oral-maxillofacial Surgery, Dentistry and Orthodontics, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Atsuhiko Hikita
- Department of Tissue Engineering, the University of Tokyo Hospital, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Zivile Tachtamisevaite
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406, Vilnius, Lithuania
| | - Greta Rakauskiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406, Vilnius, Lithuania
| | | | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406, Vilnius, Lithuania.,Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, FI-90014, Oulu, Finland.,Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 508 GA, Utrecht, The Netherlands.,Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406, Vilnius, Lithuania.
| |
Collapse
|
28
|
Dolmans MM, Donnez J, Cacciottola L. Fertility Preservation: The Challenge of Freezing and Transplanting Ovarian Tissue. Trends Mol Med 2020; 27:777-791. [PMID: 33309205 DOI: 10.1016/j.molmed.2020.11.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/06/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023]
Abstract
Cancer treatments are increasingly effective, but can result in iatrogenic premature ovarian insufficiency. Ovarian tissue cryopreservation is the only option available to preserve fertility in prepubertal girls and young women who require immediate chemotherapy. Ovarian tissue transplantation has been shown to restore hormonal cycles and fertility, but a large proportion of the follicle reserve is lost as a consequence of exposure to hypoxia. Another crucial concern is the risk of reimplanting malignant cells together with the grafted tissue. In this review, the authors advance some challenging propositions, from prevention of chemotherapy-related gonadotoxicity to ovarian tissue cryopreservation and transplantation, including the artificial ovary approach.
Collapse
Affiliation(s)
- Marie-Madeleine Dolmans
- Gynecology Department, Cliniques universitaires St-Luc, Brussels, Belgium; Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.
| | - Jacques Donnez
- Prof. Em. Catholic University of Louvain, Brussels, Belgium; Société de Recherche pour l'Infertilité (SRI), Brussels, Belgium
| | - Luciana Cacciottola
- Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
29
|
Treatment potential of bone marrow-derived stem cells in women with diminished ovarian reserves and premature ovarian failure. Curr Opin Obstet Gynecol 2020; 31:156-162. [PMID: 30855290 DOI: 10.1097/gco.0000000000000531] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW We review the techniques recently tested in both animal models and humans to provide a state-of-the-art on adult stem cell ovarian transplant to achieve ovarian rejuvenation in patients with diminished ovarian reserves. RECENT FINDINGS As the firsts reports of spontaneous pregnancies achieved after bone marrow transplantation in oncologic women with primary ovarian insufficiency, increasing evidence supports the regenerative effects of stem cell-based therapies in the ovarian niche. Adult stem cells from several origins promote follicular development, increase ovarian local vascularization, increase follicle and stromal cell proliferation and reduce cell apoptosis and follicular atresia, although they do not modify embryo quality. Therefore, residual quiescent follicles of aged or damaged ovaries might produce competent oocytes in an adequate ovarian environment. Nevertheless, further research is needed to properly evaluate underlying mechanisms, identify best cell sources and design less invasive infusion techniques. SUMMARY Stem cells may be a relevant therapeutic alternative for ovary regeneration and follicular development in patients with impaired ovaries, such as poor ovarian responders or women diagnosed with primary ovarian insufficiency.
Collapse
|
30
|
Noory P, Navid S, Zanganeh BM, Talebi A, Borhani-Haghighi M, Gholami K, Manshadi MD, Abbasi M. Human Menstrual Blood Stem Cell-Derived Granulosa Cells Participate in Ovarian Follicle Formation in a Rat Model of Premature Ovarian Failure In Vivo. Cell Reprogram 2020; 21:249-259. [PMID: 31596622 DOI: 10.1089/cell.2019.0020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We recently reported the application of human menstrual blood stem cells' (HuMenSCs) transplantation as a treatment modality in a rat model of premature ovarian failure (POF). We continued to investigate further in this respect. Female rats were injected intraperitoneally with 36 mg/kg busulfan. HuMenSCs were obtained, grown, and analyzed for immunophenotypic features at passage three. The cells were labeled with CM-Dil and infused into the rats. There were four groups: normal, negative control, treatment, and Sham. One month after treatment, the ovaries were collected and weighed. Histological sections were prepared from the ovary and HuMenSCs were tracking. Subsequently, we examined the changes of expression of Bax and B cell lymphoma 2 (Bcl2) genes by real-time polymerase chain reaction assay. One month after HuMenSCs transplantation, these cells were located in the ovarian interstitium and granulosa cells (GCs). The number of TUNEL-positive cells significantly decreased in the treatment group. Also the expression level of Bax genes, unlike Bcl2 gene, significantly decreased compared with negative and sham groups. In our study, HuMenSCs were tracked in ovarian tissues within 2 months after transplantation, and they differentiated into GCs. Therefore, the use of these cells can be a practical and low-cost method for the treatment of POF patients.
Collapse
Affiliation(s)
- Parastoo Noory
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shadan Navid
- Department of Anatomy, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Bagher Minaee Zanganeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Talebi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.,Clinical Research Development Unit, Bahar Hospital, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Maryam Borhani-Haghighi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Keykavos Gholami
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Marjan Dehghan Manshadi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Abbasi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Gonçalves MF, Asensi KD, Nascimento ALL, de Barros JHO, Santos RDA, Andrade CBVD, Kasai-Brunswick TH, Frajblat M, Ortiga-Carvalho TM, Goldenberg RCDS. Human Menstrual Blood-Derived Mesenchymal Cells Improve Mouse Embryonic Development. Tissue Eng Part A 2020; 26:769-779. [PMID: 32493133 DOI: 10.1089/ten.tea.2020.0034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
There is a constant need for improving embryo culture conditions in assisted reproduction. One possibility is to use mesenchymal stem/stromal cells derived from menstrual blood (mbMSCs), with an endometrial origin. In this study, we sought to analyze the expansion of mouse embryos in a direct coculture model with mbMSCs. Our results showed that after five passages, mbMSCs presented a spindle-shaped morphology, with surface markers that were comparable with the normal mesenchymal cell phenotype. mbMSCs could differentiate into adipogenic and osteogenic lineages and secrete angiopoetin-2 and hepatocyte growth factor. The coculture experiments employed 103 two-cell-stage embryos that were randomly divided into two groups: control (n = 50), embryos cultured in GV-Blast medium, and cocultured mbMSCs (n = 53), embryos cocultured with GV-Blast and mbMSCs. Typically, two to three embryos were placed in a well with 200 μL of culture medium and observed until developmental day 5. After 5 days, the cocultured group had more embryos in the blastocyst stage (69.8%) when compared with the control group (30%) (p < 0.001). It was also found that nearly 57% of blastocysts in the cocultured group reached the hatching stage, while only 13% achieved this stage in the control group (p < 0.001). Analyses of cultured mbMSCs and growth media, in the presence or absence of an embryo, were also performed. Immunofluorescence detected similar levels of collagen I and III and fibronectin in both mbMSCs and cocultured mbMSCs, and similar amounts of growth factors, VEGF, PDGF-AA, and PDGF-BB, were also observed in the conditioned medium, regardless of embryo presence. The present study describes, for the first time, an easy, noninvasive, and autologous method that could potentially increase blastocyst growth rates during assisted reproductive procedures (i.e., in vitro fertilization). It is proposed that this mbMSC coculture strategy enriches the embryonic microenvironment and promotes embryo development. This technique may complement or replace existing assisted reproduction methods and is directly relevant to the field of personalized medicine. Impact statement The study demonstrates a novel and potentially personalized assisted reproduction approach. The search for alternative and autologous methods provides assisted reproduction patients with a better chance of a successful pregnancy. In this study, mesenchymal cells derived from menstrual blood resembled the outside uterine surface and could potentially be employed for improving embryo outgrowth. Our protocol enriches the embryonic microenvironment and facilitates high-quality single-embryo transfer.
Collapse
Affiliation(s)
| | - Karina Dutra Asensi
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anna Luiza Lima Nascimento
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Rosana de Almeida Santos
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Tais Hanae Kasai-Brunswick
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcel Frajblat
- Health Sciences Center, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tania M Ortiga-Carvalho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Regina Coeli Dos Santos Goldenberg
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Chang QY, Zhang SW, Li PP, Yuan ZW, Tan JC. Safety of menstrual blood-derived stromal cell transplantation in treatment of intrauterine adhesion. World J Stem Cells 2020; 12:368-380. [PMID: 32547685 PMCID: PMC7280865 DOI: 10.4252/wjsc.v12.i5.368] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/03/2020] [Accepted: 04/23/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Intrauterine adhesion (IUA) can cause serious damage to women's reproductive health, yet current treatment methods are difficult to achieve satisfactory results. In our previous studies, we demonstrated that menstrual-derived stromal stem cells (MenSCs), with high proliferative capacity and self-renewal ability, have a powerful therapeutic effect in patients with severe IUA. However, safety assessment of MenSCs transplantation is essential for its further application.
AIM To evaluate the short-, medium-, and long-term biosafety of MenSCs via intrauterine transplantation in a rat model of IUA, with a focus on toxicity and tumorigenicity.
METHODS MenSCs were injected into the sub-serosal layer of the uterus in an IUA rat model, for 3 d, 3 mo, and 6 mo separately, to monitor the corresponding acute, sub-chronic, and chronic effects. Healthy rats of the same age served as negative controls. Toxicity effects were evaluated by body weight, organ weight, histopathology, hematology, and biochemistry tests. Tumorigenicity of MenSCs was investigated in Balb/c-nu mice in vivo and by colony formation assays in vitro.
RESULTS Compared with the same week-old control group, all of the IUA rats receiving MenSC transplantation demonstrated no obvious changes in body weight, main organ weight, or blood cell composition during the acute, sub-chronic, and chronic observation periods. At the same time, serum biochemical tests showed no adverse effects on metabolism or liver and kidney function. After 4 wk of subcutaneous injection of MenSCs in Balb/c-nu nude mice, no tumor formation or cell metastasis was observed. Moreover, there was no tumor colony formation of MenSCs during soft agar culture in vitro.
CONCLUSION There is no acute, sub-chronic, or chronic poisoning, infection, tumorigenesis, or endometriosis in rats with IUA after MenSC transplantation. The above results suggest that intrauterine transplantation of MenSCs is safe for endometrial treatment.
Collapse
Affiliation(s)
- Qi-Yuan Chang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110022, Liaoning Province, China
- Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang 110004, Liaoning Province, China
| | - Si-Wen Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110022, Liaoning Province, China
- Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang 110004, Liaoning Province, China
| | - Ping-Ping Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110022, Liaoning Province, China
- Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang 110004, Liaoning Province, China
| | - Zheng-Wei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Benxi 117004, Liaoning Province, China
| | - Ji-Chun Tan
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110022, Liaoning Province, China
- Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang 110004, Liaoning Province, China
| |
Collapse
|
33
|
Chen L, Qu J, Cheng T, Chen X, Xiang C. Menstrual blood-derived stem cells: toward therapeutic mechanisms, novel strategies, and future perspectives in the treatment of diseases. Stem Cell Res Ther 2019; 10:406. [PMID: 31864423 PMCID: PMC6925480 DOI: 10.1186/s13287-019-1503-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/07/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022] Open
Abstract
Menstrual blood-derived stem cells (MenSCs) have great potential in the treatment of various diseases. As a novel type of mesenchymal stem cells (MSCs), MenSCs have attracted more interest due to their therapeutic effects in both animal models and clinical trials. Here, we described the differentiation, immunomodulation, paracrine, homing, and engraftment mechanisms of MenSCs. These include differentiation into targeting cells, immunomodulation with various immune cells, the paracrine effect on secreting cytokines, and homing and engraftment into injured sites. To better conduct MenSC-based therapy, some novel hotspots were proposed such as CRISPR (clustered regularly interspaced short palindromic repeats)/cas9-mediated gene modification, exosomes for cell-free therapy, single-cell RNA sequence for precision medicine, engineered MenSC-based therapy for the delivery platform, and stem cell niches for improving microenvironment. Subsequently, current challenges were elaborated on, with regard to age of donor, dose of MenSCs, transplantation route, and monitoring time. The management of clinical research with respect to MenSC-based therapy in diseases will become more normative and strict. Thus, a more comprehensive horizon should be considered that includes a combination of traditional solutions and novel strategies. In summary, MenSC-based treatment has a great potential in treating diseases through diverse strategies, and more therapeutic mechanisms and novel strategies need to be elucidated for future regenerative medicine and clinical applications.
Collapse
Affiliation(s)
- Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China.,Stowers Institute for Medical Research, 1000 E 50th Street, Kansas City, MO, 64110, USA
| | - Jingjing Qu
- Lung Cancer and Gastroenterology Department, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical, School of Central South University, Changsha, 410008, People's Republic of China.,Department of Respiratory Disease, Thoracic Disease Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Tianli Cheng
- Thoracic Medicine Department 1, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical, School of Central South University, Changsha, 410008, People's Republic of China
| | - Xin Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China.
| |
Collapse
|