1
|
Rezayat F, Esmaeil N, Nikpour P, Feizi A, Rezaei A. Different behavior of NK cells isolated from healthy women and women with recurrent spontaneous abortion after treatment with human amniotic epithelial cells. J Leukoc Biol 2025; 117:qiaf020. [PMID: 40276927 DOI: 10.1093/jleuko/qiaf020] [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: 06/26/2024] [Revised: 12/03/2024] [Indexed: 04/26/2025] Open
Abstract
Maternal immunotolerance during pregnancy is heavily dependent on the critical properties of human amniotic epithelial cells (hAECs). Recurrent spontaneous abortion (RSA) is one of the most common diseases in women and is caused by feto-maternal immunotolerance disruption. The objective of this study is to investigate how hAEECs affect pNK cells isolated from RSA and healthy women in terms of immunomodulation. Peripheral blood NK cells were isolated from 20 women with RSA and 20 healthy women. Purified NK cells were co-cultured with hAECs, obtained from full-term healthy pregnant women at different cellular ratios. After 24 and 72 h of incubation, the expression of immunomodulatory genes in hAECs, immunophenotype, and cytotoxicity of NK cells, and cytokine production were investigated using real-time PCR, flow cytometry, and ELISA techniques, respectively. We observed a significant increase in TGF-β and IL-10 production, and CD56bright CD16+ subpopulation in pNK cells, a significant decrease in IFN-γ production and CD107a and FasL expression on NK cells. Also, NK cells' cytotoxicity against K562 cells was diminished after co-culture with hAECs. The expression of TGF-β and HLA-G genes by hAECs was diminished after co-culture with NK cells isolated from women with RSA. Our research indicates that the interaction between NK cells and hAECs influences the phenotype and function of both cells. Also, NK cells belonging to women with RSA and healthy women exhibit different behavior during treatment with hAECs, possibly due to NK cell dysfunction. However, extensive research is required to assess NK cell defects and their mutual interaction with hAECs.
Collapse
Affiliation(s)
- Fatemeh Rezayat
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan 8174673461, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan 8174673461, Iran
- Pooya Zist-Mabna Hakim Company, Isfahan Health Center, Aghababaei Highway, Isfahan 8159611119, Iran
| | - Parvaneh Nikpour
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan 8174673461, Iran
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Post box 100 405 30, Gothenburg, Sweden
| | - Awat Feizi
- Department of Epidemiology and Biostatistics, Isfahan Endocrine and Metabolism Research Center, School of Public Health, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan 8174673461, Iran
| | - Abbas Rezaei
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan 8174673461, Iran
- Immunodeficiency Diseases Research Center, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan 8174673461, Iran
| |
Collapse
|
2
|
Hao W, Luo Y, Tian J, Lu Y, Cui Y, Zhang Y, Jin X, Ye H, Lu M, Song J, Zhou W, Zhang W, He Z. Scale-Up of Human Amniotic Epithelial Cells Through Regulation of Epithelial-Mesenchymal Plasticity Under Defined Conditions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408581. [PMID: 39804851 PMCID: PMC11923953 DOI: 10.1002/advs.202408581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/12/2024] [Indexed: 01/16/2025]
Abstract
Human amniotic epithelial cells (hAECs) have shown excellent efficacy in clinical research and have prospective applications in the treatment of many diseases. However, the properties of the hAECs and their proliferative mechanisms remain unclear. Here, single-cell RNA sequencing (scRNA-seq) is performed on hAECs obtained from amniotic tissues at different gestational ages and passages during in vitro culture. The results showed that the proliferation of hAECs is associated with epithelial-mesenchymal plasticity (EMP) during amniogenesis. Freshly isolated, full-term hAECs are identified as mature epithelial cells. Once cultured in vitro, they are observed to rapidly undergo epithelial-mesenchymal transition (EMT) and enter a partial epithelial-mesenchymal transition (pEMT) state to regain their EMP properties and proliferation capacities. With the continuous development of EMT, hAECs eventually enter a senescent state. The addition of SB431542 and microcarrier screening enabled the effective 3D expansion of hAECs by 50 fold while maintaining the EMP status in hAECs for further proliferation. This study not only elucidated the central proliferation mechanism of hAECs during development and expansion but also optimized the in vitro culture system so that it is sufficient to generate hAECs for 50 patients from a single donor amniotic membrane.
Collapse
Affiliation(s)
- Wangping Hao
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
| | - Yi Luo
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
- Shanghai Institute of Stem Cell Research and Clinical TranslationShanghai200120P. R. China
| | - Jia Tian
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
- Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
- College of Chemical EngineeringUniversity of the Chinese Academy of SciencesBeijing101408P. R. China
| | - Yuefeng Lu
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
| | - Yangyang Cui
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
| | - Ying Zhang
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
| | - Xiao Jin
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
| | - Hongjuan Ye
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
| | - Mengqi Lu
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
- Shanghai Institute of Stem Cell Research and Clinical TranslationShanghai200120P. R. China
- Postgraduate Training Base of Shanghai East HospitalJinzhou Medical UniversityJinzhouLiaoning121001P. R. China
| | - Jinjia Song
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
- Shanghai Institute of Stem Cell Research and Clinical TranslationShanghai200120P. R. China
| | - Weiqing Zhou
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
- Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
- College of Chemical EngineeringUniversity of the Chinese Academy of SciencesBeijing101408P. R. China
| | - Wencheng Zhang
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
- Shanghai Institute of Stem Cell Research and Clinical TranslationShanghai200120P. R. China
| | - Zhiying He
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
- Shanghai Institute of Stem Cell Research and Clinical TranslationShanghai200120P. R. China
- Postgraduate Training Base of Shanghai East HospitalJinzhou Medical UniversityJinzhouLiaoning121001P. R. China
| |
Collapse
|
3
|
Cerveró-Varona A, Prencipe G, Peserico A, Canciello A, House AH, Santos HA, Perugini M, Sulcanese L, Takano C, Miki T, Iannetta A, Russo V, Mattioli M, Barboni B. Amniotic epithelial Cell microvesicles uptake inhibits PBMCs and Jurkat cells activation by inducing mitochondria-dependent apoptosis. iScience 2025; 28:111830. [PMID: 39967871 PMCID: PMC11834128 DOI: 10.1016/j.isci.2025.111830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/15/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Amniotic epithelial cells (AECs) exhibit significant immunomodulatory and pro-regenerative properties, largely due to their intrinsic paracrine functions that are currently harnessed through the collection of their secretomes. While there is increasing evidence of the role of bioactive components freely secreted or carried by exosomes, the bioactive cargo of AEC microvesicles (MVs) and their crosstalk with the immune cells remains to be fully explored. We showed that under intrinsic conditions or in response to LPS, AEC-derived MV carries components such as lipid-mediated signaling molecules, ER, and mitochondria. They foster the intra/interspecific mitochondrial transfer into immune cells (PBMCs and Jurkat cells) in vitro and in vivo on the zebrafish larvae model of injury. The internalization of MV cargoes through macropinocytosis induces hyperpolarization of PBMC mitochondrial membranes and triggers MV-mediated apoptosis. This powerful immune suppressive mechanism triggered by AEC-MV cargo delivery paves the way for controlled and targeted cell-free therapeutic approaches.
Collapse
Affiliation(s)
- Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Alessia Peserico
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Angelo Canciello
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Andrew H. House
- Helsinki University Lipidomics Unit, Helsinki Institute for Life Science (HiLIFE), Biocenter 3, Viikinkaari 1, 00790 Helsinki, Finland
| | - Hélder A. Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, 9713 AV Groningen, the Netherlands
| | - Monia Perugini
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Ludovica Sulcanese
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Chika Takano
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Toshio Miki
- Department of Physiology, Nihon University School of Medicine, Tokyo, Japan
| | - Annamaria Iannetta
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Valentina Russo
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Mauro Mattioli
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
4
|
Zhu C, Liu Y, Xu H, Wang S, Zhou H, Cao J, Meng F, Zhang Y. Production of second-generation sheep clones via somatic cell nuclear transfer using amniotic cells as nuclear donors. Theriogenology 2025; 232:79-86. [PMID: 39515062 DOI: 10.1016/j.theriogenology.2024.11.001] [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: 04/17/2024] [Revised: 10/20/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Somatic Cell Nuclear Transfer (SCNT) has transformed animal genetic improvement, gene-editing in model production, xenotransplantation, and conservation efforts for endangered species. However, SCNT-derived embryos occasionally display developmental abnormalities, and following embryo transfer, the miscarriage rate is high. Gene-edited fetuses may experience birth defects, resulting in decreased survival rates. Correct selection of nuclear donor cells is essential for the success of somatic cell cloning. Fibroblasts are the most commonly used cells, but their rapid proliferation increases the risk of genetic mutation, impairing embryo development and production. Conversely, amniotic cells have slower proliferation rates, decreasing the mutation risk during cultivation. Amniotic cells are thus better SCNT candidates than fibroblasts because they offer genomic stability, low tumorigenic and teratogenic risks, reduced immunogenicity, high differentiation potential, ease of accessibility, and fewer ethical concerns. Cells derived from first-generation gene-edited animals exhibit stable genetic structures, reduced susceptibility to genetic alterations and artificial modifications, closely resembling natural cells, and enhanced compatibility with SCNT procedures. Amniotic cells derived from gene-edited sheep fetuses used as nuclear donor cells for SCNT successfully recloned three healthy second-generation gene-edited sheep. Using amniotic cells as nuclear donor cells for SCNT did not significantly alter embryo cleavage rates, blastocyst formation, or fetal birth compared to edited fibroblasts (p > 0.05). However, fetal survival rates were significantly higher than edited fibroblasts (p < 0.05). The results support the potential of amniotic cells as SCNT alternatives, suggesting a promising strategy to improve gene-edited fetus survival rates using first-generation gene-edited sheep-derived amniotic cells.
Collapse
Affiliation(s)
- Chunxiao Zhu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; Department of Pediatrics, Xinqiao Hospital, Army Medical University, Chongqing, 400073, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China
| | - Yiyi Liu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China
| | - Hongyang Xu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Shenyuan Wang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China
| | - Huanmin Zhou
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China
| | - Junwei Cao
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China.
| | - Fanhuan Meng
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China.
| | - Yanru Zhang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China; College of Medicine, Hainan Vocational University of Science and Technology, Haikou, 571126, China; Inner Mongolia Key Laboratory of Biomanufacture, Hohhot, 010018, China.
| |
Collapse
|
5
|
Zhou J, Luo S, Yao X, Li H, Wen Y. Analysis of changes in the chemokine CXC ligand 13 in serum and cerebrospinal fluid of patients with neuromyelitis optica. Sci Rep 2025; 15:2113. [PMID: 39814995 PMCID: PMC11735840 DOI: 10.1038/s41598-025-85650-7] [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: 07/25/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
To determine correlation between the Extended Disability Status Scale(EDSS) grade and the progression of neuromyelitis optica(NMO) patients' levels of the chemokine CXC ligand 13 (CXCL13) in their serum and cerebrospinal fluid. This research included forty-one patients diagnosed with neuromyelitis optica(NMO) and forty-three patients diagnosed with multiple sclerosis(MS). The control group consisted of forty-three non-inflammatory neurological disease(NND) patients. The patients' serum and cerebrospinal fluid CXCL13 levels were measured. Patients in NMO group and MS group had serum and cerebrospinal fluid with CXCL13 levels that were substantially greater than those in the NND group. When comparing the CXCL13 levels of blood and cerebrospinal fluid between patients in the EDSS ≥ 3.5 group and the EDSS<3.5 group, with the EDSS ≥ 3.5 group's CXCL13 levels being greater(P<0.05). There was a positive correlation between the serum CXCL13 and the EDSS grades of both the NMO and MS groups(r = 0.884, P < 0.001); The cerebrospinal fluid CXCL13 of the NMO and MS groups showed a positive correlation with their EDSS grades(r = 0.681, P < 0.001). EDSS scores of NMO patients were positively correlated with their serum BLC-1 (r = 0.896, P < 0.001); EDSS scores of NMO patients were positively correlated with their cerebrospinal fluid BLC-1 (r = 0.678, P < 0.001).EDSS scores of MS patients were positively correlated with their serum BLC-1 (r = 0.852, P < 0.001); EDSS scores of MS patients were positively correlated with their cerebrospinal fluid BLC-1 (r = 0.613, P < 0.001). Serum and cerebrospinal fluid levels of CXCL13 may serve as an important biomarker for the presumptive assessment of the degree of disability in NMO and MS disease, providing a basis for the treatment and control of the disease.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Neurology, Chenzhou First People's Hospital, Chenzhou City, 423000, Hunan Province, China
| | - Shalin Luo
- Department of Neurology, Chenzhou First People's Hospital, Chenzhou City, 423000, Hunan Province, China
| | - Xiaoxi Yao
- Department of Neurology, Chenzhou First People's Hospital, Chenzhou City, 423000, Hunan Province, China
| | - Haipeng Li
- Department of Neurology, Chenzhou First People's Hospital, Chenzhou City, 423000, Hunan Province, China
| | - Ying Wen
- Department of Neurology, Chenzhou First People's Hospital, Chenzhou City, 423000, Hunan Province, China.
- Institute of Neuromedicine, Chenzhou First People's Hospital, Chenzhou First People's Hospital, No. 102 Luojiajing, Chenzhou City, 423000, Hunan Province, China.
| |
Collapse
|
6
|
Yang Z, Chen K, Zhang Y, Xu B, Huang Y, Zhang X, Liu Z, Wang T, Wu D, Peng T, Lu T, Cai H, Wang X. Study on pharmacokinetic and tissue distribution of hyperin, astragalin, kaempferol-3-O-β-D-glucuronide from rats with multiple administrations of Semen Cuscutae processed with salt solution with effect of treating recurrent spontaneous abortion. Front Pharmacol 2024; 15:1440810. [PMID: 39351087 PMCID: PMC11439818 DOI: 10.3389/fphar.2024.1440810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 06/28/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Semen Cuscutae is a traditional Chinese medicine (TCM) that tonifies the kidneys and prevents miscarriage. According to Chinese medicine theory, kidney deficiency is one of the main causes of recurrent spontaneous abortion (RSA). The previous studies showed that raw product of Semen Cuscutae (SP) and Semen Cuscutae processed with salt solution (YP) have ameliorative effects on RSA, and that YP is superior to SP. However, the active components of YP to ameliorate RSA remain unclear and require further studies. The objective of this study is to investigate the active components of YP in ameliorating RSA. Methods First, a rat model of RSA was established using hydroxyurea in combination with mifepristone. Aqueous decoction of YP was given by gavage to rats. Second, pregnant rats were sampled on days 5, 7, 9, 10 and 12 during the modelling period. The content of Hyperin (HY), astragalin (AS) and kaempferol-3-O-β-D-glucuronide (KA) in blood and liver, heart, spleen, lung and kidney tissues were detected by liquid chromatography-mass spectrometry (LC-MS). The pharmacodynamic indicators including progesterone (P), chorionic gonadotropin β (β-HCG), estradiol (E2), tumor necrosis factor-α (TFN-α), interleukin 4 (IL-4), and tryptophan (TRP) were measured by enzyme-linked immunosorbent assay (ELISA) Pearson's correlation analysis and grey relational analysis were used to establish the relationship between the pharmacodynamic indexes and chemical constituents. Results The pharmacokinetic results showed that the area under curve (AUC) value of KA was the largest. The tissue distribution results showed that astragalin was widely distributed in liver, heart, spleen, lung and kidney in the RSA model rats, while HY was detected only in the uterus, and KA was detected only in the kidney. The pearson correlationl analysis showed that KA was significantly and positively correlated with the contents of E2, P, β-HCG and TRP. Both AS and HY were significantly negatively correlated with the content of TNF-α, respectively. Discussion This study reveals the pharmacokinetics and tissue distribution of KA, AS and HY in rats with RSA. It was elucidated that all three were involved in the regulation of progesterone levels and immune function. It initially revealed the mechanism of action of YP in enhancing the improvement of RSA, and it provided a theoretical basis for the quality assessment of YP.
Collapse
Affiliation(s)
- Zhitong Yang
- Anhui Province Key Laboratory of Research, Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Kaiwen Chen
- Anhui Province Key Laboratory of Research, Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yuting Zhang
- Anhui Province Key Laboratory of Research, Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Baiyang Xu
- Anhui Province Key Laboratory of Research, Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yu Huang
- Anhui Province Key Laboratory of Research, Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Xue Zhang
- Anhui Province Key Laboratory of Research, Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Zilu Liu
- Anhui Province Key Laboratory of Research, Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Tongsheng Wang
- Anhui Province Key Laboratory of Research, Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Deling Wu
- Anhui Province Key Laboratory of Research, Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
- Heritage Base of TCM Processing Technology of NATCM, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Hefei, China
| | - Tangyi Peng
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Tulin Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hao Cai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoli Wang
- Anhui Province Key Laboratory of Research, Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
- Heritage Base of TCM Processing Technology of NATCM, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Hefei, China
| |
Collapse
|
7
|
Cao W, Zhang Q, Huang Y, Zhang Q, Lai D. Pretreatment with Inflammatory Factors Altered the Secretome of Human Amniotic Epithelial Cells. Tissue Eng Part C Methods 2024; 30:255-267. [PMID: 38756098 DOI: 10.1089/ten.tec.2024.0065] [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] [Indexed: 05/18/2024] Open
Abstract
Human amniotic epithelial cells (hAECs) are novel and promising therapeutic agents for patients suffering from degenerative diseases. Studies have demonstrated that the therapeutic effects of hAECs mainly depend on their paracrine components. Currently, appropriate pretreatment is a widely confirmed strategy for enhancing the repair potential of stem cells; however, the effect of proinflammatory factor pretreatment on hAECs and their secretome is still unclear. In this study, we used the well-characterized proinflammatory factors tumor necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ) to stimulate hAECs and analyzed the effect of TNF-α and IFN-γ on hAECs, including gene expression profile, paracrine proteins, and microRNAs (miRNAs) in exosomes. Results showed that TNF-α and IFN-γ pretreatment improved the viability of hAECs but inhibited the proliferation of hAECs. TNF-α and IFN-γ pretreatment altered the gene expression profile of hAECs, and upregulated differentially expressed genes were predominantly enriched in biological adhesion, antioxidant activity, and response to IFN-beta. In addition, TNF-α and IFN-γ pretreatment enhanced the paracrine secretion of cytokines by hAECs. The upregulated differentially expressed proteins were mainly enriched in tissue remodeling proteins and cytokine-cytokine receptor. Notably, the expression of miRNAs in exosomes from hAECs was also changed by TNF-α and IFN-γ pretreatment. The target genes of upregulated exosomal miRNAs substantially contributed to the response to stimulus, metabolic pathways, and PI3K-Akt signaling pathway. Our findings improve our understanding of the biological characteristics of hAECs after proinflammatory factor pretreatment and provide novel insights to strengthen and optimize the therapeutic potential of hAECs and their secretome in regenerative medicine.
Collapse
Affiliation(s)
- Wenjiao Cao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qinyu Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yating Huang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiuwan Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| |
Collapse
|
8
|
Morandi F, Airoldi I, Faini A, Horenstein A, Malavasi F, Matysiak N, Kopaczka K, Marimpietri D, Gramignoli R. Immune-regulatory properties carried by human amnion epithelial cells: Focus on the role of HLA-G and adenosinergic ectoenzymes. Hum Immunol 2023:S0198-8859(23)00068-X. [PMID: 37169599 DOI: 10.1016/j.humimm.2023.04.006] [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: 11/28/2022] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/13/2023]
Abstract
Human amnion epithelial cells (hAEC) can be efficiently isolated from full-term amnion membrane and have been gaining recognition as advanced medical products. Such cells originate directly from the embryo during the early phase of development and exert a crucial function in the establishment of a tolerogenic environment, to avoid maternal immune rejection. Amnion cell immuno-modulation may be exploited, but additional efforts are required to establish the mechanisms underlying such capacity. The way to fully clarify such an issue is so far long. Here we overview current knowledge on the effects on innate or adaptive immune cells offered by intact hAEC or secreted mediators, pinpointing the mechanisms to date elucidated by our group and others. We move from the description of hAEC general features to molecular intermediaries generating effects directly or indirectly on immune cells. We focus on the role of non-canonical HLA class I molecules, with emphasis on HLA-G, but expand such analysis on adenosinergic mediators, cytokines, and hAEC-derived microvesicles. Finally, we report the ongoing clinical trials exploiting hAEC multipotency and immune modulation.
Collapse
Affiliation(s)
- F Morandi
- UOSD Cell Factory, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy.
| | - I Airoldi
- UOSD Cell Factory, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - A Faini
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - A Horenstein
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - F Malavasi
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, 10126 Torino, Italy; Fondazione Ricerca Molinette, Torino, Italy
| | - N Matysiak
- Department of Histology and Cell Pathology in Zabrze, Medical University of Silesia in Katowice, Poland
| | - K Kopaczka
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - D Marimpietri
- UOSD Cell Factory, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - R Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
9
|
Estúa-Acosta GA, Buentello-Volante B, Magaña-Guerrero FS, Flores JEA, Vivanco-Rojas O, Castro-Salas I, Zarco-Ávila K, García-Mejía MA, Garfias Y. Human Amniotic Membrane Mesenchymal Stem Cell-Synthesized PGE 2 Exerts an Immunomodulatory Effect on Neutrophil Extracellular Trap in a PAD-4-Dependent Pathway through EP2 and EP4. Cells 2022; 11:cells11182831. [PMID: 36139406 PMCID: PMC9496826 DOI: 10.3390/cells11182831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Human amniotic membrane mesenchymal stem cells (hAM-MSC) secrete a myriad of components with immunosuppressive activities. In the present research, we aimed to describe the effect of prostaglandin E2 (PGE2) secreted by hAM-MSCs on neutrophil extracellular trap (NET) release and to characterize the role of its receptors (EP2/EP4) in PAD-4 and NFκB activity in neutrophils. Human peripheral blood neutrophils were ionomycin-stimulated in the presence of hAM-MSC conditioned medium (CM) treated or not with the selective PGE2 inhibitor MF-63, PGE2, EP2/EP4 agonists, and the selective PAD-4 inhibitor GSK-484. NET release, PAD-4, and NFκB activation were analyzed. Ionomycin induced NET release, which was inhibited in the presence of hAM-MSC-CM, while CM from hAM-MSCs treated with MF-63 prevented NET release inhibition. PGE2 and EP2/EP4 agonists, and GSK-484 inhibited NET release. EP2/EP4 agonists and GSK-484 inhibited H3-citrullination but did not affect PAD-4 protein expression. Finally, PGE2 and EP2/EP4 agonists and GSK-484 increased NFκB phosphorylation. Taken together, these results suggest that hAM-MSC exert their immunomodulatory activities through PGE2, inhibiting NET release in a PAD-4-dependent pathway. This research proposes a new mechanism by which hAM-MSC exert their activities when modulating the innate immune response and inhibiting NET release.
Collapse
Affiliation(s)
| | - Beatriz Buentello-Volante
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | - Fátima Sofía Magaña-Guerrero
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | - José Eduardo-Aguayo Flores
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | - Oscar Vivanco-Rojas
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | - Ilse Castro-Salas
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | - Karla Zarco-Ávila
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | - Mariana A. García-Mejía
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
| | - Yonathan Garfias
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology Conde de Valenciana, Mexico City 06800, Mexico
- Department of Biochemistry, Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Correspondence: or
| |
Collapse
|
10
|
In Vitro Differentiation of Human Amniotic Epithelial Cells into Hepatocyte-like Cells. Cells 2022; 11:cells11142138. [PMID: 35883581 PMCID: PMC9317663 DOI: 10.3390/cells11142138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 02/05/2023] Open
Abstract
Human amniotic epithelial cells (hAECs) represent an interesting clinical alternative to human embryonic (hESCs) and induced pluripotent (hiPSCs) stem cells in regenerative medicine. The potential of hAECs can be enhanced ex vivo by their partial pre-differentiation. The aim of this study was to evaluate the effectiveness of 18-day differentiation of hAECs into endodermal cells, hepatic precursor cells, and cells showing functional features of hepatocytes using culture media supplemented with high (100 ng/mL) concentrations of EGF or HGF. The cells obtained after differentiation showed changes in morphology and increased expression of AFP, ALB, CYP3A4, CYP3A7, and GSTP1 genes. HGF was more effective than EGF in increasing the expression of liver-specific genes in hAECs. However, EGF stimulated the differentiation process more efficiently and yielded more hepatocyte-like cells capable of synthesizing α-fetoprotein during differentiation. Additionally, after 18 days, GST transferases, albumin, and CYP P450s, which proved their partial functionality, were expressed. In summary, HGF and EGF at a dose of 100 ng/mL can be successfully used to obtain hepatocyte-like cells between days 7 and 18 of hAEC differentiation. However, the effectiveness of this process is lower compared with hiPSC differentiation; therefore, optimization of the composition of the medium requires further research.
Collapse
|
11
|
Ferdousi F, Isoda H. Regulating Early Biological Events in Human Amniotic Epithelial Stem Cells Using Natural Bioactive Compounds: Extendable Multidirectional Research Avenues. Front Cell Dev Biol 2022; 10:865810. [PMID: 35433672 PMCID: PMC9011193 DOI: 10.3389/fcell.2022.865810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/07/2022] [Indexed: 12/21/2022] Open
Abstract
Stem cells isolated from perinatal tissue sources possess tremendous potential for biomedical and clinical applications. On the other hand, emerging data have demonstrated that bioactive natural compounds regulate numerous cellular and biochemical functions in stem cells and promote cell migration, proliferation, and attachment, resulting in maintaining stem cell proliferation or inducing controlled differentiation. In our previous studies, we have reported for the first time that various natural compounds could induce targeted differentiation of hAESCs in a lineage-specific manner by modulating early biological and molecular events and enhance the therapeutic potential of hAESCs through modulating molecular signaling. In this perspective, we will discuss the advantages of using naturally occurring active compounds in hAESCs and their potential implications for biological research and clinical applications.
Collapse
Affiliation(s)
- Farhana Ferdousi
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan.,Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.,AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan
| | - Hiroko Isoda
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan.,Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.,AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan.,R&D Center for Tailor-made QOL, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
12
|
Picerno A, Castellano G, Curci C, Kopaczka K, Stasi A, Pertosa GB, Sabbà C, Gesualdo L, Gramignoli R, Sallustio F. The Icarus Flight of Perinatal Stem and Renal Progenitor Cells Within Immune System. Front Immunol 2022; 13:840146. [PMID: 35355984 PMCID: PMC8959820 DOI: 10.3389/fimmu.2022.840146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/11/2022] [Indexed: 12/02/2022] Open
Abstract
Our immune system actively fights bacteria and viruses, and it must strike a delicate balance between over- and under-reaction, just like Daedalus and Icarus in Greek mythology, who could not escape their imprisonment by flying too high or too low. Both human amniotic epithelial and mesenchymal stromal cells and the conditioned medium generated from their culture exert multiple immunosuppressive activities. They have strong immunomodulatory properties that are influenced by the types and intensity of inflammatory stimuli present in the microenvironment. Notably, very recently, the immunomodulatory activity of human adult renal stem/progenitor cells (ARPCs) has been discovered. ARPCs cause a decrease in Tregs and CD3+ CD4- CD8- (DN) T cells in the early stages of inflammation, encouraging inflammation, and an increase in the late stages of inflammation, favoring inflammation quenching. If the inflammatory trigger continues, however, ARPCs cause a further increase in DN T cells to avoid the development of a harmful inflammatory state. As in the flight of Daedalus and Icarus, who could not fly too high or too low to not destroy their wings by the heat of the sun or the humidity of the sea, in response to an inflammatory environment, stem cells seem to behave by paying attention to regulating T cells in the balance between immune tolerance and autoimmunity. Recognizing the existence of both suppressive and stimulatory properties, and the mechanisms that underpin the duality of immune reaction, will aid in the development of active immunotherapeutic approaches that manipulate the immune system to achieve therapeutic benefit.
Collapse
Affiliation(s)
- Angela Picerno
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Bari, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Renal Transplant Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Claudia Curci
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Katarzyna Kopaczka
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Alessandra Stasi
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Battista Pertosa
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Carlo Sabbà
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Bari, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Fabio Sallustio
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Bari, Italy
| |
Collapse
|
13
|
Kolanko E, Grajoszek A, Czekaj P. Immunosuppressive Potential of Activated Human Amniotic Cells in an Experimental Murine Model of Skin Allo- and Xenotransplantation. Front Med (Lausanne) 2021; 8:715590. [PMID: 34631739 PMCID: PMC8494785 DOI: 10.3389/fmed.2021.715590] [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: 05/27/2021] [Accepted: 08/26/2021] [Indexed: 11/25/2022] Open
Abstract
Isolated human amniotic cells (hAC) could be used as a source of immunomodulatory factors in regenerative medicine and transplantation. However, in previous experimental studies, native hAC administered to skin graft recipients did not induce graft immunotolerance. To strengthen the immunomodulatory properties of hAC prior to administration to the recipient, we activated them ex vivo using pro-inflammatory cytokines. In this study, we compared the transplantation efficiency of skin allografts (mouse to mouse) and xnografts (rat to mouse) in recipient mice divided into three main groups receiving: 1. Placebo (control group); 2. Cyclosporine A (CsA) [10 or 50 mg/kg body weight (bw)]; 3. suspension of hAC activated ex vivo by IL-1β and INFγ, administered into a tail vein or subcutaneously. During 15 days of observation, hAC administered intravenously or subcutaneously after allotransplantation appeared to be as safe and efficient as CsA at the dose of 10 mg/kg bw in preventing rejection of skin allo- and xenografts. After xenotransplantation, however, only hAC administered intravenously prevented rejection to an extent comparable to CsA. Both CsA (10 mg/kg bw) and activated hAC reduced inflammatory infiltration in the skin (after intravenous injection) and did not increase the concentration of the inflammation marker SAP in serum or percentage of leukocytes in blood. Finally, we concluded that administration of activated hAC is safe and efficient in the presented animal model of skin allo- and xenotransplantation in a route-dependent manner. Activated hAC injected intravenously exhibit an immunosuppressive effect comparable to CsA administered at the dose of 10 mg/kg bw in both allo- and xenotransplantation.
Collapse
Affiliation(s)
- Emanuel Kolanko
- Department of Cytophysiology, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Aniela Grajoszek
- Department of Experimental Medicine, Medical University of Silesia in Katowice, Katowice, Poland
| | - Piotr Czekaj
- Department of Cytophysiology, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
14
|
Mechanisms of Immunomodulation and Cytoprotection Conferred to Pancreatic Islet by Human Amniotic Epithelial Cells. Stem Cell Rev Rep 2021; 18:346-359. [PMID: 34613550 PMCID: PMC8799589 DOI: 10.1007/s12015-021-10269-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2021] [Indexed: 12/19/2022]
Abstract
Inhibiting pro-inflammatory cytokine activity can reverse inflammation mediated dysfunction of islet grafts. Human amniotic epithelial cells (hAECs) possess regenerative, immunomodulatory and anti-inflammatory properties. We hypothesized that hAECs could protect islets from cellular damage induced by pro-inflammatory cytokines. To verify our hypothesis, hAEC monocultures, rat islets (RI), or RI-hAEC co-cultures where exposed to a pro-inflammatory cytokine cocktail (Interferon γ: IFN-γ, Tumor necrosis factor α: TNF-α and Interleukin-1β: IL-1β). The secretion of anti-inflammatory cytokines and gene expression changes in hAECs and viability and function of RI were evaluated. The expression of non-classical Major Histocompatibility Complex (MHC) class I molecules by hAECs cultured with various IFN-γ concentrations were assessed. Exposure to the pro-inflammatory cocktail significantly increased the secretion of the anti-inflammatory cytokines IL6, IL10 and G-CSF by hAECs, which was confirmed by upregulation of IL6, and IL10 gene expression. HLA-G, HLA-E and PDL-1 gene expression was also increased. This correlated with an upregulation of STAT1, STAT3 and NF-κB1gene expression levels. RI co-cultured with hAECs maintained normal function after cytokine exposure compared to RI cultured alone, and showed significantly lower apoptosis rate. Our results show that exposure to pro-inflammatory cytokines stimulates secretion of anti-inflammatory and immunomodulatory factors by hAECs through the JAK1/2 – STAT1/3 and the NF-κB1 pathways, which in turn protects islets against inflammation-induced damages. Integrating hAECs in islet transplants appears as a valuable strategy to achieve to inhibit inflammation mediated islet damage, prolong islet survival, improve their engraftment and achieve local immune protection allowing reducing systemic immunosuppressive regimens.
Collapse
|
15
|
Zhang Q, Lai D. Application of human amniotic epithelial cells in regenerative medicine: a systematic review. Stem Cell Res Ther 2020; 11:439. [PMID: 33059766 PMCID: PMC7559178 DOI: 10.1186/s13287-020-01951-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/24/2020] [Indexed: 12/16/2022] Open
Abstract
Human amniotic epithelial cells (hAECs) derived from placental tissues have gained considerable attention in the field of regenerative medicine. hAECs possess embryonic stem cell-like proliferation and differentiation capabilities, and adult stem cell-like immunomodulatory properties. Compared with other types of stem cell, hAECs have special advantages, including easy isolation, plentiful numbers, the obviation of ethical debates, and non-immunogenic and non-tumorigenic properties. During the past two decades, the therapeutic potential of hAECs for treatment of various diseases has been extensively investigated. Accumulating evidence has demonstrated that hAEC transplantation helps to repair and rebuild the function of damaged tissues and organs by different molecular mechanisms. This systematic review focused on summarizing the biological characteristics of hAECs, therapeutic applications, and recent advances in treating various tissue injuries and disorders. Relevant studies published in English from 2000 to 2020 describing the role of hAECs in diseases and phenotypes were comprehensively sought out using PubMed, MEDLINE, and Google Scholar. According to the research content, we described the major hAEC characteristics, including induced differentiation plasticity, homing and differentiation, paracrine function, and immunomodulatory properties. We also summarized the current status of clinical research and discussed the prospects of hAEC-based transplantation therapies. In this review, we provide a comprehensive understanding of the therapeutic potential of hAECs, including their use for cell replacement therapy as well as secreted cytokine and exosome biotherapy. Moreover, we showed that the powerful immune-regulatory function of hAECs reveals even more possibilities for their application in the treatment of immune-related diseases. In the future, establishing the optimal culture procedure, achieving precise and accurate treatment, and enhancing the therapeutic potential by utilizing appropriate preconditioning and/or biomaterials would be new challenges for further investigation.
Collapse
Affiliation(s)
- Qiuwan Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Key Laboratory of Embryo Original Diseases; Shanghai Municipal Key Clinical Speciality, 145, Guang-Yuan Road, Shanghai, 200030, People's Republic of China
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Key Laboratory of Embryo Original Diseases; Shanghai Municipal Key Clinical Speciality, 145, Guang-Yuan Road, Shanghai, 200030, People's Republic of China.
| |
Collapse
|
16
|
Michalik M, Gładyś A, Czekaj P. Differentiation of Cells Isolated from Afterbirth Tissues into Hepatocyte-Like Cells and Their Potential Clinical Application in Liver Regeneration. Stem Cell Rev Rep 2020; 17:581-603. [PMID: 32974851 PMCID: PMC8036182 DOI: 10.1007/s12015-020-10045-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2020] [Indexed: 12/11/2022]
Abstract
Toxic, viral and surgical injuries can pose medical indications for liver transplantation. The number of patients waiting for a liver transplant still increases, but the number of organ donors is insufficient. Hepatocyte transplantation was suggested as a promising alternative to liver transplantation, however, this method has some significant limitations. Currently, afterbirth tissues seem to be an interesting source of cells for the regenerative medicine, because of their unique biological and immunological properties. It has been proven in experimental animal models, that the native stem cells, and to a greater extent, hepatocyte-like cells derived from them and transplanted, can accelerate regenerative processes and restore organ functioning. The effective protocol for obtaining functional mature hepatocytes in vitro is still not defined, but some studies resulted in obtaining functionally active hepatocyte-like cells. In this review, we focused on human stem cells isolated from placenta and umbilical cord, as potent precursors of hepatocyte-like cells for regenerative medicine. We summarized the results of preclinical and clinical studies dealing with the introduction of epithelial and mesenchymal stem cells of the afterbirth origin to the liver failure therapy. It was concluded that the use of native afterbirth epithelial and mesenchymal cells in the treatment of liver failure could support liver function and regeneration. This effect would be enhanced by the use of hepatocyte-like cells obtained from placental and/or umbilical stem cells. Graphical abstract ![]()
Collapse
Affiliation(s)
- Marcin Michalik
- Department of Cytophysiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Aleksandra Gładyś
- Department of Cytophysiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Piotr Czekaj
- Department of Cytophysiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland.
| |
Collapse
|
17
|
Morandi F, Marimpietri D, Görgens A, Gallo A, Srinivasan RC, El-Andaloussi S, Gramignoli R. Human Amnion Epithelial Cells Impair T Cell Proliferation: The Role of HLA-G and HLA-E Molecules. Cells 2020; 9:E2123. [PMID: 32961693 PMCID: PMC7563681 DOI: 10.3390/cells9092123] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/01/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
The immunoprivilege status characteristic of human amnion epithelial cells (hAECs) has been recently highlighted in the context of xenogenic transplantation. However, the mechanism(s) involved in such regulatory functions have been so far only partially been clarified. Here, we have analyzed the expression of HLA-Ib molecules in isolated hAEC obtained from full term placentae. Moreover, we asked whether these molecules are involved in the immunoregulatory functions of hAEC. Human amnion-derived cells expressed surface HLA-G and HLA-F at high levels, whereas the commonly expressed HLA-E molecule has been measured at a very low level or null on freshly isolated cells. HLA-Ib molecules can be expressed as membrane-bound and soluble forms, and in all hAEC batches analyzed we measured high levels of sHLA-G and sHLA-E when hAEC were maintained in culture, and such a release was time-dependent. Moreover, HLA-G was present in extracellular vesicles (EVs) released by hAEC. hAEC suppressed T cell proliferation in vitro at different hAEC:T cell ratios, as previously reported. Moreover, inhibition of T cell proliferation was partially reverted by pretreating hAEC with anti-HLA-G, anti-HLA-E and anti-β2 microglobulin, thus suggesting that HLA-G and -E molecules are involved in hAEC-mediated suppression of T cell proliferation. Finally, either large-size EV (lsEV) or small-size EV (ssEV) derived from hAEC significantly modulated T-cell proliferation. In conclusion, we have here characterized one of the mechanism(s) underlying immunomodulatory functions of hAEC, related to the expression and release of HLA-Ib molecules.
Collapse
Affiliation(s)
- Fabio Morandi
- Stem Cell Laboratory and Cell Therapy Center, IRCCS Istituto Giannina Gaslini, Via Gaslini5, 16147 Genova, Italy;
| | - Danilo Marimpietri
- Stem Cell Laboratory and Cell Therapy Center, IRCCS Istituto Giannina Gaslini, Via Gaslini5, 16147 Genova, Italy;
| | - Andre Görgens
- Department of Laboratory Medicine, Division of Biomolecular and Cellular Medicine, Karolinska Institutet, 14157 Stockholm, Sweden; (A.G.); (S.E.-A.)
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Alessia Gallo
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Via E.Tricomi 5, 90127 Palermo, Italy;
| | - Raghuraman Chittor Srinivasan
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Alfred Nobels Alle 8, Huddinge SE-141 83, 14157 Stockholm, Sweden;
| | - Samir El-Andaloussi
- Department of Laboratory Medicine, Division of Biomolecular and Cellular Medicine, Karolinska Institutet, 14157 Stockholm, Sweden; (A.G.); (S.E.-A.)
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Alfred Nobels Alle 8, Huddinge SE-141 83, 14157 Stockholm, Sweden;
| |
Collapse
|
18
|
Srinivasan RC, Strom SC, Gramignoli R. Effects of Cryogenic Storage on Human Amnion Epithelial Cells. Cells 2020; 9:cells9071696. [PMID: 32679793 PMCID: PMC7407665 DOI: 10.3390/cells9071696] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/17/2020] [Accepted: 07/10/2020] [Indexed: 02/07/2023] Open
Abstract
Perinatal stem cells and epithelial cells isolated from full term amnion membrane, in particular, have attracted interest over the last decade, as a promising source of multipotent cells for cellular therapies. Human amnion epithelial cells (hAEC) have been used to treat monogenetic liver disease such as maple syrup urine disease or fibrosis of the liver in preclinical studies. In most studies xeno-transplants of hAEC were conducted without providing immunosuppression to recipients, reflecting the tolerogenic properties of hAEC. For many cell types, successful cryopreservation is critical for providing a readily available, off-the-shelf product. In this study, hAEC were isolated from full-term human placenta from 14 different donors, cryopreserved using a protocol and reagents commonly adopted for epithelial cell preservation. The cells were analyzed in terms of survival, recovery, and homogeneity, profiled for surface markers characteristic of epithelial, mesenchymal, endothelial, or hematopoietic cells. There were no significant differences observed in the percentage of cells with epithelial cell markers before and after cryopreservation. The relative proportion of stromal and hematopoietic cells was significantly reduced in hAEC preparations after cryopreservation. The expression of stem cell and immunomodulatory molecules were confirmed in the final product. Since multipotent cells are readily available from full-term placenta, this novel cell source might significantly increase the number of patients eligible to receive cellular therapies for liver and other diseases.
Collapse
|
19
|
Lebreton F, Bellofatto K, Wassmer CH, Perez L, Lavallard V, Parnaud G, Cottet-Dumoulin D, Kerr-Conte J, Pattou F, Bosco D, Othenin-Girard V, Martinez de Tejada B, Berishvili E. Shielding islets with human amniotic epithelial cells enhances islet engraftment and revascularization in a murine diabetes model. Am J Transplant 2020; 20:1551-1561. [PMID: 32031745 DOI: 10.1111/ajt.15812] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/12/2020] [Accepted: 01/28/2020] [Indexed: 01/25/2023]
Abstract
Hypoxia is a major cause of considerable islet loss during the early posttransplant period. Here, we investigate whether shielding islets with human amniotic epithelial cells (hAECs), which possess anti-inflammatory and regenerative properties, improves islet engraftment and survival. Shielded islets were generated on agarose microwells by mixing rat islets (RIs) or human islets (HI) and hAECs (100 hAECs/IEQ). Islet secretory function and viability were assessed after culture in hypoxia (1% O2 ) or normoxia (21% O2 ) in vitro. In vivo function was evaluated after transplant under the kidney capsule of diabetic immunodeficient mice. Graft morphology and vascularization were evaluated by immunohistochemistry. Both shielded RIs and HIs show higher viability and increased glucose-stimulated insulin secretion after exposure to hypoxia in vitro compared with control islets. Transplant of shielded islets results in considerably earlier normoglycemia and vascularization, an enhanced glucose tolerance, and a higher β cell mass. Our results show that hAECs have a clear cytoprotective effect against hypoxic damages in vitro. This strategy improves β cell mass engraftment and islet revascularization, leading to an improved capacity of islets to reverse hyperglycemia, and could be rapidly applicable in the clinical situation seeing that the modification to HIs are minor.
Collapse
Affiliation(s)
- Fanny Lebreton
- Cell Isolation and Transplantation Center, Department of Surgery, Faculty Diabetes Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Kevin Bellofatto
- Cell Isolation and Transplantation Center, Department of Surgery, Faculty Diabetes Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Charles H Wassmer
- Cell Isolation and Transplantation Center, Department of Surgery, Faculty Diabetes Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Lisa Perez
- Cell Isolation and Transplantation Center, Department of Surgery, Faculty Diabetes Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Vanessa Lavallard
- Cell Isolation and Transplantation Center, Department of Surgery, Faculty Diabetes Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Géraldine Parnaud
- Cell Isolation and Transplantation Center, Department of Surgery, Faculty Diabetes Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - David Cottet-Dumoulin
- Cell Isolation and Transplantation Center, Department of Surgery, Faculty Diabetes Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Julie Kerr-Conte
- INSERM U1190, Translational Research for Diabetes, University of Lille, France
| | - François Pattou
- INSERM U1190, Translational Research for Diabetes, University of Lille, France
| | - Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, Faculty Diabetes Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Véronique Othenin-Girard
- Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals, Geneva, Switzerland
| | - Begoña Martinez de Tejada
- Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, Department of Surgery, Faculty Diabetes Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Institute of Medical Research, Ilia State University, Tbilisi, Georgia
| |
Collapse
|
20
|
Morandi F, Airoldi I, Marimpietri D, Bracci C, Faini AC, Gramignoli R. CD38, a Receptor with Multifunctional Activities: From Modulatory Functions on Regulatory Cell Subsets and Extracellular Vesicles, to a Target for Therapeutic Strategies. Cells 2019; 8:E1527. [PMID: 31783629 PMCID: PMC6953043 DOI: 10.3390/cells8121527] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
CD38 is a multifunctional cell surface protein endowed with receptor/enzymatic functions. The protein is generally expressed at low/intermediate levels on hematological tissues and some solid tumors, scoring the highest levels on plasma cells (PC) and PC-derived neoplasia. CD38 was originally described as a receptor expressed by activated cells, mainly T lymphocytes, wherein it also regulates cell adhesion and cooperates in signal transduction mediated by major receptor complexes. Furthermore, CD38 metabolizes extracellular NAD+, generating ADPR and cyclic ADPR. This ecto-enzyme controls extra-cellular nucleotide homeostasis and intra-cellular calcium fluxes, stressing its relevance in multiple physiopathological conditions (infection, tumorigenesis and aging). In clinics, CD38 was adopted as a cell activation marker and in the diagnostic/staging of leukemias. Quantitative surface CD38 expression by multiple myeloma (MM) cells was the basic criterion used for therapeutic application of anti-CD38 monoclonal antibodies (mAbs). Anti-CD38 mAbs-mediated PC depletion in autoimmunity and organ transplants is currently under investigation. This review analyzes different aspects of CD38's role in regulatory cell populations and how these effects are obtained. Characterizing CD38 functional properties may widen the extension of therapeutic applications for anti-CD38 mAbs. The availability of therapeutic mAbs with different effects on CD38 enzymatic functions may be rapidly translated to immunotherapeutic strategies of cell immune defense.
Collapse
Affiliation(s)
- Fabio Morandi
- Laboratory of Stem Cell and Cell Therapy, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (I.A.); (D.M.)
| | - Irma Airoldi
- Laboratory of Stem Cell and Cell Therapy, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (I.A.); (D.M.)
| | - Danilo Marimpietri
- Laboratory of Stem Cell and Cell Therapy, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (I.A.); (D.M.)
| | - Cristiano Bracci
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, 10126 Torino, Italy; (C.B.); (A.C.F.)
- CeRMS, University of Torino, 10126 Torino, Italy
| | - Angelo Corso Faini
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, 10126 Torino, Italy; (C.B.); (A.C.F.)
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, SE-171 77 Stockholm, Sweden;
| |
Collapse
|