1
|
Hu W, Bei HP, Jiang H, Wu D, Yu X, Zhou X, Sun Q, Lu Q, Du Q, Wang L, Luo Z, Wu G, Zhao X, Wang S. DLM-GelMA/tumor slice sandwich structured tumor on a chip for drug efficacy testing. LAB ON A CHIP 2024; 24:3718-3727. [PMID: 38953554 DOI: 10.1039/d4lc00278d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The in vitro recapitulation of tumor microenvironment is of great interest to preclinical screening of drugs. Compared with culture of cell lines, tumor organ slices can better preserve the complex tumor architecture and phenotypic activity of native cells, but are limited by their exposure to fluid shear and gradual degradation under perfusion culture. Here, we established a decellularized liver matrix (DLM)-GelMA "sandwich" structure and a perfusion-based microfluidic platform to support long-term culture of tumor slices with excellent structural integrity and cell viability over 7 days. The DLM-GelMA was able to secrete cytokines and growth factors while providing shear protection to the tumor slice via the sandwich structure, leading to the preservation of the tumor microenvironment where immune cells (CD3, CD8, CD68), tumor-associated fibroblasts (α-SMA), and extracellular matrix components (collagen I, fibronectin) were well maintained. Furthermore, this chip presented anti-tumor efficacy at cisplatin (20 μM) on tumor patients, demonstrating our platform's efficacy to design patient-specific treatment regimens. Taken together, the successful development of this DLM-GelMA sandwich structure on the chip could faithfully reflect the tumor microenvironment and immune response, accelerating the screening process of drug molecules and providing insights for practical medicine.
Collapse
Affiliation(s)
- Wenqi Hu
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| | - Ho-Pan Bei
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China.
| | - Hongwei Jiang
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| | - Di Wu
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| | - Xiaorui Yu
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| | - Xintong Zhou
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China.
| | - Qiuwan Sun
- Sichuan Diya BioTechnology Group Company, Chengdu, 641400, China
| | - Qinrui Lu
- Sichuan Diya BioTechnology Group Company, Chengdu, 641400, China
| | - Qijun Du
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| | - Liangwen Wang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhi Luo
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Guohua Wu
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| | - Xin Zhao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China.
- Research Institute for Intelligent Wearable Systems, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, 999077, China
| | - Shuqi Wang
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| |
Collapse
|
2
|
Antarianto RD, Pragiwaksana A, Septiana WL, Mazfufah NF, Mahmood A. Hepatocyte Differentiation from iPSCs or MSCs in Decellularized Liver Scaffold: Cell–ECM Adhesion, Spatial Distribution, and Hepatocyte Maturation Profile. Organogenesis 2022; 18:2061263. [PMID: 35435152 PMCID: PMC9037523 DOI: 10.1080/15476278.2022.2061263] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSC) and induced pluripotent stem cells (iPSC) have been reported to be able to differentiate to hepatocyte in vitro with varying degree of hepatocyte maturation. A simple method to decellularize liver scaffold has been established by the Department of Histology, Faculty of Medicine, Universitas Indonesia, in SCTE IMERI lab.15 This study aims to evaluate hepatocyte differentiation from iPSCs compared to MSCs derived in our decellularized liver scaffold. The research stages started with iPSC culture, decellularization, seeding cell culture into the scaffold, and differentiation into hepatocytes for 21 days. Hepatocyte differentiation from iPSCs and MSCs in the scaffolds was characterized using hematoxylin–eosin, Masson Trichrome, and immunohistochemistry staining to determine the fraction of the differentiation area. RNA samples were isolated on days 7 and 21. Expression of albumin, CYP450, and CK-19 genes were analyzed using the qRT-PCR method. Electron microscopy images were obtained by SEM. Immunofluorescence examination was done using HNF4-α and CEBPA markers. The results of this study in hepatocyte-differentiated iPSCs compared with hepatocyte-differentiated MSCs in decellularized liver scaffold showed lower adhesion capacity, single-cell-formation and adhered less abundant, decreased trends of albumin, and lower CYP450 expression. Several factors contribute to this result: lower initial seeding number, which causes only a few iPSCs to attach to certain parts of decellularized liver scaffold, and manual syringe injection for recellularization, which abruptly and unevenly creates pattern of single-cell-formation by hepatocyte-differentiated iPSC in the scaffold. Hepatocyte-differentiated MSCs have the advantage of higher adhesion capacity to collagen fiber decellularized liver scaffold. This leads to positive result: increase trends of albumin and higher CYP450 expression. Hepatocyte maturation is shown by diminishing CK-19, which is more prominent in hepatocyte-differentiated iPSCs in decellularized liver scaffold. Confirmation of mature hepatocyte-differentiated iPSCs in decellularized liver scaffold maturation is positive for HNF4-a and CEBPA. The conclusion of this study is hepatocyte-differentiated iPSCs in decellularized liver scaffold is mature with lower cell–ECM adhesion, spatial cell distribution, albumin, and CYP450 expression than hepatocyte-differentiated MSCs in decellularized liver scaffold.
Collapse
Affiliation(s)
- Radiana Dhewayani Antarianto
- Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Stem cell and tissue engineering research cluster, (IMERI) Indonesian Medical Education and Research Institute, Jakarta Indonesia
- Program Doktor Ilmu Biomedik, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Adrian Pragiwaksana
- Program Master Ilmu Biomedik, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Wahyunia Likhayati Septiana
- Program Doktor Ilmu Biomedik, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Department of Histology, Faculty of Medicine, Universitas Gunadarma, Depok, Indonesia
| | - Nuzli Fahdia Mazfufah
- Stem cell and tissue engineering research cluster, (IMERI) Indonesian Medical Education and Research Institute, Jakarta Indonesia
| | - Ameer Mahmood
- Stem cell unit Department of Anatomy, King Saud University, Riyadh, Kingdom Saudi Arabia
| |
Collapse
|
3
|
Rahmani-Moghadam E, Zarrin V, Mahmoodzadeh A, Owrang M, Talaei-Khozani T. Comparison of the Characteristics of Breast Milk-derived Stem Cells with the Stem Cells Derived from the Other Sources: A Comparative Review. Curr Stem Cell Res Ther 2021; 17:71-90. [PMID: 34161214 DOI: 10.2174/1574888x16666210622125309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/14/2021] [Accepted: 03/28/2021] [Indexed: 11/22/2022]
Abstract
Breast milk (BrM) not only supplies nutrition, but it also contains a diverse population of cells. It has been estimated that up to 6% of the cells in human milk possess the characteristics of mesenchymal stem cells (MSC). Available data also indicate that these cells are multipotent and capable of self-renewal and differentiation with other cells. In this review, we have compared different characteristics, such as CD markers, differentiation capacity, and morphology of stem cells, derived from human breast milk (hBr-MSC) with human bone marrow (hBMSC), Wharton's jelly (WJMSC), and human adipose tissue (hADMSC). Through the literature review, it was revealed that human breast milk-derived stem cells specifically express a group of cell surface markers, including CD14, CD31, CD45, and CD86. Importantly, a group of markers, CD13, CD29, CD44, CD105, CD106, CD146, and CD166, were identified, which were common in the four sources of stem cells. WJMSC, hBMSC, hADMSC, and hBr-MSC are potently able to differentiate into the mesoderm, ectoderm, and endoderm cell lineages. The ability of hBr-MSCs todifferentiate into the neural stem cells, neurons, adipocyte, hepatocyte, chondrocyte, osteocyte, and cardiomyocytes has made these cells a promising source of stem cells in regenerative medicine, while isolation of stem cells from the commonly used sources, such as bone marrow, requires invasive procedures. Although autologous breast milk-derived stem cells are an accessible source for women who are in the lactation period, breast milk can be considered as a source of stem cells with high differentiation potential without any ethical concern.
Collapse
Affiliation(s)
- Ebrahim Rahmani-Moghadam
- Department of Anatomical sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahideh Zarrin
- Laboratory for Stem Cell Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Mahmoodzadeh
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Marzieh Owrang
- Department of Anatomical sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Talaei-Khozani
- Department of Anatomical sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
4
|
Nishino D, Kotake A, Yun CS, Rahman ANMI, El-Sharawy M, Yamanaka KI, Khandoker MAMY, Yamauchi N. Gene expression of bovine endometrial epithelial cells cultured in matrigel. Cell Tissue Res 2021; 385:265-275. [PMID: 33837849 DOI: 10.1007/s00441-021-03418-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 01/15/2021] [Indexed: 10/21/2022]
Abstract
Glandular epithelial cells (GE) in the endometrium are thought to support the elongation and survival of ruminant embryos by secreting histotrophs. In the present study, the gene expression of bovine endometrial epithelial cells cultured in matrigel was analyzed and examined whether it could be an in vitro model of GE. Bovine endometrial epithelial cells (BEE) and stromal cells (BES) were isolated from the slaughterhouse uteri and cultured in DMEM/F12 + 10% FBS. BEE showed the gland-like structure morphological changes when cultured in 15% matrigel but could not be identified in higher concentrations of the matrigel (30% or 60%). The expression of typical genes expressed in GE, SERPINA14 and GRP, was substantially high in matrigel-cultured BEE than in monolayer (P < 0.05). P4 and INFα have no significant effect on the SERPINA14 expression of BEE cultured in matrigel without co-culture with BES. On the other hand, when BEE were co-cultured with BES in matrigel culture, the expression of FGF13 was increased by the P4 treatment (P < 0.05). Furthermore, SERPINA14 and TXN expressions were increased by P4 + IFNα treatment (P < 0.05). These results demonstrate the appropriate conditions for BEE to form glandular structures in matrigel and the effect of co-culture with BES. The present study highlighted the possible use of matrigel for the culture of BEE to investigate the expression of cell-specific glandular epithelial genes as well as P4 and type-I IFN as factors controlling endometrial function during the implantation period.
Collapse
Affiliation(s)
- Daichi Nishino
- Department of Animal and Marine Bioresource Sciences, Faculty of Agriculture, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Ai Kotake
- Department of Animal and Marine Bioresource Sciences, Faculty of Agriculture, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Chi Sun Yun
- Department of Animal and Marine Bioresource Sciences, Faculty of Agriculture, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Al-Nur Md Iftekhar Rahman
- Department of Animal and Marine Bioresource Sciences, Faculty of Agriculture, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan.,Department of Animal Nutrition, Genetics and Breeding, Faculty of Animal Science and Veterinary Medicine, Sher-e-Bangla Agricultural University, Dhaka, 1207, Bangladesh
| | - Mohamed El-Sharawy
- Animal Production Department, Faculty of Agriculture, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | | | - M A M Yahia Khandoker
- Department of Animal Breeding and Genetics, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Nobuhiko Yamauchi
- Department of Animal and Marine Bioresource Sciences, Faculty of Agriculture, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan.
| |
Collapse
|
5
|
Chen F, Wang H, Xiao J. Regulated differentiation of stem cells into an artificial 3D liver as a transplantable source. Clin Mol Hepatol 2020; 26:163-179. [PMID: 32098013 PMCID: PMC7160355 DOI: 10.3350/cmh.2019.0022n] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/02/2020] [Indexed: 02/07/2023] Open
Abstract
End-stage liver disease is one of the leading causes of death around the world. Since insufficient sources of transplantable liver and possible immune rejection severely hinder the wide application of conventional liver transplantation therapy, artificial three-dimensional (3D) liver culture and assembly from stem cells have become a new hope for patients with end-stage liver diseases, such as cirrhosis and liver cancer. However, the induced differentiation of single-layer or 3D-structured hepatocytes from stem cells cannot physiologically support essential liver functions due to the lack of formation of blood vessels, immune regulation, storage of vitamins, and other vital hepatic activities. Thus, there is emerging evidence showing that 3D organogenesis of artificial vascularized liver tissue from combined hepatic cell types derived from differentiated stem cells is practical for the treatment of end-stage liver diseases. The optimization of novel biomaterials, such as decellularized matrices and natural macromolecules, also strongly supports the organogenesis of 3D tissue with the desired complex structure. This review summarizes new research updates on novel differentiation protocols of stem cell-derived major hepatic cell types and the application of new supportive biomaterials. Future biological and clinical challenges of this concept are also discussed.
Collapse
Affiliation(s)
- Feng Chen
- National Key Disciplines for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital, Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Jia Xiao
- Clinical Medicine Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
6
|
Conductive hydrogels based on agarose/alginate/chitosan for neural disorder therapy. Carbohydr Polym 2019; 224:115161. [DOI: 10.1016/j.carbpol.2019.115161] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 07/20/2019] [Accepted: 08/01/2019] [Indexed: 12/19/2022]
|
7
|
Zhou M, Shen L, Qiao Y, Sun Z. Inducing differentiation of human urine-derived stem cells into hepatocyte-like cells by coculturing with human hepatocyte L02 cells. J Cell Biochem 2019; 121:566-573. [PMID: 31407401 DOI: 10.1002/jcb.29301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 06/27/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVES To investigate the possibility of inducing differentiation of human urine-derived stem cells (hUSCs) into hepatocyte-like cells by coculturing with human hepatocyte L02 cells in vitro. METHODS HUSCs were isolated from fresh urine samples collected from healthy adult volunteers by centrifugation. Cells were observed under an inverted phase contrast microscope, and proliferative activity was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Stem cell surface markers were detected by flow cytometry. HUSCs were induced to differentiate into hepatocyte-like cells by coculturing with human hepatocyte L02 cells, which were confirmed by cellular morphology, messenger RNA expression of albumin (ALB), α-fetoprotein (AFP) and hepatocyte cytochrome P450 (CYP450) analyzed with quantitative reverse transcription polymerase chain reaction and the expression of glycogen detected by glycogen staining kits at 5, 10, and 15 days after coculturing. RESULTS HUSCs from urine were successfully isolated and cultured in vitro. At passages 3, the growth curve of hUSCs was S-shaped with good proliferation activity. Mesenchymal stem cell surface markers CD44 and CD90 were detected positive by flow cytometry. CD31 for endothelial cells and CD34 for hematopoietic stem cell markers were not detected. HUSCs gained the cellular morphology and function of hepatocyte cells including higher expression of several hepatocyte-specific genes such as ALB and some CYP450, lower expression of AFP and positive glycogen expression (P < .05) in coculturing with human hepatocyte L02 cells for 10-15d. CONCLUSIONS HUSCs can be induced to differentiate into hepatocyte-like cells by coculturing with human hepatocyte L02 cells for a certain number of days.
Collapse
Affiliation(s)
- Ming Zhou
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Liangliang Shen
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yinggu Qiao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhenxiao Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
8
|
Alizadeh R, Ramezanpour F, Mohammadi A, Eftekharzadeh M, Simorgh S, Kazemiha M, Moradi F. Differentiation of human olfactory system-derived stem cells into dopaminergic neuron-like cells: A comparison between olfactory bulb and mucosa as two sources of stem cells. J Cell Biochem 2019; 120:19712-19720. [PMID: 31297865 DOI: 10.1002/jcb.29277] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/20/2019] [Indexed: 12/11/2022]
Abstract
Cell transplantation has become a possible therapeutic approach in the treatment of neurodegenerative diseases of the nervous system by replacing lost cells. The current study aimed to make a comparison between the differentiation capacity of the olfactory bulb neural stem cells (OB-NSCs) and olfactory ectomesenchymal stem cells (OE-MSCs) into dopaminergic-like neurons under the inductive effect of transforming growth factor β (TGF-β). After culturing and treating with TGF-β, the differentiation capacities of both types of stem cells into dopaminergic neuron-like cells were evaluated. Quantitative real-time polymerase chain reaction analysis 3 weeks after induction demonstrated that the mRNA expression of the dopaminergic activity markers tyrosine hydroxylase (TH), dopamine transporter (DAT), paired box gene 2 (PAX2), and PAX5 in the neuron-like cells derived from OB-NSCs was significantly higher than those derived from OE-MSCs. These findings were further supported by the immunocytochemistry staining showing that the expression of the tyrosine hydroxylase, DAT, PAX2, and paired like homeodomain 3 seemed to be slightly higher in OB-NSCs compared with OE-MSCs. Despite the lower differentiation capacity of OE-MSCs, other considerations such as a noninvasive and easier harvesting process, faster proliferation attributes, longer life span, autologous transplantability, and also the easier and inexpensive cultural process of the OE-MSCs, cumulatively make these cells the more appropriate alternative in the case of autologous transplantation during the treatment process of neurodegenerative disorders like Parkinson's disease.
Collapse
Affiliation(s)
- Rafieh Alizadeh
- ENT and Head & Neck Research Center and Department, The Five Senses Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Farnaz Ramezanpour
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Mohammadi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mina Eftekharzadeh
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Simorgh
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Milad Kazemiha
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Moradi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Alizadeh R, Bagher Z, Kamrava SK, Falah M, Ghasemi Hamidabadi H, Eskandarian Boroujeni M, Mohammadi F, Khodaverdi S, Zare-Sadeghi A, Olya A, Komeili A. Differentiation of human mesenchymal stem cells (MSC) to dopaminergic neurons: A comparison between Wharton's Jelly and olfactory mucosa as sources of MSCs. J Chem Neuroanat 2019; 96:126-133. [PMID: 30639339 DOI: 10.1016/j.jchemneu.2019.01.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/15/2018] [Accepted: 01/09/2019] [Indexed: 12/28/2022]
Abstract
The generation of dopaminergic neurons from stem cells is a potential therapeutic approach to treat neurodegenerative disorders, such as Parkinson's disease. The current study aims to investigate the potential of two different types of mesenchymal stem cells derived from human Wharton's jelly and nasal cavity for differentiation into dopaminergic neurons. The differentiation capacities of both cell types were evaluated using real-time PCR, immunocytochemistry, flow cytometry and HPLC. Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) are noted for their capability to differentiate into mesodermal and non-mesodermal cells, including neurons. However, it was demonstrated that having the same neuroectodermal origin as the nervous system, the olfactory ectomesenchymal stem cells (OE-MSCs) expressed the neural marker MAP2 as well as dopaminergic markers such as tyrosine hydroxylase (TH), dopamine transporter (DAT) and PITX3 to a greater extent than the WJ-MSCs both at the level of mRNA and protein. Furthermore, quantitative flow cytometric evaluation of these markers at 12 days post-induction supported the above-mentioned results. Finally, the assessment of the functionality of differentiated cells and their ability to synthesize dopamine measured by HPLC revealed that the OE-MSC-derived dopaminergic cells released almost the same amount of dopamine as that secreted by WJ-MSC-derived cells. Thus it showed the difference in their functionality to be negligible. Overall, it may be concluded that higher proliferation and differentiation capacity of OE-MSCs, along with their easier harvestability and autologous transplantability compared with WJ-MSCs, makes them a better cell source for stem cell therapy of neurodegenerative disorders which are caused by degeneration of dopaminergic neurons.
Collapse
Affiliation(s)
- Rafieh Alizadeh
- ENT and Head & Neck Research Center and Department, The five Senses Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Zohreh Bagher
- ENT and Head & Neck Research Center and Department, The five Senses Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Kamran Kamrava
- ENT and Head & Neck Research Center and Department, The five Senses Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Falah
- ENT and Head & Neck Research Center and Department, The five Senses Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Hatef Ghasemi Hamidabadi
- Department of Anatomy & Cell Biology, Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahdi Eskandarian Boroujeni
- Department of Stem Cells and Regenerative Medicine, Faculty of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Fatemeh Mohammadi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Khodaverdi
- Endometriosis Research Center, University of Medical Sciences, Tehran, Iran
| | - Arash Zare-Sadeghi
- Skull Base Research Center, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Arta Olya
- Department of Stem Cells and Regenerative Medicine, Faculty of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Ali Komeili
- Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
10
|
Human olfactory stem cells: As a promising source of dopaminergic neuron-like cells for treatment of Parkinson's disease. Neurosci Lett 2018; 696:52-59. [PMID: 30552942 DOI: 10.1016/j.neulet.2018.12.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/11/2018] [Accepted: 12/04/2018] [Indexed: 01/06/2023]
Abstract
The production of dopaminergic (DA) neurons from stem cells holds a great promise for future clinical treatment of neurodegenerative diseases, such as Parkinson's disease (PD). Olfactory ecto-mesenchymal stem cells (OE-MSCs) derived from the adult human olfactory mucosa can be easily isolated and expanded in culture while maintaining their immense plasticity. In addition to reduced ethical concerns, OE-MSCs could be transplanted across allogeneic barriers, making them desirable stem cells for clinical applications. The goal of this study was to define the potentiality of human olfactory mucosa-derived MSCs aimed at differentiation into DA neuron-like cells. OE-MSCs were induced to differentiate to DA neuron-like cells in vitro by using sonic hedgehog (SHH), fibroblast growth factor 8 (FGF8), basic fibroblast growth factor (bFGF), Glial cell line-derived neurotrophic factor (GDNF) and brain derived neurotrophic factor (BDNF). Then the differentiated neurons were characterized for expression of DA neuron markers by Real-time PCR, immunocytochemistry and flow cytometry. Our findings showed that differentiated OE-MSCs could significantly express DA neuron markers at mRNA and protein levels along with dopamine release 12 days post-differentiation. These results support the viability and feasibility of using OE-MSCs as a source of in vitro generated DA neuron-like cells for treatment of DA disorders namely PD.
Collapse
|