1
|
Li Z, Xu T, Li X, Wang T, Tang G, Zhao H, Zhao Y, Ye K, Gao P. Viral integration promotes SV40T-induced immortalization by disturbing the expression of DNA/chromosome- and ECM-associated functional genes. Gene 2024; 896:148060. [PMID: 38048968 DOI: 10.1016/j.gene.2023.148060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/28/2023] [Accepted: 12/01/2023] [Indexed: 12/06/2023]
Abstract
Lentivirus containing simian virus 40 large T antigen (SV40T) is routinely used to induce cell immortalization. However, the roles of viral integration itself in this progress is still controversial. Here, we transformed primary mouse embryonic fibroblasts (MEFs) with SV40T lentivirus and studied the roles of viral integration in the immortalization using RNA sequencing (RNA-seq) and whole genome sequencing (WGS). During the immortalization, differentially expressed genes (DGEs) are enriched in viral infection and several diverse activities. However, DEGs between immortalized and aging cells are significantly enriched in DNA/chromosome- and extracellular matrix (ECM)-associated activities. Gene regulatory network (GRN) analysis shows that although p53 is a key regulatory factor, many other transcription factors also play critical roles in the process, like STAT1. Of these DEGs, 32 genes have viral integration in their coding and/or regulatory regions. Our findings suggest that viral integration may promote SV40T-mediated immortalization by disturbing the expression of DNA/chromosome- and ECM-associated genes.
Collapse
Affiliation(s)
- Zihang Li
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Tun Xu
- MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China; School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xiujuan Li
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Tingjie Wang
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Guangbo Tang
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Huanhuan Zhao
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yuanli Zhao
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Kai Ye
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China; School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China; School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China; Faculty of Science, Leiden University, Leiden, the Netherlands.
| | - Peng Gao
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
2
|
Heyer V, Reina-San-Martin B. Optimal AID expression and efficient immunoglobulin class switch recombination are dependent on the hypoxia-inducible factor. Eur J Immunol 2023; 53:e2350373. [PMID: 37143384 DOI: 10.1002/eji.202350373] [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: 01/12/2023] [Revised: 03/31/2023] [Accepted: 05/02/2023] [Indexed: 05/06/2023]
Abstract
During immune responses, B cells engaging a cognate antigen are recruited to GCs in secondary lymphoid organs where they will diversify their BCR to generate highly specific and adapted humoral responses. They do so, by inducing the expression of activation-induced cytidine deaminase (AID), which initiates somatic hypermutation (SHM) and class switch recombination (CSR). AID deaminates cytosines in ss DNA, generating U:G mismatches that are processed to induce ds DNA break intermediates during CSR that result in the expression of a different antibody isotype. Interestingly, hypoxia regions have been reported in GCs and suggesting that hypoxia could modulate the humoral response. Furthermore, hypoxia inducible transcription factor (HIF) can bind to the AID promoter and induce AID expression in a non-B-cell setting, suggesting that it might be involved in the transcriptional induction of AID in B cells, hence, regulating SHM and CSR. We, thus, hypothesized that HIF could regulate the efficiency of CSR. Here, we show that the inactivation of both the HIF-1α and HIF-1β subunits of the HIF transcription factor in murine CH12 B cells results in defective CSR and that this is due to the suboptimal induction of AID expression.
Collapse
Affiliation(s)
- Vincent Heyer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Bernardo Reina-San-Martin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), Illkirch, France
- Université de Strasbourg, Illkirch, France
| |
Collapse
|
3
|
Chen Y, Lu X, Gao L, Dean DC, Liu Y. Spheroid-induced heterogeneity and plasticity of uveal melanoma cells. Cell Oncol (Dordr) 2022; 45:309-321. [PMID: 35404029 PMCID: PMC9050762 DOI: 10.1007/s13402-022-00671-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2022] [Indexed: 11/29/2022] Open
Abstract
PURPOSE The mechanism underlying cancer heterogeneity and plasticity remains elusive, in spite of the fact that multiple hypotheses have been put forward. We intended to clarify this heterogeneity in uveal melanoma (UM) by looking for evidence of cancer stem cell involvement and a potential role of ZEB1 in cancer cell plasticity. METHODS Spheroids derived from human UM cells as well as xenograft tumors in nude mice were dissected for signs of heterogeneity and plasticity. Two human UM cell lines were studied: the epithelioid type C918 cell line and the spindle type OCM1 cell line. We knocked down ZEB1 in both cell lines to investigate its involvement in the regulation of stem-like cell formation and vascularization by qRT-PCR, immunohistochemistry, flow cytometry, electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) assays. RESULTS We found that a small side population (SP) in OCM1 showed stem cell-like properties such as heterogeneity, remote dissemination and nuclear dye exclusion after spheroid formation in vitro. ZEB1 regulated UM stem cell generation indirectly by promoting cell proliferation to form large size tumors in vivo and spheroid in vitro, and directly by binding to stemness genes such as TERT and ABCB1. In addition, we found that ZEB1 participates in vasculogenic mimicry system formation through the regulation of CD34 and VE-cadherin expression. CONCLUSIONS From our data we conclude that cancer stem cells may contribute to UM heterogeneity and plasticity and that ZEB1 may play a regulatory role in it.
Collapse
Affiliation(s)
- Yao Chen
- Hunan Key Laboratory of Ophthalmology, Eye Center of Xiangya Hospital, Central South University, National Clinical Medical Center for Geriatric Diseases of Xiangya Hospital, Changsha, China
| | - Xiaoqin Lu
- Department of Medicine, James Graham Brown Cancer Center, Birth Defects Center, University of Louisville School of Medicine, Louisville, KY USA
| | - Ling Gao
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Douglas C. Dean
- Department of Medicine, James Graham Brown Cancer Center, Birth Defects Center, University of Louisville School of Medicine, Louisville, KY USA
| | - Yongqing Liu
- Department of Medicine, James Graham Brown Cancer Center, Birth Defects Center, University of Louisville School of Medicine, Louisville, KY USA
| |
Collapse
|
4
|
Zeb1 Regulation of Wound Healing-Induced Inflammation in Alkali-Damaged Corneas. iScience 2022; 25:104038. [PMID: 35340433 PMCID: PMC8941209 DOI: 10.1016/j.isci.2022.104038] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/24/2021] [Accepted: 03/03/2022] [Indexed: 11/22/2022] Open
Abstract
The cornea is an avascular tissue for vision clarity. Alkali burn could cause severe traumatic damage on the cornea with inflammation and neovascularization (NV), leading to vision reduction and blindness. Mechanisms underlying corneal inflammation and NV are not as clear. We previously reported that Zeb1 is an important factor in corneal NV, and we sought to clarify whether it is also involved in regulation of corneal inflammation. We analyzed the alkali burn-induced corneal inflammation and wound healing in both Zeb1+/+ and Zeb1−/+ littermates through a multidisciplinary approach. We provide evidence that Zeb1 forms a positive regulatory loop with Tgfb to regulate early corneal inflammation by maintenance of immune cell viability and mobility and later wound healing by activation of both Nf-κb and Tgfb-related Stat3 signaling pathways. We believe that ZEB1 is a potential therapeutic target, and inactivation of ZEB1 could be a strategy to treat severe corneal inflammation condition. Traumatic wound induces inflammation in the cornea, resulting in vision reduction Zeb1 is a key factor to retain immune cell viability, mobility, and cytokine expression Zeb1 regulates cytokine gene expression through both Nf-κb and Stat3 pathways Inactivation of ZEB1 could be a strategy to treat severe corneal inflammation condition
Collapse
|
5
|
Yaglova NV, Obernikhin SS, Tsomartova DA, Nazimova SV, Yaglov VV, Tsomartova ES, Chereshneva EV, Ivanova MY, Lomanovskaya TA. Impaired Morphogenesis and Function of Rat Adrenal Zona Glomerulosa by Developmental Low-Dose Exposure to DDT Is Associated with Altered Oct4 Expression. Int J Mol Sci 2021; 22:6324. [PMID: 34204839 PMCID: PMC8231536 DOI: 10.3390/ijms22126324] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/25/2021] [Accepted: 05/29/2021] [Indexed: 12/17/2022] Open
Abstract
Dichlorodiphenyltrichloroethane (DDT) is a persistent organic pollutant and one of the most widespread endocrine disrupting chemicals. The impact of low-dose exposure to DDT on the morphogenesis of the adrenal gland is still poorly understood. The development and function of zona glomerulosa in rats has been found to be associated with changes in the expression of the transcription factor Oct4 (Octamer 4), which is the most important player in cell pluripotency. The aim of the study was to investigate the morphogenesis and function of rat adrenal zona glomerulosa in rats exposed to low doses of DDT during prenatal and postnatal development and to determine the possible role of Oct4 in DDT-mediated structural and functional changes. The DDT-exposed rats demonstrated slower development and lower functional activity of the zona glomerulosa during the pubertal period associated with higher expression of Oct4. Further, accelerated growth and restoration of hormone production was associated with, firstly, a decrease in Oct4 expressing cells and secondly, the loss of the inverse relationship between basal aldosterone levels and the number of Oct4 expressing cells. Thus, the transcriptional factor Oct4 exhibited an altered pattern of expression in the DDT-exposed rats during postnatal development. The results of the study uncover a novel putative mechanism by which low doses of DDT disrupt the development of adrenal zona glomerulosa.
Collapse
Affiliation(s)
- Nataliya V. Yaglova
- Laboratory of Endocrine System Development, Federal State Budgetary Scientific Institution “Research Institute of Human Morphology”, 117418 Moscow, Russia; (S.S.O.); (D.A.T.); (S.V.N.); (V.V.Y.); (E.S.T.)
| | - Sergey S. Obernikhin
- Laboratory of Endocrine System Development, Federal State Budgetary Scientific Institution “Research Institute of Human Morphology”, 117418 Moscow, Russia; (S.S.O.); (D.A.T.); (S.V.N.); (V.V.Y.); (E.S.T.)
| | - Dibakhan A. Tsomartova
- Laboratory of Endocrine System Development, Federal State Budgetary Scientific Institution “Research Institute of Human Morphology”, 117418 Moscow, Russia; (S.S.O.); (D.A.T.); (S.V.N.); (V.V.Y.); (E.S.T.)
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| | - Svetlana V. Nazimova
- Laboratory of Endocrine System Development, Federal State Budgetary Scientific Institution “Research Institute of Human Morphology”, 117418 Moscow, Russia; (S.S.O.); (D.A.T.); (S.V.N.); (V.V.Y.); (E.S.T.)
| | - Valentin V. Yaglov
- Laboratory of Endocrine System Development, Federal State Budgetary Scientific Institution “Research Institute of Human Morphology”, 117418 Moscow, Russia; (S.S.O.); (D.A.T.); (S.V.N.); (V.V.Y.); (E.S.T.)
| | - Elina S. Tsomartova
- Laboratory of Endocrine System Development, Federal State Budgetary Scientific Institution “Research Institute of Human Morphology”, 117418 Moscow, Russia; (S.S.O.); (D.A.T.); (S.V.N.); (V.V.Y.); (E.S.T.)
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| | - Elizaveta V. Chereshneva
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| | - Marina Y. Ivanova
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| | - Tatiana A. Lomanovskaya
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| |
Collapse
|
6
|
Park JB, Seo J, Park JW, Chun YS. Neddylation blockade induces HIF-1α driven cancer cell migration via upregulation of ZEB1. Sci Rep 2020; 10:18210. [PMID: 33097763 PMCID: PMC7585416 DOI: 10.1038/s41598-020-75286-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
Neddylation is a process by which NEDD8 is covalently conjugated to target proteins by sequential enzymatic reaction. Its role in cancer cell migration has only been recently acknowledged. Previously in cancer cell migration, the epithelial to mesenchymal transition (EMT) process has been well-known to play an important role in both invasion and metastasis by promoting mesenchymal phenotype in epithelial cells. However, the role of neddylation in the EMT process and its mechanistic details are yet to be elucidated. We recently reported that neddylation plays a crucial role in cancer cell migration through the PI3K-Akt pathway. Here, we report that inhibiting neddylation activates the hypoxia-inducible factor 1α (HIF-1α) through the PI3K-Akt pathway, which eventually regulates the EMT-activator ZEB1 (zinc finger E-box binding homeobox 1) in various cancer cell lines. As induction of HIF-1α is known to deteriorate the state of cancer and EMT process is one of the hallmarks of metastasis in cancer, our findings uncover the role of neddylation between HIF-1α and ZEB1.
Collapse
Affiliation(s)
- Jun Bum Park
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 110-799, Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 110-799, Korea
| | - Jieun Seo
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 110-799, Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 110-799, Korea
| | - Jong-Wan Park
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 110-799, Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 110-799, Korea
| | - Yang-Sook Chun
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 110-799, Korea. .,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 110-799, Korea. .,Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 110-799, Korea.
| |
Collapse
|
7
|
Li K, Ning T, Wang H, Jiang Y, Zhang J, Ge Z. Nanosecond pulsed electric fields enhance mesenchymal stem cells differentiation via DNMT1-regulated OCT4/NANOG gene expression. Stem Cell Res Ther 2020; 11:308. [PMID: 32698858 PMCID: PMC7374836 DOI: 10.1186/s13287-020-01821-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/25/2020] [Accepted: 07/09/2020] [Indexed: 02/28/2023] Open
Abstract
Background Multiple strategies have been proposed to promote the differentiation potential of mesenchymal stem cells (MSCs), which is the fundamental property in tissue formation and regeneration. However, these strategies are relatively inefficient that limit the application. In this study, we reported a novel and efficient strategy, nanosecond pulsed electric fields (nsPEFs) stimulation, which can enhance the trilineage differentiation potential of MSCs, and further explained the mechanism behind. Methods We used histological staining to screen out the nsPEFs parameters that promoted the trilineage differentiation potential of MSCs, and further proved the effect of nsPEFs by detecting the functional genes. In order to explore the corresponding mechanism, we examined the expression of pluripotency genes and the methylation status of their promoters. Finally, we targeted the DNA methyltransferase which was affected by nsPEFs. Results The trilineage differentiation of bone marrow-derived MSCs was significantly enhanced in vitro by simply pre-treating with 5 pulses of nsPEFs stimulation (energy levels as 10 ns, 20 kV/cm; 100 ns, 10 kV/cm), due to that the nsPEFs demethylated the promoters of stem cell pluripotency genes OCT4 and NANOG through instantaneous downregulation of DNA methylation transferase 1 (DNMT1), thereby increasing the expression of OCT4 and NANOG for up to 3 days, and created a treatment window period of stem cells. Conclusions In summary, nsPEFs can enhance MSCs differentiation via the epigenetic regulation and could be a safe and effective strategy for future stem cell application.
Collapse
Affiliation(s)
- Kejia Li
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Tong Ning
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Hao Wang
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Yangzi Jiang
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jue Zhang
- Institute of Biomechanics and Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Zigang Ge
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China.
| |
Collapse
|
8
|
Chen J, Huang Y, Yang J, Li K, Jiang Y, Heng BC, Cai Q, Zhang J, Ge Z. Multiple nanosecond pulsed electric fields stimulation with conductive poly(
l
‐lactic acid)/carbon nanotubes films maintains the multipotency of mesenchymal stem cells during prolonged in vitro culture. J Tissue Eng Regen Med 2020; 14:1136-1148. [DOI: 10.1002/term.3088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/03/2020] [Accepted: 06/06/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Jiaqing Chen
- Department of Biomedical Engineering, College of EngineeringPeking University Beijing China
| | - Yiqian Huang
- State Key Laboratory of Organic‐Inorganic Composites, Beijing Laboratory of Biomedical MaterialsBeijing University of Chemical Technology Beijing China
| | - Jiabei Yang
- Department of Biomedical Engineering, College of EngineeringPeking University Beijing China
| | - Kejia Li
- Department of Biomedical Engineering, College of EngineeringPeking University Beijing China
| | - Yangzi Jiang
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of MedicineThe Chinese University of Hong Kong Hong Kong China
| | - Boon Chin Heng
- Central LaboratoryPeking University School of Stomatology Beijing Beijing China
| | - Qing Cai
- State Key Laboratory of Organic‐Inorganic Composites, Beijing Laboratory of Biomedical MaterialsBeijing University of Chemical Technology Beijing China
| | - Jue Zhang
- Academy for Advanced Interdisciplinary StudiesPeking University Beijing China
| | - Zigang Ge
- Department of Biomedical Engineering, College of EngineeringPeking University Beijing China
| |
Collapse
|
9
|
Liu X, Chen F, Chen Y, Lu H, Lu X, Peng X, Kaplan HJ, Dean DC, Gao L, Liu Y. Paracrine effects of intraocularly implanted cells on degenerating retinas in mice. Stem Cell Res Ther 2020; 11:142. [PMID: 32234075 PMCID: PMC7326149 DOI: 10.1186/s13287-020-01651-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/18/2020] [Accepted: 03/12/2020] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Retinal degeneration is a leading cause of blindness in the world; its etiology is complex and involves genetic defects and stress-associated aging. In addition to gene therapies for known genetically defective retinal degeneration, cellular therapies have been widely explored for restoring vision in both preclinical animal models and clinical trials. Stem cells of distinct tissue sources and their derived lineages have been tested for treating retinal degeneration; most of them were reported to be effective to some extent in restoring/improving deteriorated vision. Whether this visual improvement is due to a functional integration of grafted cells to substitute for lost retinal neurons in recipients or due to their neuroprotective and neurotrophic effects to retain recipient functional neurons, or both, is still under debate. METHODS We compared the results of subretinal transplantation of various somatic cell types, such as stem cells and differentiated cells, into RhoP23H/+ mice, a retinal degeneration model for human retinitis pigmentosa (RP) by evaluating their optokinetic response (OKR) and retinal histology. We identified some paracrine factors in the media that cultured cells secreted by western blotting (WB) and functionally evaluated the vascular endothelial growth factor Vegfa for its potential neurotrophic and neuroprotective effects on the neuroretina of model animals by intravitreal injection of VEGF antibody. RESULTS We found that live cells, regardless of whether they were stem cells or differentiated cell types, had a positive effect on improving degenerating retinas after subretinal transplantation; the efficacy depended on their survival duration in the host tissue. A few paracrine factors were identified in cell culture media; Vegfa was the most relevant neurotrophic and neuroprotective factor identified by our experiments to extend neuron survival duration in vivo. CONCLUSIONS Cellular therapy-produced benefits for remediating retinal degeneration are mostly, if not completely, due to a paracrine effect of implanted cells on the remaining host retinal neurons.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Department of Ophthalmology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Fenghua Chen
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yao Chen
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Department of Ophthalmology, Xiangya Hospital of Central South University, Changsha, China
| | - Huayi Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiaoqin Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
| | - Xiaoyan Peng
- Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Henry J Kaplan
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
| | - Douglas C Dean
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA.
- James Graham Brown Cancer Center, Louisville, USA.
- Birth Defects Center, University of Louisville School of Medicine, Louisville, KY, USA.
| | - Ling Gao
- Department of Ophthalmology, Second Xiangya Hospital of Central South University, Changsha, China.
| | - Yongqing Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, KY, 40202, USA.
- James Graham Brown Cancer Center, Louisville, USA.
- Birth Defects Center, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
10
|
Chen F, Liu X, Chen Y, Liu JY, Lu H, Wang W, Lu X, Dean KC, Gao L, Kaplan HJ, Dean DC, Peng X, Liu Y. Sphere-induced reprogramming of RPE cells into dual-potential RPE stem-like cells. EBioMedicine 2020; 52:102618. [PMID: 31982829 PMCID: PMC6994567 DOI: 10.1016/j.ebiom.2019.102618] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 11/20/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022] Open
Abstract
Background The retinal pigment epithelium (RPE) has the potential to regenerate the entire neuroretina upon retinal injury in amphibians. In contrast, this regenerative capacity has been lost in mammals. The reprogramming of differentiated somatic cells into induced pluripotent stem cells (iPSCs) by viral transduction of exogenous stem cell factors has triggered a revolution in regenerative medicine. However, the risks of potential mutation(s) caused by random viral vector insertion in host genomes and tumor formation in recipients hamper its clinical application. One alternative is to immortalize adult stem cells with limited potential or to partially reprogram differentiated somatic cells into progenitor-like cells through non-integration protocols. Methods Sphere-induced RPE stem cells (iRPESCs) were generated from adult mouse RPE cells. Their stem cell functionality was studied in a mouse model of retinal degeneration. The molecular mechanism underlying the sphere-induced reprogramming was investigated using microarray and loss-of-function approaches. Findings We provide evidence that our sphere-induced reprogramming protocol can immortalize and transform mouse RPE cells into iRPESCs with dual potential to differentiate into cells that express either RPE or photoreceptor markers both in vitro and in vivo. When subretinally transplanted into mice with retinal degeneration, iRPESCs can integrate to the RPE and neuroretina, thereby delaying retinal degeneration in the model animals. Our molecular analyses indicate that the Hippo signaling pathway is important in iRPESC reprogramming. Interpretation The Hippo factor Yap1 is activated in the nuclei of cells at the borders of spheres. The factors Zeb1 and P300 downstream of the Hippo pathway are shown to bind to the promoters of the stemness genes Oct4, Klf4 and Sox2, thereby likely transactivate them to reprogram RPE cells into iRPESCs. Fund National Natural Science Foundation of China and the National Institute of Health USA.
Collapse
Affiliation(s)
- Fenghua Chen
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China
| | - Xiao Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; Department of Ophthalmology, Second Affiliated Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Yao Chen
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
| | - John Y Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Huayi Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Wei Wang
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Xiaoqin Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Kevin C Dean
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Ling Gao
- Department of Ophthalmology, Second Affiliated Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Henry J Kaplan
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Douglas C Dean
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA; Birth Defects Center; University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| | - Xiaoyan Peng
- Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China.
| | - Yongqing Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA; Birth Defects Center; University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| |
Collapse
|
11
|
Lo LM, Raghunath M, Lee KKH. Growing Human Dermal Fibroblasts as Spheroids Renders Them Susceptible for Early Expression of Pluripotency Genes. ACTA ACUST UNITED AC 2019; 3:e1900094. [DOI: 10.1002/adbi.201900094] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/26/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Lok Man Lo
- MOE Key Laboratory for Regenerative MedicineSchool of Biomedical SciencesChinese University of Hong Kong
- Joint Chinese University of Hong Kong – University of Southampton Laboratory for Stem Cell and Regenerative MedicineSchool of Biomedical SciencesChinese University of Hong Kong
| | - Michael Raghunath
- Center for Cell Biology and Tissue EngineeringInstitute for Chemistry and BiotechnologyZurich University for Applied Sciences (ZHAW) Switzerland
| | - Kenneth K. H. Lee
- MOE Key Laboratory for Regenerative MedicineSchool of Biomedical SciencesChinese University of Hong Kong
- Joint Chinese University of Hong Kong – University of Southampton Laboratory for Stem Cell and Regenerative MedicineSchool of Biomedical SciencesChinese University of Hong Kong
| |
Collapse
|
12
|
Atkinson SP. A Preview of Selected Articles. Stem Cells Transl Med 2019. [PMCID: PMC6646695 DOI: 10.1002/sctm.19-0198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
13
|
Primitive Cancer Cell States: A Target for Drug Screening? Trends Pharmacol Sci 2019; 40:161-171. [DOI: 10.1016/j.tips.2019.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/05/2018] [Accepted: 01/07/2019] [Indexed: 12/26/2022]
|
14
|
Chen X, Li J, Huang Y, Liu P, Fan Y. Insoluble Microenvironment Facilitating the Generation and Maintenance of Pluripotency. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:267-278. [PMID: 29327674 DOI: 10.1089/ten.teb.2017.0415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Induced pluripotent stem cells (iPSCs) hold enormous potential as a tool to generate cells for tissue engineering and regenerative medicine. Since the initial report of iPSCs in 2006, many different methods have been developed to enhance the safety and efficiency of this technology. Recent studies indicate that the extracellular signals can promote the production of iPSCs, and even replace the Yamanaka factors. Noticeably, abundant evidences suggest that the insoluble microenvironment, including the culture substrate and neighboring cells, directly regulates the expression of core pluripotency genes and the epigenetic modification of the chromatins, hence, impacts the reprogramming dynamics. These studies provide new strategies for developing safer and more efficient method for iPSC generation. In this review, we examine the publications addressing the insoluble extracellular microenvironment that boosts iPSC generation and self-renewal. We also discuss cell adhesion-mediated molecular mechanisms, through which the insoluble extracellular cues interplay with reprogramming.
Collapse
Affiliation(s)
- Xiaofang Chen
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
- 2 Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University , Beijing, China
| | - Jiaqi Li
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
| | - Yan Huang
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
- 2 Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University , Beijing, China
| | - Peng Liu
- 3 Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University , Beijing, China
| | - Yubo Fan
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
- 2 Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University , Beijing, China
- 4 National Research Center for Rehabilitation Technical Aids , Beijing, China
| |
Collapse
|
15
|
Mitotic polarization of transcription factors during asymmetric division establishes fate of forming cancer cells. Nat Commun 2018; 9:2424. [PMID: 29930325 PMCID: PMC6013470 DOI: 10.1038/s41467-018-04663-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/09/2018] [Indexed: 12/20/2022] Open
Abstract
A model of K-Ras-initiated lung cancer was used to follow the transition of precancerous adenoma to adenocarcinoma. In hypoxic, Tgf-β1-rich interiors of adenomas, we show that adenoma cells divide asymmetrically to produce cancer-generating cells highlighted by epithelial mesenchymal transition and a CD44/Zeb1 loop. In these cells, Zeb1 represses the Smad inhibitor Zeb2/Sip1, causing Pten loss and launching Tgf-β1 signaling that drives nuclear translocation of Yap1. Surprisingly, the nuclear polarization of transcription factors during mitosis establishes parent and daughter fates prior to cytokinesis in sequential asymmetric divisions that generate cancer cells from precancerous lesions. Mutation or knockdown of Zeb1 in the lung blocked the production of CD44hi, Zeb1hi cancer-generating cells from adenoma cells. A CD44/Zeb1 loop then initiates two-step transition of precancerous cells to cancer cells via a stable intermediate population of cancer-generating cells. We show these initial cancer-generating cells are independent of cancer stem cells generated in tumors by p53-regulated reprogramming of existing cancer cells. Transition from premalignant lesion to cancer cell highlights tumor initiation. Here, the authors use a model of K-Ras-initiated lung cancer to document two successive asymmetric divisions, each driven by mitotic polarization of key transcription factors, which lead to generation of initial cancer cells.
Collapse
|
16
|
Abstract
The present review aimed to assess the networks of transcription factors regulating the Oct4 expression in mice. Through a comprehensive analysis of the binding sites and the interrelationships of the transcription factors of Oct4, it is found that transcription factors of Oct4 form three regulating complexes centered by Oct4-Sox2, Nanog, and Lrh1. They bind on CR4, CR2, and CR1 regions of Oct4 promoter/enhancer, respectively, to activate Oct4 transcription synergistically. This article also discusses the mechanisms of fine-tuning the Oct4 expression. These findings have important implications in the field of stem cell and developmental biology.
Collapse
Affiliation(s)
- Yu-Qiang Li
- Marine College, Shandong University (Weihai) , Weihai, China
| |
Collapse
|
17
|
Prasad P, Mittal SA, Chongtham J, Mohanty S, Srivastava T. Hypoxia-Mediated Epigenetic Regulation of Stemness in Brain Tumor Cells. Stem Cells 2017; 35:1468-1478. [DOI: 10.1002/stem.2621] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 02/21/2017] [Accepted: 03/08/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Pankaj Prasad
- Department of Genetics; University of Delhi South Campus; New Delhi India
| | | | - Jonita Chongtham
- Department of Genetics; University of Delhi South Campus; New Delhi India
| | - Sujata Mohanty
- Stem Cell Facility, All India Institute of Medical Sciences; New Delhi India
| | - Tapasya Srivastava
- Department of Genetics; University of Delhi South Campus; New Delhi India
| |
Collapse
|
18
|
Xu N, Chen Y, Dean KC, Lu X, Liu X, Wang W, Dean DC, Kaplan HJ, Gao L, Dong F, Liu Y. Sphere-Induced Rejuvenation of Swine and Human Müller Glia Is Primarily Caused by Telomere Elongation. Stem Cells 2017; 35:1579-1591. [PMID: 28152565 DOI: 10.1002/stem.2585] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/17/2017] [Accepted: 01/24/2017] [Indexed: 02/01/2023]
Abstract
Müller cells are the major supportive and protective glial cells in the retina with important functions in histogenesis and synaptogenesis during development, and in maintenance of mature neurons as they show to secrete various cytokines and manifest potentials of self-renewal and transdifferentiation into retinal neurons following injury in the vertebrate retinas. The swine retina has a visual streak structure similar to the human macular where cone photoreceptors are highly concentrated, thereby can serve as a better model for studying retinal diseases and for formulating cell-based therapeutics than the rodent retinas. Like most differentiated somatic mammalian cells, the isolated swine and human Müller glia become senescent over passages in culture, which restricts their potential application in basic and clinic researches. Here, we demonstrate that the senescence of swine and human Müller cells is caused by telomere attrition upon multiplications in vitro; and the senescent cells can be rejuvenated by sphere suspension culture. We also provide evidence that sphere-induced extension of telomeres in swine and human Müller glia is achieved by alternative lengthening of telomeres or/and by telomerase activation. Stem Cells 2017;35:1579-1591.
Collapse
Affiliation(s)
- Ni Xu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky, USA.,Department of Ophthalmology, Peking Union Medical College Hospital, Beijing, China
| | - Yao Chen
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky, USA.,Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
| | - Kevin C Dean
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Xiaoqin Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Xiao Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky, USA.,Department of Ophthalmology, the Second Affiliated Hospital, Central South University Xiangya School of Medicine, Changsha, China
| | - Wei Wang
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Douglas C Dean
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Henry J Kaplan
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Ling Gao
- Department of Ophthalmology, the Second Affiliated Hospital, Central South University Xiangya School of Medicine, Changsha, China
| | - Fangtian Dong
- Department of Ophthalmology, Peking Union Medical College Hospital, Beijing, China
| | - Yongqing Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
19
|
Engel M, Do-Ha D, Muñoz SS, Ooi L. Common pitfalls of stem cell differentiation: a guide to improving protocols for neurodegenerative disease models and research. Cell Mol Life Sci 2016; 73:3693-709. [PMID: 27154043 PMCID: PMC5002043 DOI: 10.1007/s00018-016-2265-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/05/2016] [Accepted: 05/03/2016] [Indexed: 12/17/2022]
Abstract
Induced pluripotent stem cells and embryonic stem cells have revolutionized cellular neuroscience, providing the opportunity to model neurological diseases and test potential therapeutics in a pre-clinical setting. The power of these models has been widely discussed, but the potential pitfalls of stem cell differentiation in this research are less well described. We have analyzed the literature that describes differentiation of human pluripotent stem cells into three neural cell types that are commonly used to study diseases, including forebrain cholinergic neurons for Alzheimer's disease, midbrain dopaminergic neurons for Parkinson's disease and cortical astrocytes for neurodegenerative and psychiatric disorders. Published protocols for differentiation vary widely in the reported efficiency of target cell generation. Additionally, characterization of the cells by expression profile and functionality differs between studies and is often insufficient, leading to highly variable protocol outcomes. We have synthesized this information into a simple methodology that can be followed when performing or assessing differentiation techniques. Finally we propose three considerations for future research, including the use of physiological O2 conditions, three-dimensional co-culture systems and microfluidics to control feeding cycles and growth factor gradients. Following these guidelines will help researchers to ensure that robust and meaningful data is generated, enabling the full potential of stem cell differentiation for disease modeling and regenerative medicine.
Collapse
Affiliation(s)
- Martin Engel
- Illawarra Health and Medical Research Institute, School of Biological Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Dzung Do-Ha
- Illawarra Health and Medical Research Institute, School of Biological Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Sonia Sanz Muñoz
- Illawarra Health and Medical Research Institute, School of Biological Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute, School of Biological Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia.
| |
Collapse
|
20
|
Jeon S, Oh IH. Regeneration of the retina: toward stem cell therapy for degenerative retinal diseases. BMB Rep 2016; 48:193-9. [PMID: 25560700 PMCID: PMC4436854 DOI: 10.5483/bmbrep.2015.48.4.276] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Indexed: 01/12/2023] Open
Abstract
Degenerative retinal diseases affect millions of people worldwide, which can lead to the loss of vision. However, therapeutic approaches that can reverse this process are limited. Recent efforts have allowed the possibility of the stem cell-based regeneration of retinal cells and repair of injured retinal tissues. Although the direct differentiation of pluripotent stem cells into terminally differentiated photoreceptor cells comprises one approach, a series of studies revealed the intrinsic regenerative potential of the retina using endogenous retinal stem cells. Muller glial cells, ciliary pigment epithelial cells, and retinal pigment epithelial cells are candidates for such retinal stem cells that can differentiate into multiple types of retinal cells and be integrated into injured or developing retina. In this review, we explore our current understanding of the cellular identity of these candidate retinal stem cells and their therapeutic potential for cell therapy against degenerative retinal diseases. [BMB Reports 2015; 48(4): 193-199]
Collapse
Affiliation(s)
- Sohee Jeon
- Catholic High-Performance Cell Therapy Center & Department of Medical Lifescience; Department of Ophthalmology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul 137-701, Korea
| | - Il-Hoan Oh
- Catholic High-Performance Cell Therapy Center & Department of Medical Lifescience, The Catholic University of Korea, Seoul 137-701, Korea
| |
Collapse
|
21
|
Ni A, Wu MJ, Chavala SH. Sphere formation permits Oct4 reprogramming of ciliary body epithelial cells into induced pluripotent stem cells. Stem Cells Dev 2015; 23:3065-71. [PMID: 25027059 DOI: 10.1089/scd.2014.0064] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Somatic cells can be reprogrammed to induced pluripotent stem (iPS) cells by defined sets of transcription factors. We previously described reprogramming of monolayer-cultured adult mouse ciliary body epithelial (CE) cells by Oct4 and Klf4, but not with Oct4 alone. In this study, we report that Oct4 alone is sufficient to reprogram CE cells to iPS cells through sphere formation. Furthermore, we demonstrate that sphere formation induces a partial reprogramming state characterized by expression of retinal progenitor markers, upregulation of reprogramming transcription factors, such as Sall4 and Nanog, demethylation in the promoter regions of pluripotency associated genes, and mesenchymal to epithelial transition. The Oct4-iPS cells maintained normal karyotypes, expressed markers for pluripotent stem cells, and were capable of differentiating into derivatives of all three embryonic germ layers in vivo and in vitro. These findings suggest that sphere formation may render somatic cells more susceptible to reprogramming.
Collapse
Affiliation(s)
- Aiguo Ni
- Department of Ophthalmology, University of North Carolina , Chapel Hill, North Carolina
| | | | | |
Collapse
|
22
|
Mirakhori F, Zeynali B, Rassouli H, Shahbazi E, Hashemizadeh S, Kiani S, Salekdeh GH, Baharvand H. Induction of Neural Progenitor-Like Cells from Human Fibroblasts via a Genetic Material-Free Approach. PLoS One 2015; 10:e0135479. [PMID: 26266943 PMCID: PMC4534403 DOI: 10.1371/journal.pone.0135479] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 07/22/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND A number of studies generated induced neural progenitor cells (iNPCs) from human fibroblasts by viral delivering defined transcription factors. However, the potential risks associated with gene delivery systems have limited their clinical use. We propose it would be safer to induce neural progenitor-like cells from human adult fibroblasts via a direct non-genetic alternative approach. METHODOLOGY/PRINCIPAL FINDINGS Here, we have reported that seven rounds of TAT-SOX2 protein transduction in a defined chemical cocktail under a 3D sphere culture gradually morphed fibroblasts into neuroepithelial-like colonies. We were able to expand these cells for up to 20 passages. These cells could give rise to cells that expressed neurons and glia cell markers both in vitro and in vivo. CONCLUSIONS/SIGNIFICANCE These results show that our approach is beneficial for the genetic material-free generation of iNPCs from human fibroblasts where small chemical molecules can provide a valuable, viable strategy to boost and improve induction in a 3D sphere culture.
Collapse
Affiliation(s)
- Fahimeh Mirakhori
- School of Biology, College of Science, University of Tehran, Tehran, Iran
- Department of Stem cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Bahman Zeynali
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Hassan Rassouli
- Department of Stem cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ebrahim Shahbazi
- Department of Stem cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shiva Hashemizadeh
- Department of Stem cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sahar Kiani
- Department of Stem cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ghasem Hosseini Salekdeh
- Department of Molecular Systems Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| |
Collapse
|
23
|
Mirakhori F, Zeynali B, Kiani S, Baharvand H. Brief azacytidine step allows the conversion of suspension human fibroblasts into neural progenitor-like cells. CELL JOURNAL 2015; 17:153-8. [PMID: 25870845 PMCID: PMC4393664 DOI: 10.22074/cellj.2015.522] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Accepted: 03/10/2014] [Indexed: 12/29/2022]
Abstract
In recent years transdifferentiation technology has enabled direct conversion of human
fibroblasts to become a valuable, abundant and accessible cell source for patient-specific
induced cell generation in biomedical research. The majority of transdifferentiation approaches rely upon viral gene delivery which due to random integration with the host
genome can cause genome instability and tumorigenesis upon transplantation. Here, we
provide a simple way to induce neural progenitor-like cells from human fibroblasts without genetic manipulation by changing physicochemical culture properties from monolayer
culture into a suspension in the presence of a chemical DNA methyltransferase inhibitor
agent, Azacytidine. We have demonstrated the expression of neural progenitor-like markers, morphology and the ability to spontaneously differentiate into neural-like cells. This
approach is simple, inexpensive, lacks genetic manipulation and could be a foundation for
future chemical neural transdifferentiation and a safe induction of neural progenitor cells
from human fibroblasts for clinical applications.
Collapse
Affiliation(s)
- Fahimeh Mirakhori
- School of Biology, College of Science, University of Tehran, Tehran, Iran ; Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Bahman Zeynali
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Sahar Kiani
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran ; Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| |
Collapse
|
24
|
Dominguez PM, Shaknovich R. Epigenetic function of activation-induced cytidine deaminase and its link to lymphomagenesis. Front Immunol 2014; 5:642. [PMID: 25566255 PMCID: PMC4270259 DOI: 10.3389/fimmu.2014.00642] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 12/02/2014] [Indexed: 01/16/2023] Open
Abstract
Activation-induced cytidine deaminase (AID) is essential for somatic hypermutation and class switch recombination of immunoglobulin (Ig) genes during B cell maturation and immune response. Expression of AID is tightly regulated due to its mutagenic and recombinogenic potential, which is known to target not only Ig genes, but also non-Ig genes, contributing to lymphomagenesis. In recent years, a new epigenetic function of AID and its link to DNA demethylation came to light in several developmental systems. In this review, we summarize existing evidence linking deamination of unmodified and modified cytidine by AID to base-excision repair and mismatch repair machinery resulting in passive or active removal of DNA methylation mark, with the focus on B cell biology. We also discuss potential contribution of AID-dependent DNA hypomethylation to lymphomagenesis.
Collapse
Affiliation(s)
- Pilar M Dominguez
- Division of Hematology and Oncology, Weill Cornell Medical College , New York, NY , USA
| | - Rita Shaknovich
- Division of Hematology and Oncology, Weill Cornell Medical College , New York, NY , USA ; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College , New York, NY , USA
| |
Collapse
|
25
|
Li X, Pei D, Zheng H. Transitions between epithelial and mesenchymal states during cell fate conversions. Protein Cell 2014; 5:580-91. [PMID: 24805308 PMCID: PMC4130923 DOI: 10.1007/s13238-014-0064-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 03/23/2014] [Indexed: 12/15/2022] Open
Abstract
Cell fate conversion is considered as the changing of one type of cells to another type including somatic cell reprogramming (de-differentiation), differentiation, and trans-differentiation. Epithelial and mesenchymal cells are two major types of cells and the transitions between these two cell states as epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) have been observed during multiple cell fate conversions including embryonic development, tumor progression and somatic cell reprogramming. In addition, MET and sequential EMT-MET during the generation of induced pluripotent stem cells (iPSC) from fibroblasts have been reported recently. Such observation is consistent with multiple rounds of sequential EMT-MET during embryonic development which could be considered as a reversed process of reprogramming at least partially. Therefore in current review, we briefly discussed the potential roles played by EMT, MET, or even sequential EMT-MET during different kinds of cell fate conversions. We also provided some preliminary hypotheses on the mechanisms that connect cell state transitions and cell fate conversions based on results collected from cell cycle, epigenetic regulation, and stemness acquisition.
Collapse
Affiliation(s)
- Xiang Li
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | | | | |
Collapse
|
26
|
Zaniboni A, Bernardini C, Alessandri M, Mangano C, Zannoni A, Bianchi F, Sarli G, Calzà L, Bacci ML, Forni M. Cells derived from porcine aorta tunica media show mesenchymal stromal-like cell properties in in vitro culture. Am J Physiol Cell Physiol 2013; 306:C322-33. [PMID: 24304832 DOI: 10.1152/ajpcell.00112.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Several studies have already described the presence of specialized niches of precursor cells in vasculature wall, and it has been shown that these populations share several features with mesenchymal stromal cells (MSCs). Considering the relevance of MSCs in the cardiovascular physiopathology and regenerative medicine, and the usefulness of the pig animal model in this field, we reported a new method for MSC-like cell isolation from pig aorta. Filling the vessel with a collagenase solution for 40 min, all endothelial cells were detached and discarded and then collagenase treatment was repeated for 4 h to digest approximately one-third of the tunica media. The ability of our method to select a population of MSC-like cells from tunica media could be ascribed in part to the elimination of contaminant cells from the intimal layer and in part to the overnight culture in the high antibiotic/antimycotic condition and to the starvation step. Aortic-derived cells show an elongated, spindle shape, fibroblast-like morphology, as reported for MSCs, stain positively for CD44, CD56, CD90, and CD105; stain negatively for CD34 and CD45; and express CD73 mRNA. Moreover, these cells show the classical mesenchymal trilineage differentiation potential. Under our in vitro culture conditions, aortic-derived cells share some phenotypical features with pericytes and are able to take part in the formation of network-like structures if cocultured with human umbilical vein endothelial cells. In conclusion, our work reports a simple and highly suitable method for obtaining large numbers of precursor MSC-like cells derived from the porcine aortic wall.
Collapse
Affiliation(s)
- Andrea Zaniboni
- Department of Veterinary Medical Sciences-DIMEVET, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|