1
|
Xu Z, Song R, Chen Z, Sun Y, Xia Y, Miao H, Wang W, Zhang Y, Jiang X, Chen G. Hydrogen generators-protected mesenchymal stem cells reverse articular redox imbalance-induced immune dysfunction for osteoarthritis treatment. Biomaterials 2025; 320:123239. [PMID: 40054376 DOI: 10.1016/j.biomaterials.2025.123239] [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: 10/26/2024] [Revised: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 04/06/2025]
Abstract
Stem cell therapy has revolutionized the management of osteoarthritis (OA), but the articular dysregulated redox status diminishes cell engraftment efficiency and disrupts immune homeostasis, therefore compromising the overall therapeutic efficacy. Here, we present hydrogen (H2) generators-backpacked mesenchymal stem cells (MSCs) which preserve the biological functions and survival of transplanted cells and reverse articular immune dysfunction, mitigating OA. Specifically, post systemic transplantation, H2 generators-laden MSCs home to OA joints, and upon stimulation in acidic OA environment, H2 produced from the generators remodels articular redox balance, thereby relieving the loss of mitochondrial membrane potential, decreasing cell apoptosis rate, and maintaining pluripotent and paracrine functions of MSCs. Furthermore, the reactive oxygen species scavenging by H2 in combination with paracrine effects of the MSCs promote macrophage polarization towards the anti-inflammatory M2 phenotype, which contributes to reversing synovial immune disorder. In severe OA model, the backpacked MSCs reduce osteoarthritic degeneration, osteophyte formation and joint inflammation, and promote cartilage regeneration. In sum, our work demonstrates that arming with H2 generators effectively boosts the therapeutic efficacy of MSCs, which hold great potential for alleviating redox imbalance-related tissue lesions, including but not limited to OA.
Collapse
Affiliation(s)
- Zhou Xu
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, China; Shandong Provincial Key Medical and Health Laboratory of Neuro-oncology of Innovative Integrated Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Qingdao, 266024, China; Northern Jiangsu People's Hospital, Yangzhou, 225001, China; Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Ruilong Song
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Zhiling Chen
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, China
| | - Yu Sun
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
| | - Yinhe Xia
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Haixiang Miao
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
| | - Weijie Wang
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, China
| | - Yuankai Zhang
- Department of Orthopaedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Xinyi Jiang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Gang Chen
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, China; Shandong Provincial Key Medical and Health Laboratory of Neuro-oncology of Innovative Integrated Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Qingdao, 266024, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
2
|
Le N, Awad S, Palazzo I, Hoang T, Blackshaw S. Viral-mediated Pou5f1 (Oct4) overexpression and inhibition of Notch signaling synergistically induce neurogenic competence in mammalian Müller glia. eLife 2025; 14:RP106450. [PMID: 40388211 PMCID: PMC12088672 DOI: 10.7554/elife.106450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025] Open
Abstract
Retinal Müller glia in cold-blooded vertebrates can reprogram into neurogenic progenitors to replace neurons lost to injury, but mammals lack this ability. While recent studies have shown that transgenic overexpression of neurogenic bHLH factors and glial-specific disruption of NFI family transcription factors and Notch signaling induce neurogenic competence in mammalian Müller glia, induction of neurogenesis in wildtype glia has thus far proven elusive. Here, we report that viral-mediated overexpression of the pluripotency factor Pou5f1 (Oct4) induces transdifferentiation of mouse Müller glia into bipolar neurons, and synergistically stimulates glial-derived neurogenesis in parallel with Notch loss of function. Single-cell multiomic analysis shows that Pou5f1 overexpression leads to widespread changes in gene expression and chromatin accessibility, inducing activity of both the neurogenic transcription factor Rfx4 and the Yamanaka factors Sox2 and Klf4. This study demonstrates that viral-mediated overexpression of Pou5f1 induces neurogenic competence in adult mouse Müller glia, identifying mechanisms that could be used in cell-based therapies for treating retinal dystrophies.
Collapse
Affiliation(s)
- Nguyet Le
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Sherine Awad
- Department of Ophthalmology and Visual Sciences, University of Michigan School of MedicineAnn ArborUnited States
- Department of Cell and Developmental Biology, University of Michigan School of MedicineAnn ArborUnited States
- Michigan Neuroscience Institute, University of Michigan School of MedicineAnn ArborUnited States
| | - Isabella Palazzo
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Thanh Hoang
- Department of Ophthalmology and Visual Sciences, University of Michigan School of MedicineAnn ArborUnited States
- Department of Cell and Developmental Biology, University of Michigan School of MedicineAnn ArborUnited States
- Michigan Neuroscience Institute, University of Michigan School of MedicineAnn ArborUnited States
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Ophthalmology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Institute for Cell Engineering, Johns Hopkins University School of MedicineBaltimoreUnited States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
3
|
Matiukhova M, Ryapolova A, Andriianov V, Reshetnikov V, Zhuravleva S, Ivanov R, Karabelsky A, Minskaia E. A comprehensive analysis of induced pluripotent stem cell (iPSC) production and applications. Front Cell Dev Biol 2025; 13:1593207. [PMID: 40406420 PMCID: PMC12095295 DOI: 10.3389/fcell.2025.1593207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
The ability to reprogram mature, differentiated cells into induced pluripotent stem cells (iPSCs) using exogenous pluripotency factors opened up unprecedented opportunities for their application in biomedicine. iPSCs are already successfully used in cell and regenerative therapy, as various drug discovery platforms and for in vitro disease modeling. However, even though already 20 years have passed since their discovery, the production of iPSC-based therapies is still associated with a number of hurdles due to low reprogramming efficiency, the complexity of accurate characterization of the resulting colonies, and the concerns associated with the safety of this approach. However, significant progress in many areas of molecular biology facilitated the production, characterization, and thorough assessment of the safety profile of iPSCs. The number of iPSC-based studies has been steadily increasing in recent years, leading to the accumulation of significant knowledge in this area. In this review, we aimed to provide a comprehensive analysis of methods used for reprogramming and subsequent characterization of iPSCs, discussed barriers towards achieving these goals, and various approaches to improve the efficiency of reprogramming of different cell populations. In addition, we focused on the analysis of iPSC application in preclinical and clinical studies. The accumulated breadth of data helps to draw conclusions about the future of this technology in biomedicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ekaterina Minskaia
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
4
|
Esfandiarinezhad F, Zhan X, Tan SL, Li J, Tsang BK. A primary insight into gut microbiome, MicroRNA and stemness, in a PCOS rat model. J Ovarian Res 2025; 18:66. [PMID: 40170042 PMCID: PMC11959994 DOI: 10.1186/s13048-025-01648-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/15/2025] [Indexed: 04/03/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine disorder associated with reproductive and metabolic dysfunctions, including gut microbiome dysbiosis. This study aimed to examine the alterations in stemness in ovarian surface epithelium (OSE), gut microbiome microRNA expression in granulosa cells and plasma in a dihydrotestosterone (DHT)-induced rat model of PCOS. Female rats were administered DHT to induce PCOS, and the expression of stem cell markers in OSE was assessed to evaluate the impact on stemness. Alterations in the gut microbiome composition were assessed using 16S rRNA gene Long-Read sequencing and changes in the microRNA profile of granulosa cells and plasma were analyzed using qPCR. Our results demonstrated alterations in stemness markers and, a significant alteration in gut microbiome composition in DHT-induced rats compared to controls, characterized by shifts in the relative abundance of specific bacterial taxa, particularly Akkermansia muciniphila. Elevated levels of miR-574 and miR-378 were observed in plasma, whereas miR-21 and miR-574 showed increased expression in ovarian granulosa cells. Concurrently, increased expression of stem cell markers was observed in OSE, suggesting an enhancement of stemness in response to PCOS-like conditions. These findings imply a potential link between gut microbiome dysbiosis and increased ovarian stemness in PCOS, suggesting that the gut microbiome may contribute to ovarian dysfunction through modulation of stem cell activity. Understanding this interaction could provide novel insights into therapeutic targets in restoring ovarian function in PCOS patients.
Collapse
Affiliation(s)
- Fereshteh Esfandiarinezhad
- Inflammation and Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Departments of Obstetrics and Gynecology & Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- OriginElle Fertility Clinic and Women's Health Center, Ottawa, Canada
| | - Xiaoshu Zhan
- Department of Animal Biosciences, University of Guelph, Guelph, N1G 2W1, Canada
- School of Animal Science and Technology, Foshan University, Foshan, Guangdong, China
| | - Seang Lin Tan
- OriginElle Fertility Clinic and Women's Health Center, Ottawa, Canada
- Department of Obstetrics & Gynecology, McGill University, Montreal, Canada
| | - Julang Li
- Department of Animal Biosciences, University of Guelph, Guelph, N1G 2W1, Canada.
| | - Benjamin K Tsang
- Inflammation and Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
- Departments of Obstetrics and Gynecology & Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
5
|
Le N, Awad S, Palazzo I, Hoang T, Blackshaw S. Viral-mediated Oct4 overexpression and inhibition of Notch signaling synergistically induce neurogenic competence in mammalian Muller glia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.18.613666. [PMID: 39345433 PMCID: PMC11429848 DOI: 10.1101/2024.09.18.613666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Retinal Muller glia in cold-blooded vertebrates can reprogram into neurogenic progenitors to replace neurons lost to injury, but mammals lack this ability. While recent studies have shown that transgenic overexpression of neurogenic bHLH factors and glial-specific disruption of NFI family transcription factors and Notch signaling induce neurogenic competence in mammalian Muller glia, induction of neurogenesis in wild-type glia has thus far proven elusive. Here, we report that viral-mediated overexpression of the pluripotency factor Oct4 (Pou5f1) induces transdifferentiation of mouse Muller glia into bipolar neurons, and synergistically stimulates glial-derived neurogenesis in parallel with Notch loss of function. Single cell multiomic analysis shows that Oct4 overexpression leads to widespread changes in gene expression and chromatin accessibility, inducing activity of both the neurogenic transcription factor Rfx4 and the Yamanaka factors Sox2 and Klf4. This study demonstrates that viral-mediated overexpression of Oct4 induces neurogenic competence in retinal Muller glia, identifying mechanisms that could be used in cell-based therapies for treating retinal dystrophies.
Collapse
|
6
|
Narasimha RB, Shreya S, Jayabal VA, Yadav V, Rath PK, Mishra BP, Kancharla S, Kolli P, Mandadapu G, Kumar S, Mohanty AK, Jena MK. Stem Cell Therapy for Diseases of Livestock Animals: An In-Depth Review. Vet Sci 2025; 12:67. [PMID: 39852942 PMCID: PMC11768649 DOI: 10.3390/vetsci12010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 01/26/2025] Open
Abstract
Stem cells are unique, undifferentiated cells that have the ability to both replicate themselves and develop into specialized cell types. This dual capability makes them valuable in the development of regenerative medicine. Current development in stem cell research has widened their application in cell therapy, drug discovery, reproductive cloning in animals, and cell models for various diseases. Although there are substantial studies revealing the treatment of human degenerative diseases using stem cells, this is yet to be explored in livestock animals. Many diseases in livestock species such as mastitis, laminitis, neuromuscular disorders, autoimmune diseases, and some debilitating diseases are not covered completely by the existing drugs and treatment can be improved by using different types of stem cells like embryonic stem cells, adult stem cells, and induced pluripotent stem cells. This review mainly focuses on the use of stem cells for disease treatment in livestock animals. In addition to the diseases mentioned, the potential of stem cells can be helpful in wound healing, skin disease therapy, and treatment of some genetic disorders. This article explores the potential of stem cells from various sources in the therapy of livestock diseases and also their role in the conservation of endangered species as well as disease model preparation. Moreover, the future perspectives and challenges associated with the application of stem cells in livestock are discussed. Overall, the transformative impact of stem cell research on the livestock sector is comprehensively studied which will help researchers to design future research work on stem cells related to livestock diseases.
Collapse
Affiliation(s)
- Raghavendra B. Narasimha
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India; (R.B.N.); (S.S.)
| | - Singireddy Shreya
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India; (R.B.N.); (S.S.)
| | - Vijay Anand Jayabal
- Department of Animal Biotechnology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai 600051, Tamil Nadu, India;
| | - Vikas Yadav
- Department of Clinical Sciences, Clinical Research Centre, Skåne University Hospital, Lund University, SE 20213 Malmö, Sweden
| | - Prasana Kumar Rath
- College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar 751003, Odisha, India; (P.K.R.); (B.P.M.)
| | - Bidyut Prava Mishra
- College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar 751003, Odisha, India; (P.K.R.); (B.P.M.)
| | - Sudhakar Kancharla
- Devansh Lab Werks, 234 Aquarius Drive, Homewood, AL 35209, USA; (S.K.); (G.M.)
| | - Prachetha Kolli
- Microgen Health Inc., 14225 Sullyfield Cir Suite E, Chantilly, VA 20151, USA;
| | - Gowtham Mandadapu
- Devansh Lab Werks, 234 Aquarius Drive, Homewood, AL 35209, USA; (S.K.); (G.M.)
| | - Sudarshan Kumar
- Cell, Molecular and Proteomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (ICAR-NDRI), Karnal 132001, Haryana, India;
| | - Ashok Kumar Mohanty
- ICAR-Central Institute for Research on Cattle (ICAR-CIRC), Meerut 250001, Uttar Pradesh, India;
| | - Manoj Kumar Jena
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India; (R.B.N.); (S.S.)
| |
Collapse
|
7
|
He XD, Taylor LF, Miao X, Shi Y, Lin X, Yang Z, Liu X, Miao YL, Alfandari D, Cui W, Tremblay KD, Mager J. Overlapping peri-implantation phenotypes of ZNHIT1 and ZNHIT2 despite distinct functions during early mouse development†. Biol Reprod 2024; 111:1017-1029. [PMID: 39194072 PMCID: PMC11565232 DOI: 10.1093/biolre/ioae128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/10/2024] [Accepted: 08/27/2024] [Indexed: 08/29/2024] Open
Abstract
Mammalian preimplantation development culminates in the formation of a blastocyst that undergoes extensive gene expression regulation to successfully implant into the maternal endometrium. Zinc-finger HIT domain-containing (ZNHIT) 1 and 2 are members of a highly conserved family, yet they have been identified as subunits of distinct complexes. Here, we report that knockout of either Znhit1 or Znhit2 results in embryonic lethality during peri-implantation stages. Znhit1 and Znhit2 mutant embryos have overlapping phenotypes, including reduced proportion of SOX2-positive inner cell mass cells, a lack of Fgf4 expression, and aberrant expression of NANOG and SOX17. Furthermore, we find that the similar phenotypes are caused by distinct mechanisms. Specifically, embryos lacking ZNHIT1 likely fail to incorporate sufficient H2A.Z at the promoter region of Fgf4 and other genes involved in cell projection organization resulting in impaired invasion of trophoblast cells during implantation. In contrast, Znhit2 mutant embryos display a complete lack of nuclear EFTUD2, a key component of U5 spliceosome, indicating a global splicing deficiency. Our findings unveil the indispensable yet distinct roles of ZNHIT1 and ZNHIT2 in early mammalian embryonic development.
Collapse
Affiliation(s)
- Xinjian Doris He
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Louis F Taylor
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Xiaosu Miao
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Yingchao Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School of Nanjing University, Nanjing, China
| | - Xinhua Lin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School of Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Xin Liu
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yi-Liang Miao
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Dominique Alfandari
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Wei Cui
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Kimberly D Tremblay
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Jesse Mager
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
8
|
Rathore R, Kaul S, Sharma JB, Mathur SR. Exploring the role of SOX 2 and OCT 4 in the pathogenesis of gliomatosis peritonei: the clinicopathological profile of eleven cases. J Turk Ger Gynecol Assoc 2024; 25:66-73. [PMID: 38867687 DOI: 10.4274/jtgga.galenos.2024.2023-9-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
Objective Gliomatosis peritonei (GP) is a rare entity characterized by multiple mature glial tissue implants in association with ovarian teratomas in the peritoneum and omentum. To date, only 100 cases have been published. Not much is known about the origin, clinicopathological profile or prognosis of GP. SOX2 and OCT4 are recently recognized markers of embryonic stem cell differentiation. Here, the role of SOX2 and OCT4 in the pathogenesis of 11 cases of GP are reported and clinicopathological factors are described. Material and Methods This was a retrospective study of six years duration (2017-2022). All the cases of GP were retrieved from archives, the diagnosis was confirmed and clinicopathological factors were noted. Immunohistochemical (IHC) investigation for glial fibrillary acid protein (GFAP) and S100 was noted wherever available. IHC for SOX2 and OCT4 was performed using an avidin-biotin technique. Results There were 11 cases of GP identified. The median age was 29 years and 1/11 cases had nodal gliomatosis as well. There were eight cases of immature teratoma and three cases of mature cystic teratoma. SOX2 was positive in all foci of GP, while OCT4 was negative. These foci were also positive for GFAP and S100. Conclusion A possibility of GP should be considered as a differential, clinically and radiologically, in cases of omental nodularity. Adequate sampling at the time of surgery is essential to rule out metastasis or growing teratoma syndrome. SOX2, a stem cell marker inducing neural differentiation, may play a crucial role in the development of GP in association with other transcription factors.
Collapse
Affiliation(s)
- Ruchi Rathore
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Shreya Kaul
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, New Delhi, India
| | - Jai Bhagwan Sharma
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, New Delhi, India
| | - Sandeep R Mathur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
9
|
Zhang X, Wan J, Huang T, Tang P, Yang L, Bu X, Zhang W, Zhong L. Rapid and accurate identification of stem cell differentiation stages via SERS and convolutional neural networks. BIOMEDICAL OPTICS EXPRESS 2024; 15:2753-2766. [PMID: 38855654 PMCID: PMC11161375 DOI: 10.1364/boe.519093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 06/11/2024]
Abstract
Monitoring the transition of cell states during induced pluripotent stem cell (iPSC) differentiation is crucial for clinical medicine and basic research. However, both identification category and prediction accuracy need further improvement. Here, we propose a method combining surface-enhanced Raman spectroscopy (SERS) with convolutional neural networks (CNN) to precisely identify and distinguish cell states during stem cell differentiation. First, mitochondria-targeted probes were synthesized by combining AuNRs and mitochondrial localization signal (MLS) peptides to obtain effective and stable SERS spectra signals at various stages of cell differentiation. Then, the SERS spectra served as input datasets, and their distinctive features were learned and distinguished by CNN. As a result, rapid and accurate identification of six different cell states, including the embryoid body (EB) stage, was successfully achieved throughout the stem cell differentiation process with an impressive prediction accuracy of 98.5%. Furthermore, the impact of different spectral feature peaks on the identification results was investigated, which provides a valuable reference for selecting appropriate spectral bands to identify cell states. This is also beneficial for shortening the spectral acquisition region to enhance spectral acquisition speed. These results suggest the potential for SERS-CNN models in quality monitoring of stem cells, advancing the practical applications of stem cells.
Collapse
Affiliation(s)
- Xiao Zhang
- Key Laboratory of Photonics Technology for Integrated Sensing and Communication of Ministry of Education, Guangdong University of Technology, Guangzhou 510006, China
| | - Jianhui Wan
- Key Laboratory of Photonics Technology for Integrated Sensing and Communication of Ministry of Education, Guangdong University of Technology, Guangzhou 510006, China
| | - Tao Huang
- Key Laboratory of Photonics Technology for Integrated Sensing and Communication of Ministry of Education, Guangdong University of Technology, Guangzhou 510006, China
| | - Ping Tang
- Key Laboratory of Photonics Technology for Integrated Sensing and Communication of Ministry of Education, Guangdong University of Technology, Guangzhou 510006, China
- School of Physics and Optoelectronic Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Liwei Yang
- Guangdong Provincial Key Laboratory of Nanophotonic Functional Materials and Devices, South China Normal University, Guangzhou 510006, China
| | - Xiaoya Bu
- Guangdong Provincial Key Laboratory of Nanophotonic Functional Materials and Devices, South China Normal University, Guangzhou 510006, China
| | - Weina Zhang
- Key Laboratory of Photonics Technology for Integrated Sensing and Communication of Ministry of Education, Guangdong University of Technology, Guangzhou 510006, China
| | - Liyun Zhong
- Key Laboratory of Photonics Technology for Integrated Sensing and Communication of Ministry of Education, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
10
|
Hopkinson A, Notara M, Cursiefen C, Sidney LE. Increased Anti-Inflammatory Therapeutic Potential and Progenitor Marker Expression of Corneal Mesenchymal Stem Cells Cultured in an Optimized Propagation Medium. Cell Transplant 2024; 33:9636897241241992. [PMID: 38602231 PMCID: PMC11010753 DOI: 10.1177/09636897241241992] [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: 01/03/2024] [Revised: 02/19/2024] [Accepted: 03/05/2024] [Indexed: 04/12/2024] Open
Abstract
There is a huge unmet need for new treatment modalities for ocular surface inflammatory disorders (OSIDs) such as dry eye disease and meibomian gland dysfunction. Mesenchymal stem cell therapies may hold the answer due to their potent immunomodulatory properties, low immunogenicity, and ability to modulate both the innate and adaptive immune response. MSC-like cells that can be isolated from the corneal stroma (C-MSCs) offer a potential new treatment strategy; however, an optimized culture medium needs to be developed to produce the ideal phenotype for use in a cell therapy to treat OSIDs. The effects of in vitro expansion of human C-MSC in a medium of M199 containing fetal bovine serum (FBS) was compared to a stem cell medium (SCM) containing knockout serum replacement (KSR) with basic fibroblast growth factor (bFGF) and human leukemia inhibitory factor (LIF), investigating viability, protein, and gene expression. Isolating populations expressing CD34 or using siRNA knockdown of CD34 were investigated. Finally, the potential of C-MSC as a cell therapy was assessed using co-culture with an in vitro corneal epithelial cell injury model and the angiogenic effects of C-MSC conditioned medium were evaluated with blood and lymph endothelial cells. Both media supported proliferation of C-MSC, with SCM increasing expression of CD34, ABCG2, PAX6, NANOG, REX1, SOX2, and THY1, supported by increased associated protein expression. Isolating cell populations expressing CD34 protein made little difference to gene expression, however, knockdown of the CD34 gene led to decreased expression of progenitor genes. C-MSC increased viability of injured corneal epithelial cells whilst decreasing levels of cytotoxicity and interleukins-6 and -8. No pro-angiogenic effect of C-MSC was seen. Culture medium can significantly influence C-MSC phenotype and culture in SCM produced a cell phenotype more suitable for further consideration as an anti-inflammatory cell therapy. C-MSC show considerable potential for development as therapies for OSIDs, acting through anti-inflammatory action.
Collapse
Affiliation(s)
- Andrew Hopkinson
- Academic Ophthalmology, Mental Health and Clinical Neurosciences, University of Nottingham, Nottingham, UK
| | - Maria Notara
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Koln, Germany
| | - Claus Cursiefen
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Koln, Germany
| | - Laura E. Sidney
- Academic Ophthalmology, Mental Health and Clinical Neurosciences, University of Nottingham, Nottingham, UK
- Regenerating and Modelling Tissues, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
11
|
Bora J, Dey A, Lyngdoh AR, Dhasmana A, Ranjan A, Kishore S, Rustagi S, Tuli HS, Chauhan A, Rath P, Malik S. A critical review on therapeutic approaches of CRISPR-Cas9 in diabetes mellitus. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3459-3481. [PMID: 37522916 DOI: 10.1007/s00210-023-02631-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 07/14/2023] [Indexed: 08/01/2023]
Abstract
Diabetes mellitus (D.M.) is a common metabolic disorder caused mainly by combining two primary factors, which are (1) defects in insulin production by the pancreatic β-cells and (2) responsiveness of insulin-sensitive tissues towards insulin. Despite the rapid advancement in medicine to suppress elevated blood glucose levels (hyperglycemia) and insulin resistance associated with this hazard, a demand has undoubtedly emerged to find more effective and curative dimensions in therapeutic approaches against D.M. The administration of diabetes treatment that emphasizes insulin production and sensitivity may result in unfavorable side effects, reduced adherence, and potential treatment ineffectiveness. Recent progressions in genome editing technologies, for instance, in zinc-finger nucleases, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeat (CRISPR-Cas)-associated nucleases, have greatly influenced the gene editing technology from concepts to clinical practices. Improvements in genome editing technologies have also opened up the possibility to target and modify specific genome sequences in a cell directly. CRISPR/Cas9 has proven effective in utilizing ex vivo gene editing in embryonic stem cells and stem cells derived from patients. This application has facilitated the exploration of pancreatic beta-cell development and function. Furthermore, CRISPR/Cas9 enables the creation of innovative animal models for diabetes and assesses the effectiveness of different therapeutic strategies in treating the condition. We, therefore, present a critical review of the therapeutic approaches of the genome editing tool CRISPR-Cas9 in treating D.M., discussing the challenges and limitations of implementing this technology.
Collapse
Affiliation(s)
- Jutishna Bora
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, 834001, India
| | - Ankita Dey
- Department of Biochemistry, North Eastern Hill University, Shillong, Meghalaya, 793022, India
| | - Antonia R Lyngdoh
- Department of Biochemistry, North Eastern Hill University, Shillong, Meghalaya, 793022, India
| | - Archna Dhasmana
- Himalayan School of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun, Uttarakhand, India
| | - Anuj Ranjan
- Academy of Biology and Biotechnology, Southern Federal University, Stachki 194/1, Rostov-On-Don, 344090, Russia
| | - Shristi Kishore
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, 834001, India
| | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University, 22 Dehradun, Uttarakhand, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana-Ambala, 133207, India
| | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Sector 125, Noida, Uttar Pradesh, India
| | - Prangya Rath
- Amity Institute of Environmental Sciences, Amity University, Noida, Uttar Pradesh, 201303, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, 834001, India.
- School of Applied and Life Sciences, Uttaranchal University, 22 Dehradun, Uttarakhand, India.
- Guru Nanak College of Pharmaceutical Sciences, Dehradun, Uttarakhand, India.
| |
Collapse
|
12
|
Sheveleva O, Protasova E, Nenasheva T, Butorina N, Melnikova V, Gerasimova T, Sakovnich O, Kurinov A, Grigor’eva E, Medvedev S, Lyadova I. A Model of iPSC-Derived Macrophages with TNFAIP3 Overexpression Reveals the Peculiarities of TNFAIP3 Protein Expression and Function in Human Macrophages. Int J Mol Sci 2023; 24:12868. [PMID: 37629049 PMCID: PMC10454046 DOI: 10.3390/ijms241612868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/29/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
Macrophages play a crucial role in the development and control of inflammation. Understanding the mechanisms balancing macrophage inflammatory activity is important to develop new strategies for treating inflammation-related diseases. TNF-α-induced protein 3 (TNFAIP3, A20) is a negative regulator of intracellular inflammatory cascades; its deficiency induces hyper-inflammatory reactions. Whether A20 overexpression can dampen macrophage inflammatory response remains unclear. Here, we generated human-induced pluripotent stem cells with tetracycline-inducible A20 expression and differentiated them into macrophages (A20-iMacs). A20-iMacs displayed morphology, phenotype, and phagocytic activity typical of macrophages, and they displayed upregulated A20 expression in response to doxycycline. A20 overexpression dampened the A20-iMac response to TNF-α, as shown by a decreased expression of IL1B and IL6 mRNA. A dynamic analysis of A20 expression following the generation of A20-iMacs and control iMacs showed that the expression declined in iMacs and that iMacs expressed a lower molecular weight form of the A20 protein (~70 kDa) compared with less differentiated cells (~90 kDa). A low-level expression of A20 and the predominance of a low-molecular-weight A20 form were also characteristic of monocyte-derived macrophages. The study for the first time developed a model for generating macrophages with an inducible expression of a target gene and identified the peculiarities of A20 expression in macrophages that likely underlie macrophage preparedness for inflammatory reactivity. It also suggested the possibility of mitigating inflammatory macrophage responses via A20 overexpression.
Collapse
Affiliation(s)
- Olga Sheveleva
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| | - Elena Protasova
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| | - Tatiana Nenasheva
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| | - Nina Butorina
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| | - Victoria Melnikova
- Laboratory of Comparative Developmental Physiology, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia;
| | - Tatiana Gerasimova
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| | - Olga Sakovnich
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| | - Alexander Kurinov
- Laboratory of Regeneration Problems, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia;
| | - Elena Grigor’eva
- Laboratory of Developmental Epigenetics, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Lavrentyev Ave., 10, 630090 Novosibirsk, Russia; (E.G.); (S.M.)
| | - Sergey Medvedev
- Laboratory of Developmental Epigenetics, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Lavrentyev Ave., 10, 630090 Novosibirsk, Russia; (E.G.); (S.M.)
| | - Irina Lyadova
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| |
Collapse
|
13
|
Oses C, Francia MG, Verneri P, Vazquez Echegaray C, Guberman AS, Levi V. The dynamical organization of the core pluripotency transcription factors responds to differentiation cues in early S-phase. Front Cell Dev Biol 2023; 11:1125015. [PMID: 37215075 PMCID: PMC10192714 DOI: 10.3389/fcell.2023.1125015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
DNA replication in stem cells is a major challenge for pluripotency preservation and cell fate decisions. This process involves massive changes in the chromatin architecture and the reorganization of many transcription-related molecules in different spatial and temporal scales. Pluripotency is controlled by the master transcription factors (TFs) OCT4, SOX2 and NANOG that partition into condensates in the nucleus of embryonic stem cells. These condensates are proposed to play relevant roles in the regulation of gene expression and the maintenance of pluripotency. Here, we asked whether the dynamical distribution of the pluripotency TFs changes during the cell cycle, particularly during DNA replication. Since the S phase is considered to be a window of opportunity for cell fate decisions, we explored if differentiation cues in G1 phase trigger changes in the distribution of these TFs during the subsequent S phase. Our results show a spatial redistribution of TFs condensates during DNA replication which was not directly related to chromatin compaction. Additionally, fluorescence fluctuation spectroscopy revealed TF-specific, subtle changes in the landscape of TF-chromatin interactions, consistent with their particularities as key players of the pluripotency network. Moreover, we found that differentiation stimuli in the preceding G1 phase triggered a relatively fast and massive reorganization of pluripotency TFs in early-S phase. Particularly, OCT4 and SOX2 condensates dissolved whereas the lifetimes of TF-chromatin interactions increased suggesting that the reorganization of condensates is accompanied with a change in the landscape of TF-chromatin interactions. Notably, NANOG showed impaired interactions with chromatin in stimulated early-S cells in line with its role as naïve pluripotency TF. Together, these findings provide new insights into the regulation of the core pluripotency TFs during DNA replication of embryonic stem cells and highlight their different roles at early differentiation stages.
Collapse
Affiliation(s)
- Camila Oses
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marcos Gabriel Francia
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Paula Verneri
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Camila Vazquez Echegaray
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alejandra Sonia Guberman
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Valeria Levi
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
14
|
Molecular Biomarkers of Malignant Transformation in Head and Neck Dysplasia. Cancers (Basel) 2022; 14:cancers14225581. [PMID: 36428690 PMCID: PMC9688631 DOI: 10.3390/cancers14225581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) and its treatments are associated with substantial morbidity, often resulting in cosmetic deformity and loss of physiologic functions including speech and swallowing. Despite advancements in treatment, 5-year survival rates for mucosal malignancies remain below 70%. Effective prevention of HNSCC demands an understanding of the molecular pathways of carcinogenesis. Specifically, defining features of pre-cancerous dysplastic lesions that indicate a better or worse prognosis is necessary to help identify patients who are likely to develop a carcinoma and allow a more aggressive approach to management. There remains a need for identification of biomarkers that can provide both early prognostic and predictive value in clinical decision-making by serving as both therapeutic targets as well as predictors of therapy response. Here, we comprehensively review the most frequently altered molecular biomarkers of malignant transformation in head and neck dysplasia. These markers are involved in a wide range of cellular processes in head and neck carcinogenesis, including extracellular matrix degradation, cell motility and invasion, cell-cell adhesion, solute transport, immortalization, metabolism, the cell cycle and apoptosis, transcription, and cell signaling.
Collapse
|
15
|
Lufkin L, Samanta A, Baker D, Lufkin S, Schulze J, Ellis B, Rose J, Lufkin T, Kraus P. Glis1 and oxaloacetate in nucleus pulposus stromal cell somatic reprogramming and survival. Front Mol Biosci 2022; 9:1009402. [PMID: 36406265 PMCID: PMC9671658 DOI: 10.3389/fmolb.2022.1009402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Regenerative medicine aims to repair degenerate tissue through cell refurbishment with minimally invasive procedures. Adipose tissue (FAT)-derived stem or stromal cells are a convenient autologous choice for many regenerative cell therapy approaches. The intervertebral disc (IVD) is a suitable target. Comprised of an inner nucleus pulposus (NP) and an outer annulus fibrosus (AF), the degeneration of the IVD through trauma or aging presents a substantial socio-economic burden worldwide. The avascular nature of the mature NP forces cells to reside in a unique environment with increased lactate levels, conditions that pose a challenge to cell-based therapies. We assessed adipose and IVD tissue-derived stromal cells through in vitro transcriptome analysis in 2D and 3D culture and suggested that the transcription factor Glis1 and metabolite oxaloacetic acid (OAA) could provide NP cells with survival tools for the harsh niche conditions in the IVD.
Collapse
Affiliation(s)
- Leon Lufkin
- Department of Statistics and Data Science, Yale University, New Haven, CT, United States,The Clarkson School, Clarkson University, Potsdam, NY, United States
| | - Ankita Samanta
- Department of Biology, Clarkson University, Potsdam, NY, United States
| | - DeVaun Baker
- The Clarkson School, Clarkson University, Potsdam, NY, United States,Department of Biology, Clarkson University, Potsdam, NY, United States
| | - Sina Lufkin
- The Clarkson School, Clarkson University, Potsdam, NY, United States,Department of Biology, Clarkson University, Potsdam, NY, United States
| | | | - Benjamin Ellis
- Department of Biology, Clarkson University, Potsdam, NY, United States
| | - Jillian Rose
- Department of Biology, Clarkson University, Potsdam, NY, United States
| | - Thomas Lufkin
- Department of Biology, Clarkson University, Potsdam, NY, United States
| | - Petra Kraus
- Department of Biology, Clarkson University, Potsdam, NY, United States,*Correspondence: Petra Kraus,
| |
Collapse
|
16
|
Lim C, Roh YH, Yoo SJ, Jeong DK, Nam KW. Identification of Stem Cell Related Gene Expression from the Osteosarcoma Cell Core Side. J Cancer Prev 2022; 27:122-128. [PMID: 35864855 PMCID: PMC9271406 DOI: 10.15430/jcp.2022.27.2.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 11/04/2022] Open
Abstract
Osteosarcoma is the most frequent primary malignant bone tumor with higher incidences in children and adolescents. Despite clinical evolutions, patients with osteosacoma have had a poor prognosis. There has been increasing evidence that cancer is a stem cell disease. This study sought to isolate and characterize cancer stem cells from human osteosarcoma with relevant literature reviews. Here we show that the emerging evidence suggests osteosarcoma should be regarded as a differentiation disease such as stem cell disease. Two human osteosarcoma cell lines were cultured in non-adherent culture conditions as sarcospheres. Sarcospheres were observed using histomorphology and alkaline phosphatase (ALP) staining. Expression of the embryonic stem cell marker was analyzed with use of reverse transcriptase-PCR. Sarcospheres could be reproduced consistently throughout multiple passages and produced adherent osteosarcoma cell cultures. Expression of stem cell-associated genes such as those encoding Nanog, octamer-binding transcription factor 3/4, sex determining region Y box 2 , c-Myc and ALP indicated pluripotent stem-like cells. These results support the extension of the cancer stem cell theory to include osteosarcoma. Understanding the cancer stem cell derived from human osteosarcoma could lead to the evolution of diagnosis and treatment for osteosarcoma patients.
Collapse
Affiliation(s)
- Chaemoon Lim
- Department of Orthopaedic Surgery, Jeju National University Hospital, Jeju, Korea
| | - Young Ho Roh
- Department of Orthopaedic Surgery, Jeju National University Hospital, Jeju, Korea
| | - Seung Jin Yoo
- Department of Orthopaedic Surgery, Jeju National University Hospital, Jeju, Korea
| | - Dong Kee Jeong
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Korea
| | - Kwang Woo Nam
- Department of Orthopaedic Surgery, Uijeongbu Eulji Medical Center, Eulji University, Uijeongbu, Korea
| |
Collapse
|
17
|
Pandey SK, Machlof-Cohen R, Santhanam M, Shteinfer-Kuzmine A, Shoshan-Barmatz V. Silencing VDAC1 to Treat Mesothelioma Cancer: Tumor Reprograming and Altering Tumor Hallmarks. Biomolecules 2022; 12:biom12070895. [PMID: 35883451 PMCID: PMC9312978 DOI: 10.3390/biom12070895] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 12/10/2022] Open
Abstract
Mesothelioma, an aggressive cancer with a poor prognosis, is linked to asbestos exposure. However, carbon nanotubes found in materials we are exposed to daily can cause mesothelioma cancer. Cancer cells reprogram their metabolism to support increased biosynthetic and energy demands required for their growth and motility. Here, we examined the effects of silencing the expression of the voltage-dependent anion channel 1 (VDAC1), controlling the metabolic and energetic crosstalk between mitochondria and the rest of the cell. We demonstrate that VDAC1 is overexpressed in mesothelioma patients; its levels increase with disease stage and are associated with low survival rates. Silencing VDAC1 expression using a specific siRNA identifying both mouse and human VDAC1 (si-m/hVDAC1-B) inhibits cell proliferation of mesothelioma cancer cells. Treatment of xenografts of human-derived H226 cells or mouse-derived AB1 cells with si-m/hVDAC1-B inhibited tumor growth and caused metabolism reprogramming, as reflected in the decreased expression of metabolism-related proteins, including glycolytic and tricarboxylic acid (-)cycle enzymes and the ATP-synthesizing enzyme. In addition, tumors depleted of VDAC1 showed altered microenvironments and inflammation, both associated with cancer progression. Finally, tumor VDAC1 silencing also eliminated cancer stem cells and induced cell differentiation to normal-like cells. The results show that silencing VDAC1 expression leads to reprogrammed metabolism and to multiple effects from tumor growth inhibition to modulation of the tumor microenvironment and inflammation, inducing differentiation of malignant cells. Thus, silencing VDAC1 is a potential therapeutic approach to treating mesothelioma.
Collapse
Affiliation(s)
- Swaroop Kumar Pandey
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (S.K.P.); (R.M.-C.); (M.S.)
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| | - Renen Machlof-Cohen
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (S.K.P.); (R.M.-C.); (M.S.)
| | - Manikandan Santhanam
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (S.K.P.); (R.M.-C.); (M.S.)
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| | - Anna Shteinfer-Kuzmine
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| | - Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (S.K.P.); (R.M.-C.); (M.S.)
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
- Correspondence: ; Tel.: +972-528795939; Fax: +972-86479207
| |
Collapse
|
18
|
Mamun MMA, Khan MR, Zhu Y, Zhang Y, Zhou S, Xu R, Bukhari I, Thorne RF, Li J, Zhang XD, Liu G, Chen S, Wu M, Song X. Stub1 maintains proteostasis of master transcription factors in embryonic stem cells. Cell Rep 2022; 39:110919. [PMID: 35675767 DOI: 10.1016/j.celrep.2022.110919] [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: 10/02/2021] [Revised: 04/01/2022] [Accepted: 05/10/2022] [Indexed: 12/01/2022] Open
Abstract
The pluripotency and differentiation states of embryonic stem cells (ESCs) are regulated by a set of core transcription factors, primarily Sox2, Oct4, and Nanog. Although their transcriptional regulation has been studied extensively, the contribution of posttranslational modifications in Sox2, Oct4, and Nanog are poorly understood. Here, using a CRISPR-Cas9 knockout library screen in murine ESCs, we identify the E3 ubiquitin ligase Stub1 as a negative regulator of pluripotency. Manipulation of Stub1 expression in murine ESCs shows that ectopic Stub1 expression significantly reduces the protein half-life of Sox2, Oct4, and Nanog. Mechanistic investigations reveal Stub1 catalyzes the polyubiquitination and 26S proteasomal degradation of Sox2 and Nanog through K48-linked ubiquitin chains and Oct4 via K63 linkage. Stub1 deficiency positively enhances somatic cell reprogramming and delays differentiation, whereas its enforced expression triggers ESC differentiation. The discovery of Stub1 as an integral pluripotency regulator strengthens our understanding of ESC regulation beyond conventional transcriptional control mechanisms.
Collapse
Affiliation(s)
- Md Mahfuz Al Mamun
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China
| | - Muhammad Riaz Khan
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China; Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada; Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1E 4K8 Canada
| | - Yifu Zhu
- CAS Key Laboratory of Innate Immunity and Chronic Disease, CAS Centre for Excellence in Molecular Cell Science, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230027, China
| | - Yuwei Zhang
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China
| | - Shuai Zhou
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China
| | - Ran Xu
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Ihtisham Bukhari
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China; Molecular Pathology Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China; School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2258, Australia
| | - Jinming Li
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China
| | - Xu Dong Zhang
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China; Molecular Pathology Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China; School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Guangzhi Liu
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China.
| | - Song Chen
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China; Molecular Pathology Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China; Institute of Medicinal Biotechnology, Jiangsu College of Nursing, Huai'an, Jiangsu 223300, China.
| | - Mian Wu
- Translational Research Institute, Henan Provincial People's Hospital, Henan Key Laboratory of Stem Cell Differentiation and Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China; Zhengzhou City Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan Provincial Key Laboratory of Long Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou 450003, China; CAS Key Laboratory of Innate Immunity and Chronic Disease, CAS Centre for Excellence in Molecular Cell Science, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230027, China; Molecular Pathology Center, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China.
| | - Xiaoyuan Song
- MOE Key Laboratory for Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
19
|
Zhao X, Tang W, Wang H, He H. Femtosecond-laser stimulation induces senescence of tumor cells in vitro and in vivo. BIOMEDICAL OPTICS EXPRESS 2022; 13:791-804. [PMID: 35284179 PMCID: PMC8884195 DOI: 10.1364/boe.449456] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/18/2021] [Accepted: 12/23/2021] [Indexed: 05/30/2023]
Abstract
Tumor cells present anti-apoptosis and abnormal proliferation during development. Senescence and stemness of tumor cells play key roles in tumor development and malignancy. In this study, we show the transient stimulation by a single-time scanning of tightly focused femtosecond laser to tumor cells can modulate the stemness and senescence in vitro and in vivo. The laser-induced cellular senescence and stemness present distinct transitions in vitro and in vivo. The cells 1.2 mm deep in tumor tissue are found with significant senescence induced by the transient photostimulations in 100-200 µm shallow layer in vivo, which suppresses the growth of whole tumor in living mice.
Collapse
|
20
|
Alcina A, Fedetz M, Vidal-Cobo I, Andrés-León E, García-Sánchez MI, Barroso-Del-Jesus A, Eichau S, Gil-Varea E, Villar LM, Saiz A, Leyva L, Vandenbroeck K, Otaegui D, Izquierdo G, Comabella M, Urcelay E, Matesanz F. Identification of the genetic mechanism that associates L3MBTL3 to multiple sclerosis. Hum Mol Genet 2022; 31:2155-2163. [PMID: 35088080 PMCID: PMC9262392 DOI: 10.1093/hmg/ddac009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/19/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a complex and demyelinating disease of the central nervous system. One of the challenges of the post-GWAS era is to understand the molecular basis of statistical associations to reveal gene networks and potential therapeutic targets. The L3MBTL3 locus has been associated with MS risk by GWAS. To identify the causal variant of the locus, we performed fine mapping in a cohort of 3440 MS patients and 1688 healthy controls. The variant that best explained the association was rs6569648 (P = 4.13E-10, OR = 0.71, 95% CI = 0.64-0.79), which tagged rs7740107, located in intron 7 of L3MBTL3. The rs7740107 (A/T) variant has been reported to be the best expression and splice quantitative trait locus (eQTL and sQTL) of the region in up to 35 human GTEx tissues. By sequencing RNA from blood of 17 MS patients and quantification by digital qPCR, we determined that this eQTL/sQTL originated from the expression of a novel short transcript starting in intron 7 near rs7740107. The short transcript was translated into three proteins starting at different translation initiation codons. These N-terminal truncated proteins lacked the region where L3MBTL3 interacts with the transcriptional regulator RBPJ (Recombination Signal Binding Protein for Immunoglobulin Kappa J Region) which, in turn, regulates the Notch signaling pathway. Our data and other functional studies suggest that the genetic mechanism underlying the MS association of rs7740107 affects not only the expression of L3MBTL3 isoforms, but might also involve the Notch signaling pathway.
Collapse
Affiliation(s)
- Antonio Alcina
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas (IPBLN-CSIC) 18016 Granada, Spain
| | - Maria Fedetz
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas (IPBLN-CSIC) 18016 Granada, Spain
| | - Isabel Vidal-Cobo
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas (IPBLN-CSIC) 18016 Granada, Spain
| | - Eduardo Andrés-León
- Bioinformatic Unit, Instituto de Parasitología y Biomedicina López Neyra (IPBLN-CSIC), Granada, Spain
| | - Maria-Isabel García-Sánchez
- UGC Neurología. Nodo Hospital Universitario Virgen Macarena, Biobanco del Sistema Sanitario Público de Andalucía, Sevilla, (Spain)
| | - Alicia Barroso-Del-Jesus
- Genomics Unit, Instituto de Parasitología y Biomedicina López Neyra (IPBLN-CSIC), Granada, Spain
| | - Sara Eichau
- UGC Neurología. Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Elia Gil-Varea
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat). Institut de Recerca Vall d'Hebron (VHIR). Hospital Universitari Vall d'Hebron. Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Luisa-Maria Villar
- Departments of Immunology, Hospital Ramon y Cajal, (IRYCIS), Madrid, Spain
| | - Albert Saiz
- Servicio de Neurología, Hospital Clinic and Institut d'Investigació Biomèdica Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Laura Leyva
- Instituto de Investigación Biomédica de Málaga-IBIMA, UGC Neurología, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Koen Vandenbroeck
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain.,IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| | - David Otaegui
- Neurosciences Area, Biodonostia Health Research Institute, 20014 San Sebastián, Spain
| | - Guillermo Izquierdo
- Multiple Sclerosis Unit, Neurology Service, Vithas Nisa Hospital, 41950 Seville, Spain
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat). Institut de Recerca Vall d'Hebron (VHIR). Hospital Universitari Vall d'Hebron. Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Elena Urcelay
- Lab. of Genetics of Complex Diseases, Hospital Clinico San Carlos, Instituto de Investigacion Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Fuencisla Matesanz
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas (IPBLN-CSIC) 18016 Granada, Spain
| |
Collapse
|
21
|
Meesuwan S, Ketpun D, Piyaviriyakul P, Rattanapinyopituk K, Theewasutrakul P, Sailasuta A. Immunohistochemical and molecular profiling of CD 117, Oct-4, and Sox-2 in canine cutaneous mast cell tumor of the crossbred dogs in Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand. Vet World 2021; 14:2646-2654. [PMID: 34903921 PMCID: PMC8654761 DOI: 10.14202/vetworld.2021.2646-2654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/02/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND AIM CD 117 (c-KIT) internal tandem duplication (ITD), octamer-binding transcription factor 4 (Oct-4), and sex-determining region Y-box 2 (Sox-2) may govern the oncogenicity and aggressiveness of canine cutaneous mast cell tumor (MCT) in the crossbred dogs. Thus, a comprehension of this matter may help us establishing a novel platform to treat the disease in those dogs. However, evidence has lacked so far. Thus, this study aimed to survey CD 117 ITD, Oct-4, and Sox-2 expressions and their relations to the 2-tier grading in a group of Thai crossbreed dogs. The study was done using polymerase chain reaction (PCR), Reverse transcription PCR (RT-PCR), and immunohistochemistry. MATERIALS AND METHODS Thirty-three MCT specimens graded by the 2-tier histopathology grading were collected from the crossbred and purebred dogs. CD 117 ITD was detected by conventional PCR and immunohistochemistry. While, Oct-4 and Sox-2 expression levels were determined at the protein and mRNA levels by immunohistochemistry and RT-PCR, respectively. The expression magnitude of each parameter was then related to the grades and breeds. RESULTS About 60.61% of specimens were low grade, while 39.39% were high grade. CD 117 ITD was not detected in all specimens. A significant increase of Oct-4 expression was found in the high-grade, crossbred dogs. Meanwhile, Sox-2 expressions were increased both in the purebred and crossbred dogs with high-grade MCT. CONCLUSION The study finding has indicated that the level of Sox-2 expression may be a useful tumorigenic and prognostic biomarker because it correlates to the 2-tier grades but not dog breeds.
Collapse
Affiliation(s)
- Sirilak Meesuwan
- Veterinary Pathobiology Program, Graduate School, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330 Thailand
- Companion Animal Cancer Research Unit, CAC-RU, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Dettachai Ketpun
- Companion Animal Cancer Research Unit, CAC-RU, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
- Veterinary Pathology and Diagnosis Centre, Akkhraratchakumari Veterinary College, Walailak University, Nakhon Si Thammarat 80160, Thailand
- One Health Research Centre, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Prapruddee Piyaviriyakul
- Companion Animal Cancer Research Unit, CAC-RU, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Physiology, Biochemistry Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kasem Rattanapinyopituk
- Companion Animal Cancer Research Unit, CAC-RU, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Pattharakrit Theewasutrakul
- Companion Animal Cancer Research Unit, CAC-RU, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
- Oncology Clinic, Small Animal Teaching Hospital, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Achariya Sailasuta
- Companion Animal Cancer Research Unit, CAC-RU, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
22
|
Hsu LJ, Liu CL, Kuo ML, Shen CN, Shen CR. An Alternative Cell Therapy for Cancers: Induced Pluripotent Stem Cell (iPSC)-Derived Natural Killer Cells. Biomedicines 2021; 9:1323. [PMID: 34680440 PMCID: PMC8533510 DOI: 10.3390/biomedicines9101323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022] Open
Abstract
Cell therapy is usually defined as the treatment or prevention of human disease by supplementation with cells that have been selected, manipulated, and pharmacologically treated or altered outside the body (ex vivo). Induced pluripotent stem cells (iPSCs), with their unique characteristics of indefinite expansion in cultures and genetic modifications, represent an ideal cell source for differentiation into specialized cell types. Cell therapy has recently become one of the most promising therapeutic approaches for cancers, and different immune cell types are selected as therapeutic platforms. Natural killer (NK) cells are shown to be effective tumor cell killers and do not cause graft-vs-host disease (GVHD), making them excellent candidates for, and facilitating the development of, "off-the-shelf" cell therapies. In this review, we summarize the progress in the past decade in the advent of iPSC technology and review recent developments in gene-modified iPSC-NK cells as readily available "off-the-shelf" cellular therapies.
Collapse
Affiliation(s)
- Li-Jie Hsu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- PhD Program in Biotechnology Industry, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chao-Lin Liu
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei 243, Taiwan;
- Biochemical Technology R&D Center, Ming Chi University of Technology, New Taipei 243, Taiwan
| | - Ming-Ling Kuo
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Center of Molecular and Clinical Immunology, Chang Gung University, Taoyuan 333, Taiwan
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Lin-Kou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Department of Pediatrics, New Taipei Municipal TuCheng Hospital, New Taipei 236, Taiwan
| | - Chia-Ning Shen
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
| | - Chia-Rui Shen
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- PhD Program in Biotechnology Industry, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Center of Molecular and Clinical Immunology, Chang Gung University, Taoyuan 333, Taiwan
- Department of Ophthalmology, Lin-Kou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| |
Collapse
|
23
|
Li J, Haque M, Shang C, Hassan B, Liu D, Chen W, Lai R. Identification and Characterization of Cancer Stem-Like Cells in ALK-Positive Anaplastic Large Cell Lymphoma Using the SORE6 Reporter. Curr Issues Mol Biol 2021; 43:543-557. [PMID: 34287231 PMCID: PMC8929104 DOI: 10.3390/cimb43020041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/17/2021] [Accepted: 06/24/2021] [Indexed: 01/04/2023] Open
Abstract
Transcription factors Sox2 and Oct4 are essential in maintaining the pluripotency of embryonic stem cells and conferring stemness in cancer stem-like (CSL) cells. SORE6, an in-vitro reporter system, was designed to quantify the transcription activity of Sox2/Oct4 and identify CSL cells in non-hematologic cancers. Using SORE6, we identified and enriched CSL cells in ALK-positive anaplastic large cell lymphoma (ALK + ALCL). Two ALK + ALCL cell lines, SupM2 and UCONN-L2, contained approximately 20% of SORE6+ cells, which were purified based on their expression of green fluorescent protein. We then performed functional studies using single-cell clones derived from SORE6− and SORE6+ cells. Compared to SORE6− cells, SORE6+ cells were significantly more chemoresistant and clonogenic in colony-formation assays. Sox2/Oct4 are directly involved in conferring these CSL properties, since the shRNA knockdown of Sox2 in SORE6+ significantly lowered their chemoresistance, while enforced expression of Sox2/Oct4 in SORE6− cells produced opposite effects. Using Western blots, we found that the expression and subcellular localization of Sox2/Oct4 were similar between SORE6− and SORE6+ cells. However, in SORE6+ but not SORE6− cells, Sox2 and Oct4 abundantly bound to a probe containing the SORE6 consensus sequence. c-Myc, previously shown to regulate cancer stemness in ALK + ALCL, regulated the SORE6 activity. In conclusion, SORE6 is useful in identifying/enriching CSL cells in ALK + ALCL.
Collapse
Affiliation(s)
- Jing Li
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (J.L.); (M.H.); (C.S.); (B.H.); (D.L.); (W.C.)
- Electron Microscopy Center, Basic Medical Science College, Harbin Medical University, Harbin 150080, China
| | - Moinul Haque
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (J.L.); (M.H.); (C.S.); (B.H.); (D.L.); (W.C.)
- College of Medicine and Health, University College Cork, T12 AK54 Cork, Ireland
| | - Chuquan Shang
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (J.L.); (M.H.); (C.S.); (B.H.); (D.L.); (W.C.)
| | - Bardes Hassan
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (J.L.); (M.H.); (C.S.); (B.H.); (D.L.); (W.C.)
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Dongzhe Liu
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (J.L.); (M.H.); (C.S.); (B.H.); (D.L.); (W.C.)
- Laboratory of Biology and Chemistry, Basic Medical Science College, Harbin Medical University, Harbin 150080, China
| | - Will Chen
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (J.L.); (M.H.); (C.S.); (B.H.); (D.L.); (W.C.)
| | - Raymond Lai
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (J.L.); (M.H.); (C.S.); (B.H.); (D.L.); (W.C.)
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2R7, Canada
- Correspondence:
| |
Collapse
|
24
|
Aspartame induces cancer stem cell enrichment through p21, NICD and GLI1 in human PANC-1 pancreas adenocarcinoma cells. Food Chem Toxicol 2021; 153:112264. [PMID: 33992720 DOI: 10.1016/j.fct.2021.112264] [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: 08/01/2020] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 11/21/2022]
Abstract
This study aimed to investigate the molecular effects of the common natural sugar glucose and artificial sweetener aspartame on cancer stem cell (CSC) population and cancer aggressiveness of PANC-1 human pancreas adenocarcinoma cells. According to our findings while aspartame exposure significantly increased the CSC population, high glucose had no effect on it. The epithelial-mesenchymal transition marker N-cadherin increased only in the aspartame group. The findings indicate that a high level of glucose exposure does not effect the invasion and migration of PANC-1 cells, while aspartame increases both of these aggressiveness criteria. The findings also suggest that a high concentration of glucose maintains CSC population through induction of nuclear Oct3/4 and differentiation to parental cells via increasing cytoplasmic c-myc. Aspartame exposure to PANC-1 cells activated AKT and deactivated GSK3β by increasing levels of ROS and cytoplasmic Ca+2, respectively, through T1R2/T1R3 stimulation. Then p-GSK3β(Ser9) boosted the CSC population by increasing pluripotency factors Oct3/4 and c-myc via NICD, GLI1 and p21. In the aspartame group, T1R1 silencing further increased the CSC population but decreased cell viability and suppressed the p21, NICD and GLI activation. The presence and amount of T1R subunits in the membrane fraction of PANC-1 cells are demonstrated for the first time in this study, as is the regulatory effect of T1R1's on CSC population. In conclusion, the present study demonstrated that long-term aspartame exposure increases CSC population and tumor cell aggressiveness through p21, NICD, GLI1. Moreover, while aspartame had no tumorigenic effect, it could potentially advance an existing tumor.
Collapse
|
25
|
Fus-Kujawa A, Mendrek B, Trybus A, Bajdak-Rusinek K, Stepien KL, Sieron AL. Potential of Induced Pluripotent Stem Cells for Use in Gene Therapy: History, Molecular Bases, and Medical Perspectives. Biomolecules 2021; 11:biom11050699. [PMID: 34067183 PMCID: PMC8151405 DOI: 10.3390/biom11050699] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 01/14/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) are defined as reprogrammed somatic cells exhibiting embryonic stem cell characteristics. Since their discovery in 2006, efforts have been made to utilize iPSCs in clinical settings. One of the promising fields of medicine, in which genetically patient-specific stem cells may prove themselves useful, is gene therapy. iPSCs technology holds potential in both creating models of genetic diseases and delivering therapeutic agents into the organism via auto-transplants, which reduces the risk of rejection compared to allotransplants. However, in order to safely administer genetically corrected stem cells into patients’ tissues, efforts must be made to establish stably pluripotent stem cells and reduce the risk of insertional tumorigenesis. In order to achieve this, optimal reprogramming factors and vectors must be considered. Therefore, in this review, the molecular bases of reprogramming safe iPSCs for clinical applications and recent attempts to translate iPSCs technology into the clinical setting are discussed.
Collapse
Affiliation(s)
- Agnieszka Fus-Kujawa
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland; (A.F.-K.); (A.T.); (K.L.S.)
| | - Barbara Mendrek
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Sklodowskiej 34, 41-819 Zabrze, Poland;
| | - Anna Trybus
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland; (A.F.-K.); (A.T.); (K.L.S.)
| | - Karolina Bajdak-Rusinek
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland;
| | - Karolina L. Stepien
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland; (A.F.-K.); (A.T.); (K.L.S.)
| | - Aleksander L. Sieron
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Medykow 18 Street, 40-752 Katowice, Poland; (A.F.-K.); (A.T.); (K.L.S.)
- Correspondence:
| |
Collapse
|
26
|
Synthesis and biological evaluation of novel isoxazole-piperazine hybrids as potential anti-cancer agents with inhibitory effect on liver cancer stem cells. Eur J Med Chem 2021; 221:113489. [PMID: 33951549 DOI: 10.1016/j.ejmech.2021.113489] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023]
Abstract
In our effort for the development of novel anticancer therapeutics, a series of isoxazole-piperazine analogues were prepared, and primarily screened for their antiproliferative potential against hepatocellular carcinoma (HCC; Huh7/Mahlavu) and breast (MCF-7) cancer cells. All compounds demonstrated potent to moderate cytotoxicity on all cell lines with IC50 values in the range of 0.09-11.7 μM. Further biological studies with 6a and 13d in HCC cells have shown that both compounds induced G1 or G2/M arrests resulting in apoptotic cell death. Subsequent analysis of proteins involved in cell cycle progression as well as proliferation of HCC cells revealed that 6a and 13d may affect cellular survival pathways differently depending on the mutation profiles of cells (p53 and PTEN), epidermal/mesenchymal characteristics, and activation of cell mechanisms through p53 dependent/independent pathways. Lastly, we have demonstrated the potential anti-stemness properties of these compounds in which the proportion of liver CSCs in Huh7 cells (CD133+/EpCAM+) were significantly reduced by 6a and 13d. Furthermore, both compounds caused a significant reduction in expression of stemness markers, NANOG or OCT4 proteins, in Mahlavu and Huh7 cells, as well as resulted in a decreased sphere formation capacity in Huh7 cells. Together, these novel isoxazole-piperazine derivatives may possess potential as leads for development of effective anti-cancer drugs against HCC cells with stem cell-like properties.
Collapse
|
27
|
Abbasi Y, Hajiaghalou S, Baniasadi F, Mahabadi VP, Ghalamboran MR, Fathi R. Fe 3O 4 magnetic nanoparticles improve the vitrification of mouse immature oocytes and modulate the pluripotent genes expression in derived pronuclear-stage embryos. Cryobiology 2021; 100:81-89. [PMID: 33781804 DOI: 10.1016/j.cryobiol.2021.03.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 03/20/2021] [Accepted: 03/20/2021] [Indexed: 01/15/2023]
Abstract
The vitrification of Germinal Vesicle (immature) oocytes is beneficial for preservation of fertility in cases involving reproductive problems. The use of nanoparticles (NP(s)) as vitrification aid is a novel approach towards improving vitrification efficiency. The efficacy of use of iron oxide (Fe3O4) nanoparticles as vitrification aid is reported in this paper. Immature oocytes from NMRI mice were collected and divided into non-vitrified (nVit), Vitrified (Vit) and Vitrified + NP (Vit+NP) groups. In the Vit+NP group, solutions containing Fe3O4 nanoparticles at three different concentrations (0.004%, 0.008% and 0.016% w/v) were separately added to the vitrification solution and their effects on the vitrification of the oocytes were compared. The concentration that was found to be best performing (0.004% w/v) was used in vitrification studies in subsequent experiments. Mitochondrial function, apoptosis incidence, ultrastructure alteration, nuclear maturity, embryo formation and genes expression (Nanog, Oct4, Cdx2, and Sox2) were evaluated in response to the addition of the nanoparticle solution during vitrification. Nuclear maturity of oocyte and embryo formation increased significantly (P ≤ 0.05) in the vitrified + NP group. Expression of Sox2 also increased significantly in both vitrified and vitrified + NP groups. While there was a significant increase in Oct4 expression in the vitrified group as compared to control, there was no significant difference between vitrified and Vit+NP groups. The expression of Cdx2 decreased significantly (P ≤ 0.05) in the Vit+NP group. From these observations, Fe3O4 nanoparticles could protect immature oocytes from cryodamages, positively affect vitrification and modulate the pluripotency of derived pronuclear-stage embryos.
Collapse
Affiliation(s)
- Yasaman Abbasi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Department of Cell and Molecular Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Samira Hajiaghalou
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Farzaneh Baniasadi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Faculty of Science, Physics Department, Shahid Beheshti University, Iran
| | - Vahid Pirhajati Mahabadi
- Neuroscience Research Center, Iran University of Medical Science, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Science, Tehran, Iran
| | | | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
28
|
Postnatal Pluripotent Cells: Quarter of a Century of Research. Bull Exp Biol Med 2021; 170:515-521. [PMID: 33713237 DOI: 10.1007/s10517-021-05099-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Indexed: 10/21/2022]
Abstract
Almost quarter of a century long studies aimed at identification, isolation, culturing, and use of postnatal pluripotent cells for the development of cell-based technologies have not met with success and failed to provide reliable and reproducible protocols of cell isolation, identification, and culturing. At the same time, experimental data in this field suggest that postnatal pluripotent cells are not the copies of embryonic cells and, therefore, the tests routinely used for identification of embryonic pluripotent cells are not fully adequate for characterization of their postnatal analogues. Therefore, cell lineage tracing methods showing the differentiation routes of the studied cells in human or animal body after birth should be developed and used.
Collapse
|
29
|
Teo AKK, Nguyen L, Gupta MK, Lau HH, Loo LSW, Jackson N, Lim CS, Mallard W, Gritsenko MA, Rinn JL, Smith RD, Qian WJ, Kulkarni RN. Defective insulin receptor signaling in hPSCs skews pluripotency and negatively perturbs neural differentiation. J Biol Chem 2021; 296:100495. [PMID: 33667549 PMCID: PMC8050001 DOI: 10.1016/j.jbc.2021.100495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 02/18/2021] [Accepted: 03/01/2021] [Indexed: 11/26/2022] Open
Abstract
Human embryonic stem cells are a type of pluripotent stem cells (hPSCs) that are used to investigate their differentiation into diverse mature cell types for molecular studies. The mechanisms underlying insulin receptor (IR)-mediated signaling in the maintenance of human pluripotent stem cell (hPSC) identity and cell fate specification are not fully understood. Here, we used two independent shRNAs to stably knock down IRs in two hPSC lines that represent pluripotent stem cells and explored the consequences on expression of key proteins in pathways linked to proliferation and differentiation. We consistently observed lowered pAKT in contrast to increased pERK1/2 and a concordant elevation in pluripotency gene expression. ERK2 chromatin immunoprecipitation, luciferase assays, and ERK1/2 inhibitors established direct causality between ERK1/2 and OCT4 expression. Of importance, RNA sequencing analyses indicated a dysregulation of genes involved in cell differentiation and organismal development. Mass spectrometry–based proteomic analyses further confirmed a global downregulation of extracellular matrix proteins. Subsequent differentiation toward the neural lineage reflected alterations in SOX1+PAX6+ neuroectoderm and FOXG1+ cortical neuron marker expression and protein localization. Collectively, our data underscore the role of IR-mediated signaling in maintaining pluripotency, the extracellular matrix necessary for the stem cell niche, and regulating cell fate specification including the neural lineage.
Collapse
Affiliation(s)
- Adrian Kee Keong Teo
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Brigham and Women's Hospital, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA; Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Proteos, Singapore, Singapore; Department of Biochemistry and Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Linh Nguyen
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Proteos, Singapore, Singapore; Department of Biochemistry and Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Manoj K Gupta
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Brigham and Women's Hospital, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | - Hwee Hui Lau
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Proteos, Singapore, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Larry Sai Weng Loo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Proteos, Singapore, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Nicholas Jackson
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Brigham and Women's Hospital, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | - Chang Siang Lim
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Proteos, Singapore, Singapore
| | - William Mallard
- Department of Stem Cell and Regenerative Biology, Harvard University, and Broad Institute of MIT, Cambridge, Massachusetts, USA
| | - Marina A Gritsenko
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - John L Rinn
- Department of Stem Cell and Regenerative Biology, Harvard University, and Broad Institute of MIT, Cambridge, Massachusetts, USA
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Rohit N Kulkarni
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Brigham and Women's Hospital, Harvard Medical School, and Harvard Stem Cell Institute, Boston, Massachusetts, USA.
| |
Collapse
|
30
|
Simultaneous and quantitative monitoring transcription factors in human embryonic stem cell differentiation using mass spectrometry-based targeted proteomics. Anal Bioanal Chem 2021; 413:2081-2089. [PMID: 33655347 DOI: 10.1007/s00216-021-03160-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/15/2020] [Accepted: 01/06/2021] [Indexed: 11/27/2022]
Abstract
Human embryonic stem cells (hESCs) can be self-propagated indefinitely in culture while holding the capacity to generate almost all cell types. Although this powerful differentiation ability of hESCs has become a potential source of cell replacement therapies, application of stem cells in clinical practice relies heavily on the exquisite control of their developmental fate. In general, an essential first step in differentiation is to exit the pluripotent state, which is precariously balanced and depends on a variety of factors, mainly centering on the core transcriptional mechanism. To date, much evidence has indicated that transcription factors such as Sox2, Oct4, and Nanog control the self-renewal and pluripotency of hESCs. Their expression displays a restricted spatial-temporal pattern and their small changes in level can significantly affect directed differentiation and the cell type derived. So far, few assays have been developed to monitor this process. Herein, we provided a mass spectrometry (MS)-based approach for simultaneous and quantitative monitoring of these transcription factors, in an attempt to provide insight into their contributions in hESC differentiation.
Collapse
|
31
|
Mehravar M, Ghaemimanesh F, Poursani EM. An Overview on the Complexity of OCT4: at the Level of DNA, RNA and Protein. Stem Cell Rev Rep 2021; 17:1121-1136. [PMID: 33389631 DOI: 10.1007/s12015-020-10098-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2020] [Indexed: 10/22/2022]
Abstract
OCT4 plays critical roles in self-renewal and pluripotency maintenance of embryonic stem cells, and is considered as one of the main stemness markers. It also has pivotal roles in early stages of embryonic development. Most studies on OCT4 have focused on the expression and function of OCT4A, which is the biggest isoform of OCT4 known so far. Recently, many studies have shown that OCT4 has various transcript variants, protein isoforms, as well as pseudogenes. Distinguishing the expression and function of these variants and isoforms is a big challenge in expression profiling studies of OCT4. Understanding how OCT4 is functioning in different contexts, depends on knowing of where and when each of OCT4 transcripts, isoforms and pseudogenes are expressed. Here, we review OCT4 known transcripts, isoforms and pseudogenes, as well as its interactions with other proteins, and emphasize the importance of discriminating each of them in order to understand the exact function of OCT4 in stem cells, normal development and development of diseases.
Collapse
Affiliation(s)
- Majid Mehravar
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Fatemeh Ghaemimanesh
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Ensieh M Poursani
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Gómez-Varela AI, Gaspar R, Miranda A, Assis JL, Valverde RHF, Einicker-Lamas M, Silva BFB, De Beule PAA. Fluorescence cross-correlation spectroscopy as a valuable tool to characterize cationic liposome-DNA nanoparticle assembly. JOURNAL OF BIOPHOTONICS 2021; 14:e202000200. [PMID: 32827206 DOI: 10.1002/jbio.202000200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 06/11/2023]
Abstract
The development of nonviral gene delivery vehicles for therapeutic applications requires methods capable of quantifying the association between the genes and their carrier counterparts. Here we investigate the potential of fluorescence cross-correlation spectroscopy (FCCS) to characterize and optimize the assembly of nonviral cationic liposome (CL)-DNA complexes based on a CL formulation consisting of the cationic lipid DOTAP and zwitterionic lipid DOPC. We use a DNA plasmid for lipoplex loading encoding the Oct4 gene, critically involved in reprogramming somatic cells into induced pluripotent stem cells. We demonstrate that FCCS is able to quantitatively determine the extent of the association between DNA and the liposomes and assess its loading capacity. We also establish that the cationic lipid fraction, being proportional to the liposome membrane charge density, as well as charge ratio between the CLs and anionic DNA play an important role in the degree of interaction between the liposomes and DNA.
Collapse
Affiliation(s)
- Ana I Gómez-Varela
- International Iberian Nanotechnology Laboratory, Braga, Portugal
- Department of Applied Physics, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ricardo Gaspar
- International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - Adelaide Miranda
- International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - Juliane L Assis
- Biomembranes Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael H F Valverde
- Biomembranes Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Einicker-Lamas
- Biomembranes Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno F B Silva
- International Iberian Nanotechnology Laboratory, Braga, Portugal
| | | |
Collapse
|
33
|
Safitri E. Effect of low oxygen tension on transcriptional factor OCT4 and SOX2 expression in New Zealand rabbit bone marrow-derived mesenchymal stem cells. Vet World 2020; 13:2469-2476. [PMID: 33363343 PMCID: PMC7750229 DOI: 10.14202/vetworld.2020.2469-2476] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 10/14/2020] [Indexed: 01/09/2023] Open
Abstract
Background and Aim: Octamer-binding transcription factor 4 (OCT4) and sex-determining region Y-box 2 (SOX2) are transcription factors whose functions are essential to maintain the pluripotency of embryonic stem cells. The purpose of this study was to derive stem cells for in vitro culture and to maintain their viability and pluripotency, with the goal to obtain a cell line for transplantation in patients with degenerative diseases or injuries. This research focused on examining the effect of low oxygen tension on the ability of bone marrow-derived mesenchymal stem cells (BM-MSCs) to express OCT4 and SOX2 in vitro. Materials and Methods: BM-MSCs were obtained from femurs of 2000 to 3000 g New Zealand male rabbits. BM-MSCs were divided into three groups to test different culture conditions: A control group under hyperoxia condition (21% O2) and two treatment groups with low oxygen tension (1% and 3% O2). We characterized the BM-MSCs using flow cytometric measurement of cluster differentiation 44 (CD44) and cluster differentiation 90 (CD90) expression. The expression of OCT4 and SOX2 was measured by immunofluorescence staining after 48 h of incubation in chambers with normal or low oxygen tension with controlled internal atmosphere consisting of 95% N2, 5% CO2, and 1% O2 (T1) and 3% O2 (T2). We considered OCT4 and SOX2 as two markers of pluripotency induction. All immunofluorescence data were subjected to a post hoc normality Tukey’s honestly significant difference test; all differences with p<5% were considered significant. Results: BM-MSCs were positive for CD44 and CD90 expression after isolation. Oxygen tension culture conditions of 1% and 3% O2 led to OCT4 and SOX2 expression on culture days 2 and 4 (p<0.05), respectively, as compared to the hyperoxia condition (21% O2). Conclusion: Based on the OCT4 and SOX2 immunofluorescence data, we conclude that the stem cells were pluripotent at low O2 tension (at 1% O2 on day 2 and at 3% O2 on day 4), whereas under 21% O2 the OCT4 and SOX2 were not expressed.
Collapse
Affiliation(s)
- Erma Safitri
- Department of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Indonesia.,Stem Cells Research Division, Institute Tropical Disease, Universitas Airlangga, Surabaya 60115, Indonesia
| |
Collapse
|
34
|
Metz EP, Wuebben EL, Wilder PJ, Cox JL, Datta K, Coulter D, Rizzino A. Tumor quiescence: elevating SOX2 in diverse tumor cell types downregulates a broad spectrum of the cell cycle machinery and inhibits tumor growth. BMC Cancer 2020; 20:941. [PMID: 32998722 PMCID: PMC7528478 DOI: 10.1186/s12885-020-07370-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 08/31/2020] [Indexed: 11/17/2022] Open
Abstract
Background Quiescent tumor cells pose a major clinical challenge due to their ability to resist conventional chemotherapies and to drive tumor recurrence. Understanding the molecular mechanisms that promote quiescence of tumor cells could help identify therapies to eliminate these cells. Significantly, recent studies have determined that the function of SOX2 in cancer cells is highly dose dependent. Specifically, SOX2 levels in tumor cells are optimized to promote tumor growth: knocking down or elevating SOX2 inhibits proliferation. Furthermore, recent studies have shown that quiescent tumor cells express higher levels of SOX2 compared to adjacent proliferating cells. Currently, the mechanisms through which elevated levels of SOX2 restrict tumor cell proliferation have not been characterized. Methods To understand how elevated levels of SOX2 restrict the proliferation of tumor cells, we engineered diverse types of tumor cells for inducible overexpression of SOX2. Using these cells, we examined the effects of elevating SOX2 on their proliferation, both in vitro and in vivo. In addition, we examined how elevating SOX2 influences their expression of cyclins, cyclin-dependent kinases (CDKs), and p27Kip1. Results Elevating SOX2 in diverse tumor cell types led to growth inhibition in vitro. Significantly, elevating SOX2 in vivo in pancreatic ductal adenocarcinoma, medulloblastoma, and prostate cancer cells induced a reversible state of tumor growth arrest. In all three tumor types, elevation of SOX2 in vivo quickly halted tumor growth. Remarkably, tumor growth resumed rapidly when SOX2 returned to endogenous levels. We also determined that elevation of SOX2 in six tumor cell lines decreased the levels of cyclins and CDKs that control each phase of the cell cycle, while upregulating p27Kip1. Conclusions Our findings indicate that elevating SOX2 above endogenous levels in a diverse set of tumor cell types leads to growth inhibition both in vitro and in vivo. Moreover, our findings indicate that SOX2 can function as a master regulator by controlling the expression of a broad spectrum of cell cycle machinery. Importantly, our SOX2-inducible tumor studies provide a novel model system for investigating the molecular mechanisms by which elevated levels of SOX2 restrict cell proliferation and tumor growth.
Collapse
Affiliation(s)
- Ethan P Metz
- Eppley Institute for Research in Cancer and Allied Diseases Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Erin L Wuebben
- Eppley Institute for Research in Cancer and Allied Diseases Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Phillip J Wilder
- Eppley Institute for Research in Cancer and Allied Diseases Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Jesse L Cox
- Department of Pathology and Microbiology, University of Nebraska Medical Center Fred & Pamela Buffett Cancer Center, Omaha, NE, 68198-6805, USA
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Donald Coulter
- Department of Pediatrics, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Angie Rizzino
- Eppley Institute for Research in Cancer and Allied Diseases Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA. .,Department of Pathology and Microbiology, University of Nebraska Medical Center Fred & Pamela Buffett Cancer Center, Omaha, NE, 68198-6805, USA. .,Department of Biochemistry and Molecular Biology Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA.
| |
Collapse
|
35
|
Pagotto R, Santamaría CG, Harreguy MB, Abud J, Zenclussen ML, Kass L, Crispo M, Muñoz-de-Toro MM, Rodriguez HA, Bollati-Fogolín M. Perinatal exposure to Bisphenol A disturbs the early differentiation of male germ cells. Reprod Toxicol 2020; 98:117-124. [PMID: 32956838 DOI: 10.1016/j.reprotox.2020.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 09/02/2020] [Accepted: 09/05/2020] [Indexed: 12/27/2022]
Abstract
Understanding the effects of Bisphenol A (BPA) on early germ cell differentiation and their consequences in adult life is an area of growing interest in the field of endocrine disruption. Herein, we investigate whether perinatal exposure to BPA affects the differentiation of male germ cells in early life using a transgenic mouse expressing the GFP reporter protein under the Oct4 promoter. In this model, the expression of GFP reflects the expression of the Oct4 gene. This pluripotency gene is required to maintain the spermatogonial stem cells in an undifferentiated stage. Thus, GFP expression was used as a parameter to evaluate the effect of BPA on early germ cell development. Female pregnant transgenic mice were exposed to BPA by oral gavage, from embryonic day 5.5 to postnatal day 7 (PND7). The effects of BPA on male germ cell differentiation were evaluated at PND7, while sperm quality, testicular morphology, and protein expression of androgen receptor and proliferating cell nuclear antigen were studied at PND130. We found that perinatal/lactational exposure to BPA up-regulates the expression of Oct4-driven GFP in testicular cells at PND7. This finding suggests a higher proportion of undifferentiated spermatogonia in BPA-treated animals compared with non-exposed mice. Moreover, in adulthood, the number of spermatozoa per epididymis was reduced in those animals perinatally exposed to BPA. This work shows that developmental exposure to BPA disturbed the normal differentiation of male germ cells early in life, mainly by altering the expression of Oct4 and exerted long-lasting sequelae at the adult stage, affecting sperm count and testis.
Collapse
Affiliation(s)
- Romina Pagotto
- Cell Biology Unit, Institut Pasteur de Montevideo, Mataojo 2020, CP 11400 Montevideo, Uruguay
| | - Clarisa G Santamaría
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Ciudad Universitaria UNL, Ruta Nacional N°168, km 472, CPA S3000ZAA, Santa Fe, Argentina
| | - María Belén Harreguy
- Cell Biology Unit, Institut Pasteur de Montevideo, Mataojo 2020, CP 11400 Montevideo, Uruguay
| | - Julián Abud
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Ciudad Universitaria UNL, Ruta Nacional N°168, km 472, CPA S3000ZAA, Santa Fe, Argentina
| | - María Laura Zenclussen
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Ciudad Universitaria UNL, Ruta Nacional N°168, km 472, CPA S3000ZAA, Santa Fe, Argentina
| | - Laura Kass
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Ciudad Universitaria UNL, Ruta Nacional N°168, km 472, CPA S3000ZAA, Santa Fe, Argentina
| | - Martina Crispo
- Transgenic and Experimental Animal Unit, Institut Pasteur de Montevideo, Mataojo 2020, CP 11400 Montevideo, Uruguay
| | - Mónica M Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Ciudad Universitaria UNL, Ruta Nacional N°168, km 472, CPA S3000ZAA, Santa Fe, Argentina
| | - Horacio A Rodriguez
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Ciudad Universitaria UNL, Ruta Nacional N°168, km 472, CPA S3000ZAA, Santa Fe, Argentina
| | - Mariela Bollati-Fogolín
- Cell Biology Unit, Institut Pasteur de Montevideo, Mataojo 2020, CP 11400 Montevideo, Uruguay.
| |
Collapse
|
36
|
Nieto-Nicolau N, de la Torre RM, Fariñas O, Savio A, Vilarrodona A, Casaroli-Marano RP. Extrinsic modulation of integrin α6 and progenitor cell behavior in mesenchymal stem cells. Stem Cell Res 2020; 47:101899. [PMID: 32659733 DOI: 10.1016/j.scr.2020.101899] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/11/2020] [Accepted: 06/24/2020] [Indexed: 01/07/2023] Open
Abstract
Mesenchymal stem cells (MSC) are heterogeneous cells of complex nature that show different potentials while different culture conditions can modify their functionalities through interactions with the microenviroment. Here, we found that bone marrow (BM) MSC from different donor sources and passages that expressed higher levels of α6 integrin subunit (ITGA6), showed higher clonogenicity, migration and differentiation potential. ITGA6 showed important roles improving these potentials and regulating proliferation through protein kinase B (AKT) pathway and cell cycle inhibitor proteins p53 and p21. Moreover, ITGA6 downregulation impaired migration. Cell confluence regulated ITGA6, increasing its expression in low density cultures and decreasing in high density cultures. Besides, ITGA6- cells expressed ITGA6 when seeded at low densities. We found higher ITGA6 expression on fibronectin substrates at lower confluency. Fibronectin increased proliferation, clonogenicity, activation of AKT, decreased cell cycle inhibitor proteins and augmented growth factors expression. Spheres-derived MSC showed higher ITGA6 expression and enhanced potentials for migration, clonogenicity and proliferation. In conclusion, though there is an intrinsic regulation of ITGA6 expression, associated to the progenitor potential of BM-MSC, this expression is regulated by culture conditions and is translated in changes in cell behavior and proliferation. This knowledge could be used to enhance the potential of BM-MSC for clinical application.
Collapse
Affiliation(s)
- Nuria Nieto-Nicolau
- Barcelona Tissue Bank (BTB) & Donor Center, Banc de Sang i Teixits (BST), Barcelona, Spain; CellTec-UB, University of Barcelona, Barcelona, Spain; Institute of Biomedical Research Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | | | - Oscar Fariñas
- Barcelona Tissue Bank (BTB) & Donor Center, Banc de Sang i Teixits (BST), Barcelona, Spain; Institute of Biomedical Research Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Andrés Savio
- Barcelona Tissue Bank (BTB) & Donor Center, Banc de Sang i Teixits (BST), Barcelona, Spain; Institute of Biomedical Research Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Anna Vilarrodona
- Barcelona Tissue Bank (BTB) & Donor Center, Banc de Sang i Teixits (BST), Barcelona, Spain; Institute of Biomedical Research Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Ricardo P Casaroli-Marano
- Barcelona Tissue Bank (BTB) & Donor Center, Banc de Sang i Teixits (BST), Barcelona, Spain; CellTec-UB, University of Barcelona, Barcelona, Spain; Department of Surgery, School of Medicine & Hospital Clinic de Barcelona, University of Barcelona, Barcelona, Spain; Institute of Biomedical Research Sant Pau (IIB-Sant Pau), Barcelona, Spain.
| |
Collapse
|
37
|
Tsui YM, Chan LK, Ng IOL. Cancer stemness in hepatocellular carcinoma: mechanisms and translational potential. Br J Cancer 2020; 122:1428-1440. [PMID: 32231294 PMCID: PMC7217836 DOI: 10.1038/s41416-020-0823-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/30/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer stemness, referring to the stem-cell-like phenotype of cancer cells, has been recognised to play important roles in different aspects of hepatocarcinogenesis. A number of well-established cell-surface markers already exist for liver cancer stem cells, with potential new markers of liver cancer stem cells being identified. Both genetic and epigenetic factors that affect various signalling pathways are known to contribute to cancer stemness. In addition, the tumour microenvironment—both physical and cellular—is known to play an important role in regulating cancer stemness, and the potential interaction between cancer stem cells and their microenvironment has provided insight into the regulation of the tumour-initiating ability as well as the cellular plasticity of liver CSCs. Potential specific therapeutic targeting of liver cancer stemness is also discussed. With increased knowledge, effective druggable targets might be identified, with the aim of improving treatment outcome by reducing chemoresistance.
Collapse
Affiliation(s)
- Yu-Man Tsui
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Lo-Kong Chan
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong.,State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Irene Oi-Lin Ng
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong. .,State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong.
| |
Collapse
|
38
|
Verneri P, Vazquez Echegaray C, Oses C, Stortz M, Guberman A, Levi V. Dynamical reorganization of the pluripotency transcription factors Oct4 and Sox2 during early differentiation of embryonic stem cells. Sci Rep 2020; 10:5195. [PMID: 32251342 PMCID: PMC7089971 DOI: 10.1038/s41598-020-62235-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/11/2020] [Indexed: 01/29/2023] Open
Abstract
Pluripotency maintenance requires transcription factors (TFs) that induce genes necessary to preserve the undifferentiated state and repress others involved in differentiation. Recent observations support that the heterogeneous distribution of TFs in the nucleus impacts on gene expression. Thus, it is essential to explore how TFs dynamically organize to fully understand their role in transcription regulation. Here, we examine the distribution of pluripotency TFs Oct4 and Sox2 in the nucleus of embryonic stem (ES) cells and inquire whether their organization changes during early differentiation stages preceding their downregulation. Using ES cells expressing Oct4-YPet or Sox2-YPet, we show that Oct4 and Sox2 partition between nucleoplasm and a few chromatin-dense foci which restructure after inducing differentiation by 2i/LIF withdrawal. Fluorescence correlation spectroscopy showed distinct changes in Oct4 and Sox2 dynamics after differentiation induction. Specifically, we detected an impairment of Oct4-chromatin interactions whereas Sox2 only showed slight variations in its short-lived, and probably more unspecific, interactions with chromatin. Our results reveal that differentiation cues trigger early changes of Oct4 and Sox2 nuclear distributions that also include modifications in TF-chromatin interactions. This dynamical reorganization precedes Oct4 and Sox2 downregulation and may contribute to modulate their function at early differentiation stages.
Collapse
Affiliation(s)
- Paula Verneri
- CONICET - Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina
| | - Camila Vazquez Echegaray
- CONICET - Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina
| | - Camila Oses
- CONICET - Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina
| | - Martin Stortz
- CONICET - Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina.,Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Buenos Aires, Argentina
| | - Alejandra Guberman
- CONICET - Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina. .,Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Buenos Aires, Argentina.
| | - Valeria Levi
- CONICET - Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina.
| |
Collapse
|
39
|
Yu C, Liu Q, Chen C, Wang J. Quantification of the Underlying Mechanisms and Relationships Among Cancer, Metastasis, and Differentiation and Development. Front Genet 2020; 10:1388. [PMID: 32194614 PMCID: PMC7061528 DOI: 10.3389/fgene.2019.01388] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/19/2019] [Indexed: 12/28/2022] Open
Abstract
Recurrence and metastasis have been regarded as two of the greatest obstacles to cancer therapy. Cancer stem cells (CSCs) contribute to cancer development, with the distinctive features of recurrence and resistance to popular treatments such as drugs and chemotherapy. In addition, recent discoveries suggest that the epithelial mesenchymal transition (EMT) is an essential process in normal embryogenesis and tissue repair, as well as being a required step in cancer metastasis. Although there are many indications of the connections between metastasis and stem cells, these have often been studied separately or at most bi-laterally, not in an integrated way. In this study, we aimed to explore the global mechanisms and interrelationships among cancer, development, and metastasis, which are currently poorly understood. First, we constructed a core gene regulatory network containing specific genes and microRNAs of CSCs, EMT, and cancer. We uncovered seven distinct states emerging from the underlying landscape, denoted normal, premalignant, cancer, stem cell, CSC, lesion, and hyperplasia. Given the biological definition of each state, we also discuss the metastasis ability of each state. We show how and which types of cells can be transformed to a cancer state, and the connections among cancer, CSCs, and EMT. The barrier height and flux of the kinetic paths are explored to quantify how and which cells switch stochastically between the states. Our landscape model provides a quantitative approach to reveal the global mechanisms of cancer, development, and metastasis.
Collapse
Affiliation(s)
- Chong Yu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.,University of Science and Technology of China, Hefei, China
| | - Qiong Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Cong Chen
- Department of Chemistry and of Physics and Astronomy, State University of New York at Stony Brook, Stony Brook, NY, United States
| | - Jin Wang
- Department of Chemistry and of Physics and Astronomy, State University of New York at Stony Brook, Stony Brook, NY, United States
| |
Collapse
|
40
|
Hussein MNA, Cao X, Elokil AA, Huang S. Characterisation of stem and proliferating cells on the retina and lens of loach Misgurnus anguillicaudatus. JOURNAL OF FISH BIOLOGY 2020; 96:102-110. [PMID: 31674006 DOI: 10.1111/jfb.14189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 10/30/2019] [Indexed: 06/10/2023]
Abstract
The eye of the fish has a lifelong persistent neurogenesis unlike eye of mammals, so it's highly interesting to study retinal neurogenesis and its genetic control to give complete knowledge about the cause of this property in fish in comparison to mammals. We performed fluorescent in situ hybridisation for loach Misgurnus anguillicaudatus bmi1, msi1 and sox2 genes, which are used as an indicator of the sites of multipotent stem cells. Proliferating cell nuclear antigen (PCNA), bromodeoxyuridine (BRDU) and KI67 markers were used as indicators of proliferating cells and glial fibrillary acidic protein (GFAP) immunofluorescence was used for detection of the glial property of cells, as well as, immunohistochemistry detected the role of peroxisome proliferator-activated receptor (PPAR)α and γ in retinal neurogenesis. Our results determined that the lens and the retina of loach M. anguillicaudatus contain proliferative and pluripotent stem cells that have both glial and neuroepithelial properties, which add new cells continuously throughout life even without injury-induced proliferation. The PPARα has an essential function in providing energy supply for retinal neurogenesis more than PPARγ.
Collapse
Affiliation(s)
- Mona N A Hussein
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Xiaojuan Cao
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, Hubei, China
| | - Abdelmotaleb A Elokil
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Animal Productions Department, Faculty of Agriculture, Benha University, Benha, Egypt
| | - Songqian Huang
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Japan
| |
Collapse
|
41
|
Liu ML, Zang F, Zhang SJ. RBCK1 contributes to chemoresistance and stemness in colorectal cancer (CRC). Biomed Pharmacother 2019; 118:109250. [DOI: 10.1016/j.biopha.2019.109250] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023] Open
|
42
|
Niloy KK, Gulfam M, Compton KB, Li D, Huang GTJ, Lowe TL. Methacrylated Hyaluronic Acid–Based Hydrogels Maintain Stemness in Human Dental Pulp Stem Cells. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00115-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
43
|
Steevens AR, Glatzer JC, Kellogg CC, Low WC, Santi PA, Kiernan AE. SOX2 is required for inner ear growth and cochlear nonsensory formation before sensory development. Development 2019; 146:dev.170522. [PMID: 31152002 DOI: 10.1242/dev.170522] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 05/13/2019] [Indexed: 12/16/2022]
Abstract
The transcription factor sex determining region Y-box 2 (SOX2) is required for the formation of hair cells and supporting cells in the inner ear and is a widely used sensory marker. Paradoxically, we demonstrate via fate mapping that, initially, SOX2 primarily marks nonsensory progenitors in the mouse cochlea, and is not specific to all sensory regions until late otic vesicle stages. SOX2 fate mapping reveals an apical-to-basal gradient of SOX2 expression in the sensory region of the cochlea, reflecting the pattern of cell cycle exit. To understand SOX2 function, we undertook a timed-deletion approach, revealing that early loss of SOX2 severely impaired morphological development of the ear, whereas later deletions resulted in sensory disruptions. During otocyst stages, SOX2 shifted dramatically from a lateral to medial domain over 24-48 h, reflecting the nonsensory-to-sensory switch observed by fate mapping. Early loss or gain of SOX2 function led to changes in otic epithelial volume and progenitor proliferation, impacting growth and morphological development of the ear. Our study demonstrates a novel role for SOX2 in early otic morphological development, and provides insights into the temporal and spatial patterns of sensory specification in the inner ear.
Collapse
Affiliation(s)
- Aleta R Steevens
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jenna C Glatzer
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Courtney C Kellogg
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Walter C Low
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Peter A Santi
- Department of Otolaryngology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Amy E Kiernan
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY 14642, USA .,Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
44
|
Katsura Y, Ohara T, Noma K, Ninomiya T, Kashima H, Kato T, Sato H, Komoto S, Narusaka T, Tomono Y, Xing B, Chen Y, Tazawa H, Kagawa S, Shirakawa Y, Kasai T, Seno M, Matsukawa A, Fujiwara T. A Novel Combination Cancer Therapy with Iron Chelator Targeting Cancer Stem Cells via Suppressing Stemness. Cancers (Basel) 2019; 11:cancers11020177. [PMID: 30717462 PMCID: PMC6406536 DOI: 10.3390/cancers11020177] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 01/28/2019] [Accepted: 01/31/2019] [Indexed: 02/07/2023] Open
Abstract
Excess iron causes cancer and is thought to be related to carcinogenesis and cancer progression including stemness, but the details remain unclear. Here, we hypothesized that stemness in cancer is related to iron metabolism and that regulating iron metabolism in cancer stem cells (CSCs) may be a novel therapy. In this study, we used murine induced pluripotent stem cells that expressed specific stem cell genes such as Nanog, Oct3/4, Sox2, Klf4, and c-Myc, and two human cancer cell lines with similar stem cell gene expression. Deferasirox, an orally available iron chelator, suppressed expression of stemness markers and spherogenesis of cells with high stemness status in vitro. Combination therapy had a marked antitumor effect compared with deferasirox or cisplatin alone. Iron metabolism appears important for maintenance of stemness in CSCs. An iron chelator combined with chemotherapy may be a novel approach via suppressing stemness for CSC targeted therapy.
Collapse
Affiliation(s)
- Yuki Katsura
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Toshiaki Ohara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Kazuhiro Noma
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Takayuki Ninomiya
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Hajime Kashima
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Takuya Kato
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Hiroaki Sato
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Satoshi Komoto
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Toru Narusaka
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Yasuko Tomono
- Shigei Medical Research Institute, Okayama 701-0202, Japan.
| | - Boyi Xing
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Yuehua Chen
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama 700-8558, Japan.
| | - Shunsuke Kagawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Yasuhiro Shirakawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| | - Tomonari Kasai
- School of Bioscience and Biotechnology, Tokyo University of Technology, Tokyo 192-0914, Japan.
| | - Masaharu Seno
- Laboratory of Nano-Biotechnology, Okayama University Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama 700-8530, Japan.
| | - Akihiro Matsukawa
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan (Y.K.).
| |
Collapse
|
45
|
Xie Y, Nurkesh AA, Ibragimova N, Zhanzak Z, Meyerbekova A, Alexeyeva Z, Yesbolatova A, Satayeva M, Mustafa A, Manarbek L, Maipas A, Altaikyzy A, Keneskhanova Z, Akbay B, Chen Z. Systematic analysis of NLMP suggests nuclear localization of RTK/MET kinases resemble cancer cell clearance. J Exp Clin Cancer Res 2019; 38:43. [PMID: 30700325 PMCID: PMC6354337 DOI: 10.1186/s13046-018-1004-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/13/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Some membrane proteins can translocate into the nucleus, defined as nuclear localized membrane proteins (NLMPs), including receptor tyrosine kinases (RTKs). We previously showed that nuclear MET (nMET), a member of RTKs, mediates cancer stem-like cells self-renewal to promote cancer recurrence. However, it is unknown that nMET or mMET, which is the ancestor in the evolution of cancer cell survival and clearance. Here, we aim to study the NLMP functions in cell death, differentiation and survival. METHOD We applied the systematic reanalysis of functional NLMP and clinical investigations of nMET from databases. In addition, we used soft agar assay, immunoblotting, flow cytometry, and immunofluorescence confocal microscopy for examinations of nMET functions including stem-like cell formation, cell signaling, cell cycle regulation, and co-localization with regulators of cell signaling. ShRNA, antibody of recognizing surface membrane MET based treatment were used to downregulate endogenous nMET to uncover its function. RESULTS We predicted and demonstrated that nMET and nEGFR are most likely not ancestors. nMET overexpression induces both cell death and survival with drug resistance and stem cell-like characters. Moreover, the paradoxical function of nMET in both cell death and cell survival is explained by the fact that nMET induces stem cell-like cell growth, DNA damage repair, to evade the drug sensitization for survival of single cells while non-stem cell-like nMET expressing single cells may undergo clearance by cell death through cell cycle arrest induced by p21. CONCLUSION Taken together, our data suggest a link between nuclear RTK and cancer cell evolutionary clearance via cell death, and drug resistance for survival through stemness selection. Targeting evolved nuclear RTKs in cancer stem cells would be a novel avenue for precision cancer therapy.
Collapse
Affiliation(s)
- Yingqiu Xie
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Ayan A. Nurkesh
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Nazgul Ibragimova
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Zhuldyz Zhanzak
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Aizhan Meyerbekova
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Zhanna Alexeyeva
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Aiya Yesbolatova
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Madina Satayeva
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Aidana Mustafa
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Limara Manarbek
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Aisulu Maipas
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Akerke Altaikyzy
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Zhibek Keneskhanova
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Burkitkan Akbay
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Zhenbang Chen
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208 USA
| |
Collapse
|
46
|
Yu M, Wei Y, Xu K, Liu S, Ma L, Pei Y, Hu Y, Liu Z, Zhang X, Wang B, Mu Y, Li K. EGFR deficiency leads to impaired self-renewal and pluripotency of mouse embryonic stem cells. PeerJ 2019; 7:e6314. [PMID: 30713819 PMCID: PMC6357870 DOI: 10.7717/peerj.6314] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 12/17/2018] [Indexed: 12/23/2022] Open
Abstract
Background Self-renewal and pluripotency are considered as unwavering features of embryonic stem cells (ESCs). How ESCs regulate the self-renewal and differentiation is a central question in development and regenerative medicine research. Epidermal growth factor receptor (EGFR) was identified as a critical regulator in embryonic development, but its role in the maintenance of ESCs is poorly understood. Methods Here, EGFR was disrupted by its specific inhibitor AG1478 in mouse ESCs (mESCs), and its self-renewal and pluripotency were characterized according to their proliferation, expression of pluripotency markers, embryoid body (EB) formation, and mRNA expression patterns. We also used another EGFR inhibitor (gefitinib) and RNA interference assay to rule out the possibility of non-specific effects of AG1478. Results EGFR inhibition by AG1478 treatment in mESCs markedly reduced cell proliferation, caused cell cycle arrest at G0/G1 phase, and altered protein expressions of the cell cycle regulatory genes (CDK2 (decreased 11.3%) and proliferating cell nuclear antigen (decreased 25.2%)). The immunoreactivities and protein expression of pluripotency factors (OCT4 (decreased 26.9%)) also dramatically decreased, while the differentiation related genes (GATA4 (increased 1.6-fold)) were up-regulated in mESCs after EGFR inhibition. Meanwhile, EGFR inhibition in mESCs disrupted EB formation, indicating its impaired pluripotency. Additionally, the effects observed by EGFR inhibition with another inhibitor gefitinib and siRNA were consistent with those observed by AG1478 treatment in mESCs. These effects were manifested in the decreased expression of proliferative and pluripotency-related genes and the increased expression of genes involved in differentiation. Moreover, RNA-seq analysis displayed that transcript profiling was changed significantly after EGFR inhibition by AG1478. A large number of differentially expressed genes were involved in cell cycle, apoptotic process, epigenetic modification, and metabolic process, which were related to self-renewal and pluripotency, confirming that EGFR deficiency impaired self-renewal and pluripotency in mESCs. Conclusions Taken together, our results demonstrated the importance of EGFR in guarding the stemness of mESCs.
Collapse
Affiliation(s)
- Miaoying Yu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China.,College of Life Science, Shangrao Normal University, Shangrao, Jiangxi, China
| | - Yinghui Wei
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Kui Xu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shasha Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lei Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China.,College of Life Science, Shihezi University, Shihezi, Xinjiang, China
| | - Yangli Pei
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yanqing Hu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhiguo Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xue Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Bingyuan Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yulian Mu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Kui Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
47
|
Histone methyltransferase NSD2 regulates apoptosis and chemosensitivity in osteosarcoma. Cell Death Dis 2019; 10:65. [PMID: 30683853 PMCID: PMC6347630 DOI: 10.1038/s41419-019-1347-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/31/2018] [Accepted: 01/07/2019] [Indexed: 12/19/2022]
Abstract
Osteosarcoma (OS) is a primary malignant bone tumour. However, the genetic basis for the pathogenesis of OS remains elusive. In this study, we uncovered the role of the histone methyltransferase NSD2 in regulating tumourigenesis and chemosensitivity in OS. We show that NSD2 knockdown leads to increased apoptosis in OS cells in vitro and in vivo. Additionally, NSD2 knockdown significantly enhances the efficacy of cisplatin against OS cells and accordingly inhibits properties associated with cancer stem cells (CSCs). Furthermore, RNA sequencing (RNAseq) and Gene Ontology (GO) analysis revealed that NSD2 promotes transcription of genes associated with negative regulation of apoptotic signalling pathways and CSC properties. The results of chromatin immunoprecipitation quantitative polymerase chain reaction (ChIP-qPCR) assays indicated that NSD2 knockdown leads to decreased H3K36me2 modification at BCL2 and SOX2 loci, thus inhibiting the transcription of these two genes that are closely correlated with apoptosis, CSC properties and chemosensitivity in OS cells. Pathway analysis demonstrated that the ERK and AKT pathways mediate the regulation of OS progression and chemosensitivity by NSD2. Overall, our study is the first to uncover the function of NSD2 in OS chemosensitivity. NSD2 regulates the expression of the apoptosis regulatory proteins BCL2 and SOX2 through the ERK and AKT pathways. Our results suggest that NSD2 is a new target for combined chemotherapy and is a prognostic factor for OS.
Collapse
|
48
|
Lu Y, Qu H, Qi D, Xu W, Liu S, Jin X, Song P, Guo Y, Jia Y, Wang X, Li H, Li Y, Quan C. OCT4 maintains self-renewal and reverses senescence in human hair follicle mesenchymal stem cells through the downregulation of p21 by DNA methyltransferases. Stem Cell Res Ther 2019; 10:28. [PMID: 30646941 PMCID: PMC6334457 DOI: 10.1186/s13287-018-1120-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 12/11/2018] [Accepted: 12/20/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Self-renewal is dependent on an intrinsic gene regulatory network centered on OCT4 and on an atypical cell cycle G1/S transition, which is also regulated by OCT4. p21, a gene negatively associated with self-renewal and a senescence marker, is a member of the universal cyclin-dependent kinase inhibitors (CDKIs) and plays critical roles in the regulation of the G1/S transition. The expression of p21 can be regulated by OCT4-targeted DNA methyltransferases (DNMTs), which play distinct roles in gene regulation and maintaining pluripotency properties. The aim of this study was to determine the role of OCT4 in the regulation of self-renewal and senescence in human hair follicle mesenchymal stem cells (hHFMSCs) and to characterize the molecular mechanisms involved. METHODS A lentiviral vector was used to ectopically express OCT4. The influences of OCT4 on the self-renewal and senescence of hHFMSCs were investigated. Next-generation sequencing (NGS) was performed to identify the downstream genes of OCT4 in this process. Methylation-specific PCR (MSP) analysis was performed to measure the methylation level of the p21 promoter region. p21 was overexpressed in hHFMSCsOCT4 to test its downstream effect on OCT4. The regulatory effect of OCT4 on DNMTs was examined by ChIP assay. 5-aza-dC/zebularine was used to inhibit the expression of DNMTs, and then self-renewal properties and senescence in hHFMSCs were detected. RESULTS The overexpression of OCT4 promoted proliferation, cell cycle progression, and osteogenic differentiation capacity of hHFMSCs. The cell senescence of hHFMSCs was markedly suppressed due to the ectopic expression of OCT4. Through NGS, we identified 2466 differentially expressed genes (DEGs) between hHFMSCsOCT4 and hHFMSCsEGFP, including p21, which was downregulated. The overexpression of p21 abrogated the proliferation and osteogenic differentiation capacity of hHFMSCsOCT4 and promoted cell senescence. OCT4 enhanced the transcription of DNMT genes, leading to an elevation in the methylation of the p21 promoter. The inhibition of DNMTs reversed the OCT4-induced p21 reduction, depleted the self-renewal of hHFMSCsOCT4, and triggered cell senescence. CONCLUSIONS OCT4 maintains the self-renewal ability of hHFMSCs and reverses senescence by suppressing the expression of p21 through the upregulation of DNMTs.
Collapse
Affiliation(s)
- Yan Lu
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, China
| | - Huinan Qu
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, China
| | - Da Qi
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, China
| | - Wenhong Xu
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, China
| | - Shutong Liu
- Cell Processing Section, Department of Transfusion, Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xiangshu Jin
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, China
| | - Peiye Song
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, China
| | - Yantong Guo
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, China
| | - Yiyang Jia
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, China
| | - Xinqi Wang
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, China
| | - Hairi Li
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, 92093-0651, USA
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, China
| | - Chengshi Quan
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, China.
| |
Collapse
|
49
|
Zhang XM, Huang Y, Cong X, Qu LH, Zhang K, Wu LL, Zhang Y, Yu GY. Parasympathectomy increases resting secretion of the submandibular gland in minipigs in the long term. J Cell Physiol 2018; 234:9515-9524. [PMID: 30387129 DOI: 10.1002/jcp.27640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/02/2018] [Indexed: 11/09/2022]
Abstract
Parasympathectomy leads to retrogressive alteration and dysfunction of the submandibular gland (SMG) within 1 month, but its long-term effect is unclear. Excessive secretion is observed in half of the patients 4-6 months after SMG transplantation, which completely denervates the gland. Here, we investigated the long-term effect of parasympathectomy on the secretion of SMGs in minipigs. The results showed that the resting salivary secretion of SMGs decreased by 82.9% of that in control at 2 months after denervation, but increased by 156% at 6 months. Although experiencing an atrophic period, the denervated glands regained their normal morphology by 6 months. The expression of the function-related proteins, including muscarinic acetylcholine receptor (mAChR) 3, aquaporin 5 (AQP5), tight junction protein claudin-3, and claudin-4 was decreased at 2 months after denervation. Meanwhile, the protein expression of stem cell markers, including sex-determining region Y-box 2 and octamer-binding transcription factor 4, and the number of Ki67+ cells were significantly increased. However, at 6 months after denervation, the expression of mAChR3, AQP5, claudin-1, claudin-3, and claudin-4 was significantly raised, and the membrane distribution of these proteins was increased accordingly. The autonomic axonal area of the glands was reduced at 2 months after denervation but returned to the control level at 6 months, suggesting that reinnervation took place in the long term. In summary, parasympathectomy increases resting secretion of the SMGs in the long term with a possible mechanism involving improved transepithelial fluid transport. This finding may provide a new strategy for xerostomia treatment.
Collapse
Affiliation(s)
- Xue-Ming Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology and Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yan Huang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology and Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Xin Cong
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Ling-Han Qu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology and Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Kuo Zhang
- Department of Laboratory Animal Science, Peking University Health Science Center, Beijing, China
| | - Li-Ling Wu
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Yan Zhang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Guang-Yan Yu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology and Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
50
|
Vilas JM, Carneiro C, Da Silva-Álvarez S, Ferreirós A, González P, Gómez M, Ortega S, Serrano M, García-Caballero T, González-Barcia M, Vidal A, Collado M. Adult Sox2+ stem cell exhaustion in mice results in cellular senescence and premature aging. Aging Cell 2018; 17:e12834. [PMID: 30129215 PMCID: PMC6156495 DOI: 10.1111/acel.12834] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 06/25/2018] [Accepted: 07/21/2018] [Indexed: 12/14/2022] Open
Abstract
Aging is characterized by a gradual functional decline of tissues with age. Adult stem and progenitor cells are responsible for tissue maintenance, repair, and regeneration, but during aging, this population of cells is decreased or its activity is reduced, compromising tissue integrity and causing pathologies that increase vulnerability, and ultimately lead to death. The causes of stem cell exhaustion during aging are not clear, and whether a reduction in stem cell function is a cause or a consequence of aging remains unresolved. Here, we took advantage of a mouse model of induced adult Sox2+ stem cell depletion to address whether accelerated stem cell depletion can promote premature aging. After a short period of partial repetitive depletion of this adult stem cell population in mice, we observed increased kyphosis and hair graying, and reduced fat mass, all of them signs of premature aging. It is interesting that cellular senescence was identified in kidney after this partial repetitive Sox2+ cell depletion. To confirm these observations, we performed a prolonged protocol of partial repetitive depletion of Sox2+ cells, forcing regeneration from the remaining Sox2+ cells, thereby causing their exhaustion. Senescence specific staining and the analysis of the expression of genetic markers clearly corroborated that adult stem cell exhaustion can lead to cellular senescence induction and premature aging.
Collapse
Affiliation(s)
- Jéssica M. Vilas
- Laboratorio de Células Madre en Cáncer y Envejecimiento, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS); Santiago de Compostela Spain
| | - Carmen Carneiro
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Universidade de Santiago de Compostela; Santiago de Compostela Spain
| | - Sabela Da Silva-Álvarez
- Laboratorio de Células Madre en Cáncer y Envejecimiento, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS); Santiago de Compostela Spain
| | - Alba Ferreirós
- Laboratorio de Células Madre en Cáncer y Envejecimiento, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS); Santiago de Compostela Spain
| | - Patricia González
- Histopathology Core Unit; Spanish National Cancer Research Centre (CNIO); Madrid Spain
| | - María Gómez
- Histopathology Core Unit; Spanish National Cancer Research Centre (CNIO); Madrid Spain
| | - Sagrario Ortega
- Trasgenic Mice Unit; Spanish National Cancer Research Centre (CNIO); Madrid Spain
| | - Manuel Serrano
- Tumor Suppression Group; Spanish National Cancer Research Centre (CNIO); Madrid Spain
- Institute for Research in Biomedicine (IRB Barcelona); The Barcelona Institute of Science and Technology (BIST); Barcelona Spain
- Catalan Institution for Research and Advanced Studies (ICREA); Barcelona Spain
| | - Tomás García-Caballero
- Departamento de Ciencias Morfológicas, Facultad de Medicina; USC, Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS); Santiago de Compostela Spain
| | - Miguel González-Barcia
- Servicio de Farmacia; Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS); Santiago de Compostela Spain
| | - Anxo Vidal
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Universidade de Santiago de Compostela; Santiago de Compostela Spain
| | - Manuel Collado
- Laboratorio de Células Madre en Cáncer y Envejecimiento, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS); Santiago de Compostela Spain
| |
Collapse
|