1
|
Burnik T, Zupan J, Jeras M, Kandušer M. Fusion of Human Synovium-Derived Mesenchymal Stem/Stromal Cells with Primary Human Chondrocytes Using the Modified Adherence Method (MAM). Methods Mol Biol 2025. [PMID: 40106149 DOI: 10.1007/7651_2025_620] [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: 03/22/2025]
Abstract
Cell fusion is a complex phenomenon that is key in maintaining tissue homeostasis, particularly in aiding tissue regeneration processes. Studies show that mesenchymal stem/stromal cells (MSCs) are capable of restoring damaged tissue by adopting the phenotype of various cell types via cell fusion. As cell fusion of MSCs with cells of different origin remains poorly researched, we have developed a protocol that allows successful electrofusion between human synovium-derived MSCs and human chondrocytes. Building on from our protocol can help researchers study the cell fusion processes in the in vitro environment and could set basis for development of fusion cell-based advanced therapy medicinal products (ATMPs). In our protocol, we provide a detailed description on how to culture both of the fusion partner cells, how to carry out the modified adherence method (MAM) to achieve a high yield of successfully fused cells, and how to determine the yield of cell fusion using either methyl violet or fluorescent cell trackers.
Collapse
Affiliation(s)
- Tilen Burnik
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
- Institute of Pharmacy, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Janja Zupan
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Matjaž Jeras
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Maša Kandušer
- Institute of Pharmacy, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
2
|
Shabo I, Midtbö K, Bränström R, Lindström A. Monocyte-cancer cell fusion is mediated by phosphatidylserine-CD36 receptor interaction and induced by ionizing radiation. PLoS One 2025; 20:e0311027. [PMID: 39752516 PMCID: PMC11698428 DOI: 10.1371/journal.pone.0311027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 09/11/2024] [Indexed: 01/06/2025] Open
Abstract
Emerging evidence suggests that fusion of cancer cells with leucocytes, such as macrophages, plays a significant role in cancer metastasis and results in tumor hybrid cells that acquire resistance to chemo- and radiation therapy. However, the precise mechanisms behind the leukocyte-cancer cell fusion remain unclear. The present in vitro study explores the presence of fusion between the monocyte cell line (THP-1) and the breast cancer cell line (MCF-7) in relation to the expression of CD36 and phosphatidylserine with and without treatment of these cells with ionizing radiation. The study reveals that spontaneous THP-1/MCF-7 cell fusion increases significantly from 2.8% to 6% after irradiation. The interaction between CD36 and phosphatidylserine plays a pivotal role in THP-1/MCF-7 cell fusion, as inhibiting this interaction using anti-CD36 antibodies significantly reduces cell fusion. While irradiation leads to a dose-dependent escalation in phosphatidylserine expression in MCF-7 cells, it does not impact the expression of CD36 in either THP-1 or MCF-7 cells. To the best of our knowledge, this is the first study to demonstrate the involvement of the CD36-phosphatidylserine interaction in the fusion between monocytes and cancer cells, shedding light on a novel explanatory mechanism for the roles of CD36 and phosphatidylserine in tumor progression.
Collapse
Affiliation(s)
- Ivan Shabo
- Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Breast Cancer, Sarcoma and Endocrine Tumors, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Kristine Midtbö
- Division of Cell- and Neurobiology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Robert Bränström
- Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Breast Cancer, Sarcoma and Endocrine Tumors, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Annelie Lindström
- Division of Cell- and Neurobiology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
3
|
Dhanjal DS, Singh R, Sharma V, Nepovimova E, Adam V, Kuca K, Chopra C. Advances in Genetic Reprogramming: Prospects from Developmental Biology to Regenerative Medicine. Curr Med Chem 2024; 31:1646-1690. [PMID: 37138422 DOI: 10.2174/0929867330666230503144619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 05/05/2023]
Abstract
The foundations of cell reprogramming were laid by Yamanaka and co-workers, who showed that somatic cells can be reprogrammed into pluripotent cells (induced pluripotency). Since this discovery, the field of regenerative medicine has seen advancements. For example, because they can differentiate into multiple cell types, pluripotent stem cells are considered vital components in regenerative medicine aimed at the functional restoration of damaged tissue. Despite years of research, both replacement and restoration of failed organs/ tissues have remained elusive scientific feats. However, with the inception of cell engineering and nuclear reprogramming, useful solutions have been identified to counter the need for compatible and sustainable organs. By combining the science underlying genetic engineering and nuclear reprogramming with regenerative medicine, scientists have engineered cells to make gene and stem cell therapies applicable and effective. These approaches have enabled the targeting of various pathways to reprogramme cells, i.e., make them behave in beneficial ways in a patient-specific manner. Technological advancements have clearly supported the concept and realization of regenerative medicine. Genetic engineering is used for tissue engineering and nuclear reprogramming and has led to advances in regenerative medicine. Targeted therapies and replacement of traumatized , damaged, or aged organs can be realized through genetic engineering. Furthermore, the success of these therapies has been validated through thousands of clinical trials. Scientists are currently evaluating induced tissue-specific stem cells (iTSCs), which may lead to tumour-free applications of pluripotency induction. In this review, we present state-of-the-art genetic engineering that has been used in regenerative medicine. We also focus on ways that genetic engineering and nuclear reprogramming have transformed regenerative medicine and have become unique therapeutic niches.
Collapse
Affiliation(s)
- Daljeet Singh Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Reena Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Varun Sharma
- Head of Bioinformatic Division, NMC Genetics India Pvt. Ltd., Gurugram, India
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno, CZ 613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, Brno, CZ-612 00, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, 50005, Czech Republic
| | - Chirag Chopra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
4
|
Abstract
Electroporation (EP) is a commonly used strategy to increase cell permeability for intracellular cargo delivery or irreversible cell membrane disruption using electric fields. In recent years, EP performance has been improved by shrinking electrodes and device structures to the microscale. Integration with microfluidics has led to the design of devices performing static EP, where cells are fixed in a defined region, or continuous EP, where cells constantly pass through the device. Each device type performs superior to conventional, macroscale EP devices while providing additional advantages in precision manipulation (static EP) and increased throughput (continuous EP). Microscale EP is gentle on cells and has enabled more sensitive assaying of cells with novel applications. In this Review, we present the physical principles of microscale EP devices and examine design trends in recent years. In addition, we discuss the use of reversible and irreversible EP in the development of therapeutics and analysis of intracellular contents, among other noteworthy applications. This Review aims to inform and encourage scientists and engineers to expand the use of efficient and versatile microscale EP technologies.
Collapse
Affiliation(s)
- Sung-Eun Choi
- Department of Mechanical Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Harrison Khoo
- Department of Mechanical Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Soojung Claire Hur
- Department of Mechanical Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- Institute for NanoBioTechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- Department of Oncology, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 401 North Broadway, Baltimore, Maryland 21231, United States
| |
Collapse
|
5
|
Cwykiel J, Jundzill A, Klimczak A, Madajka-Niemeyer M, Siemionow M. Donor Recipient Chimeric Cells Induce Chimerism and Extend Survival of Vascularized Composite Allografts. Arch Immunol Ther Exp (Warsz) 2021; 69:13. [PMID: 33970329 PMCID: PMC8110509 DOI: 10.1007/s00005-021-00614-9] [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: 02/13/2021] [Accepted: 04/08/2021] [Indexed: 11/30/2022]
Abstract
This study evaluated the efficacy of donor recipient chimeric cell (DRCC) therapy created by fusion of donor and recipient derived bone marrow cells (BMC) in chimerism and tolerance induction in a rat vascularized composite allograft (VCA) model. Twenty-four VCA (groin flaps) from MHC-mismatched ACI (RT1a) donors were transplanted to Lewis (RT1l) recipients. Rats were randomly divided into (n = 6/group): Group 1—untreated controls, Groups 2—7-day immunosuppression controls, Group 3—DRCC, and Group 4—DRCC with 7-day anti-αβTCR monoclonal antibody and cyclosporine A protocol. DRCC created by polyethylene glycol-mediated fusion of ACI and Lewis BMC were cultured and transplanted (2–4 × 106) to VCA recipients via intraosseous delivery route. Flow cytometry assessed peripheral blood chimerism while fluorescent microscopy and PCR tested the presence of DRCC in the recipient’s blood, bone marrow (BM), and lymphoid organs at the study endpoint (VCA rejection). No complications were observed after DRCC intraosseous delivery. Group 4 presented the longest average VCA survival (79.3 ± 30.9 days) followed by Group 2 (53.3 ± 13.6 days), Group 3 (18 ± 7.5 days), and Group 1 (8.5 ± 1 days). The highest chimerism level was detected in Group 4 (57.9 ± 6.2%) at day 7 post-transplant. The chimerism declined at day 21 post-transplant and remained at 10% level during the entire follow-up period. Single dose of DRCC therapy induced long-term multilineage chimerism and extended VCA survival. DRCC introduces a novel concept of customized donor-recipient cell-based therapy supporting solid organ and VCA transplants.
Collapse
Affiliation(s)
- Joanna Cwykiel
- Department of Orthopaedics, University of Illinois At Chicago, Molecular Biology Research Building, 900 S. Ashland Ave. Room# 3356, Chicago, IL, 60607, USA.,Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Arkadiusz Jundzill
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA.,Chair of Urology, Department of Regenerative Medicine, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Bydgoszcz, Poland.,Department of Plastic, Reconstructive and Aesthetic Surgery, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Aleksandra Klimczak
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA.,Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | | | - Maria Siemionow
- Department of Orthopaedics, University of Illinois At Chicago, Molecular Biology Research Building, 900 S. Ashland Ave. Room# 3356, Chicago, IL, 60607, USA. .,Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA. .,Department of Surgery, Poznan University of Medical Sciences, Poznan, Poland.
| |
Collapse
|
6
|
Cwykiel J, Madajka-Niemeyer M, Siemionow M. Development of Donor Recipient Chimeric Cells of bone marrow origin as a novel approach for tolerance induction in transplantation. Stem Cell Investig 2021; 8:8. [PMID: 33969113 DOI: 10.21037/sci-2020-044] [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: 10/29/2020] [Accepted: 03/03/2021] [Indexed: 12/14/2022]
Abstract
Background Cell therapies and chimerism-based strategies are currently the most successful approach for tolerance induction in transplantation. This study aimed to establish and characterize novel Donor Recipient Chimeric Ccell (DRCC) therapy of bone marrow (BM) origin presenting donor-recipient phenotype to support tolerance induction. Methods Ex vivo fusions of fully MHC-mismatched BM cells from ACI (RT1a) and Lewis (RT1l) rats were performed using polyethylene-glycol (PEG). The creation of rat DRCC was tested by flow cytometry (FC), confocal microscopy and PCR. FC characterized DRCC's phenotype (CD3, CD4, CD8, CD45, CD90, CD11b/c, CD45RA, OX-82, or CD4/CD25) and apoptosis, while mixed lymphocyte reaction assessed DRCC's immunogenicity and colony forming unit assay tested DRCC's differentiation and proliferation. DRCC's polyploidy was evaluated using Hoechst33342 staining and COMET assay tested genotoxicity of fusion procedure. ELISA analyzed the secretion of IL-2, IL-4, IL-10, TGFß1, IFNγ and TNFα by DRCC at day 1, 5 and 14 post-fusion. The DRCC's phenotype after long-term culturing was assessed by reverse-transcription PCR. Results The chimeric state of DRCC was confirmed. Fusion did not change the expression of hematopoietic markers compared to BM controls. Although an increased number of early and late apoptotic (Annexin V+/Sytox blue- and Annexin V+/Sytox blue+, respectively) DRCC was detected at 24h post-fusion, the number significantly decreased at day 5 (38.4%±3.1% and 22.6%±2.5%, vs. 28.3%±2.5% and 13.9%±2.6%, respectively, P<0.05). DRCC presented decreased immunogenicity, increased expression of IL-10 and TGFβ1 and proliferative potential comparable to BM controls. The average percentage of tetraploid DRCC was 3.1%±0.2% compared to 0.96%±0.1% in BM controls. The lack of damage to the DRCC's DNA content supported the DRCC's safety. In culture, DRCC maintained proliferation for up to 28 days while preserving hematopoietic profile. Conclusions This study confirmed feasibility of DRCC creation via ex vivo PEG mediated fusion. The created DRCC revealed pro-tolerogenic properties indicating potential immunomodulatory effect of DRCC therapy when applied in vivo to support tolerance induction in solid organ and vascularized composite allograft transplantation.
Collapse
Affiliation(s)
- Joanna Cwykiel
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, USA.,Department of Plastic Surgery, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Maria Siemionow
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, USA.,Department of Plastic Surgery, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Surgery, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
7
|
Mukaida N, Tanabe Y, Baba T. Cancer non-stem cells as a potent regulator of tumor microenvironment: a lesson from chronic myeloid leukemia. MOLECULAR BIOMEDICINE 2021; 2:7. [PMID: 35006395 PMCID: PMC8607377 DOI: 10.1186/s43556-021-00030-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/17/2021] [Indexed: 01/10/2023] Open
Abstract
A limited subset of human leukemia cells has a self-renewal capacity and can propagate leukemia upon their transplantation into animals, and therefore, are named as leukemia stem cells, in the early 1990's. Subsequently, cell subpopulations with similar characteristics were detected in various kinds of solid cancers and were denoted as cancer stem cells. Cancer stem cells are presently presumed to be crucially involved in malignant progression of solid cancer: chemoresitance, radioresistance, immune evasion, and metastasis. On the contrary, less attention has been paid to cancer non-stem cell population, which comprise most cancer cells in cancer tissues, due to the lack of suitable markers to discriminate cancer non-stem cells from cancer stem cells. Chronic myeloid leukemia stem cells generate a larger number of morphologically distinct non-stem cells. Moreover, accumulating evidence indicates that poor prognosis is associated with the increases in these non-stem cells including basophils and megakaryocytes. We will discuss the potential roles of cancer non-stem cells in fostering tumor microenvironment, by illustrating the roles of chronic myeloid leukemia non-stem cells including basophils and megakaryocytes in the pathogenesis of chronic myeloid leukemia, a typical malignant disorder arising from leukemic stem cells.
Collapse
Affiliation(s)
- Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan.
| | - Yamato Tanabe
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Tomohisa Baba
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| |
Collapse
|
8
|
Jemni-Damer N, Guedan-Duran A, Fuentes-Andion M, Serrano-Bengoechea N, Alfageme-Lopez N, Armada-Maresca F, Guinea GV, Perez-Rigueiro J, Rojo F, Gonzalez-Nieto D, Kaplan DL, Panetsos F. Biotechnology and Biomaterial-Based Therapeutic Strategies for Age-Related Macular Degeneration. Part II: Cell and Tissue Engineering Therapies. Front Bioeng Biotechnol 2020; 8:588014. [PMID: 33363125 PMCID: PMC7758210 DOI: 10.3389/fbioe.2020.588014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
Age-related Macular Degeneration (AMD) is an up-to-date untreatable chronic neurodegenerative eye disease of multifactorial origin, and the main causes of blindness in over 65 y.o. people. It is characterized by a slow progression and the presence of a multitude of factors, highlighting those related to diet, genetic heritage and environmental conditions, present throughout each of the stages of the illness. Current therapeutic approaches, mainly consisting on intraocular drug delivery, are only used for symptoms relief and/or to decelerate the progression of the disease. Furthermore, they are overly simplistic and ignore the complexity of the disease and the enormous differences in the symptomatology between patients. Due to the wide impact of the AMD and the up-to-date absence of clinical solutions, Due to the wide impact of the AMD and the up-to-date absence of clinical solutions, different treatment options have to be considered. Cell therapy is a very promising alternative to drug-based approaches for AMD treatment. Cells delivered to the affected tissue as a suspension have shown poor retention and low survival rate. A solution to these inconveniences has been the encapsulation of these cells on biomaterials, which contrive to their protection, gives them support, and favor their retention of the desired area. We offer a two-papers critical review of the available and under development AMD therapeutic approaches, from a biomaterials and biotechnological point of view. We highlight benefits and limitations and we forecast forthcoming alternatives based on novel biomaterials and biotechnology methods. In this second part we review the preclinical and clinical cell-replacement approaches aiming at the development of efficient AMD-therapies, the employed cell types, as well as the cell-encapsulation and cell-implant systems. We discuss their advantages and disadvantages and how they could improve the survival and integration of the implanted cells.
Collapse
Affiliation(s)
- Nahla Jemni-Damer
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
| | - Atocha Guedan-Duran
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - María Fuentes-Andion
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
| | - Nora Serrano-Bengoechea
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
- Silk Biomed SL, Madrid, Spain
| | - Nuria Alfageme-Lopez
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
- Silk Biomed SL, Madrid, Spain
| | | | - Gustavo V. Guinea
- Silk Biomed SL, Madrid, Spain
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcon, Spain
- Department of Material Science, Civil Engineering Superior School, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - José Perez-Rigueiro
- Silk Biomed SL, Madrid, Spain
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcon, Spain
- Department of Material Science, Civil Engineering Superior School, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - Francisco Rojo
- Silk Biomed SL, Madrid, Spain
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcon, Spain
- Department of Material Science, Civil Engineering Superior School, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - Daniel Gonzalez-Nieto
- Silk Biomed SL, Madrid, Spain
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcon, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Fivos Panetsos
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
- Silk Biomed SL, Madrid, Spain
| |
Collapse
|
9
|
Abstract
In humans, various genetic defects or age-related diseases, such as diabetic retinopathies, glaucoma, and macular degeneration, cause the death of retinal neurons and profound vision loss. One approach to treating these diseases is to utilize stem and progenitor cells to replace neurons in situ, with the expectation that new neurons will create new synaptic circuits or integrate into existing ones. Reprogramming non-neuronal cells in vivo into stem or progenitor cells is one strategy for replacing lost neurons. Zebrafish have become a valuable model for investigating cellular reprogramming and retinal regeneration. This review summarizes our current knowledge regarding spontaneous reprogramming of Müller glia in zebrafish and compares this knowledge to research efforts directed toward reprogramming Müller glia in mammals. Intensive research using these animal models has revealed shared molecular mechanisms that make Müller glia attractive targets for cellular reprogramming and highlighted the potential for curing degenerative retinal diseases from intrinsic cellular sources.
Collapse
Affiliation(s)
- Manuela Lahne
- Center for Zebrafish Research, Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, USA; , .,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Mikiko Nagashima
- Department of Ophthalmology and Visual Sciences, University of Michigan School of Medicine, Ann Arbor, Michigan 48105, USA; ,
| | - David R Hyde
- Center for Zebrafish Research, Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, USA; , .,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Peter F Hitchcock
- Department of Ophthalmology and Visual Sciences, University of Michigan School of Medicine, Ann Arbor, Michigan 48105, USA; , .,Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, Michigan 48105, USA
| |
Collapse
|
10
|
Singh MS, Park SS, Albini TA, Canto-Soler MV, Klassen H, MacLaren RE, Takahashi M, Nagiel A, Schwartz SD, Bharti K. Retinal stem cell transplantation: Balancing safety and potential. Prog Retin Eye Res 2020; 75:100779. [PMID: 31494256 PMCID: PMC7056514 DOI: 10.1016/j.preteyeres.2019.100779] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 08/29/2019] [Accepted: 09/02/2019] [Indexed: 12/14/2022]
Abstract
Stem cell transplantation holds great promise as a potential treatment for currently incurable retinal degenerative diseases that cause poor vision and blindness. Recently, safety data have emerged from several Phase I/II clinical trials of retinal stem cell transplantation. These clinical trials, usually run in partnership with academic institutions, are based on sound preclinical studies and are focused on patient safety. However, reports of serious adverse events arising from cell therapy in other poorly regulated centers have now emerged in the lay and scientific press. While progress in stem cell research for blindness has been greeted with great enthusiasm by patients, scientists, doctors and industry alike, these adverse events have raised concerns about the safety of retinal stem cell transplantation and whether patients are truly protected from undue harm. The aim of this review is to summarize and appraise the safety of human retinal stem cell transplantation in the context of its potential to be developed into an effective treatment for retinal degenerative diseases.
Collapse
Affiliation(s)
- Mandeep S Singh
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Susanna S Park
- Department of Ophthalmology & Vision Science, University of California-Davis Eye Center, Sacramento, CA, 95817, USA
| | - Thomas A Albini
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - M Valeria Canto-Soler
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Henry Klassen
- Gavin Herbert Eye Institute and Stem Cell Research Center, Irvine, CA, 92697, USA
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford and Oxford University Eye Hospital, NHS Foundation Trust, NIHR Biomedical Research Centre, Oxford, OX3 9DU, UK
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, Kobe, Hyogo, 650-0047, Japan
| | - Aaron Nagiel
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA; USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90007, USA
| | - Steven D Schwartz
- Stein Eye Institute, University of California Los Angeles Geffen School of Medicine, Los Angeles, CA, 90095, USA; Edythe and Eli Broad Stem Cell Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Kapil Bharti
- National Eye Institute, National Institutes of Health, Bethesda, MD, 90892, USA
| |
Collapse
|
11
|
Rong L, Gu X, Xie J, Zeng Y, Li Q, Chen S, Zou T, Xue L, Xu H, Yin ZQ. Bone Marrow CD133 + Stem Cells Ameliorate Visual Dysfunction in Streptozotocin-induced Diabetic Mice with Early Diabetic Retinopathy. Cell Transplant 2018; 27:916-936. [PMID: 29717657 PMCID: PMC6050916 DOI: 10.1177/0963689718759463] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/15/2018] [Accepted: 01/22/2018] [Indexed: 12/16/2022] Open
Abstract
Diabetic retinopathy (DR), one of the leading causes of vision loss worldwide, is characterized by neurovascular disorders. Emerging evidence has demonstrated retinal neurodegeneration in the early pathogenesis of DR, and no treatment has been developed to prevent the early neurodegenerative changes that precede detectable microvascular disorders. Bone marrow CD133+ stem cells with revascularization properties exhibit neuroregenerative potential. However, whether CD133+ cells can ameliorate the neurodegeneration at the early stage of DR remains unclear. In this study, mouse bone marrow CD133+ stem cells were immunomagnetically isolated and analyzed for the phenotypic characteristics, capacity for neural differentiation, and gene expression of neurotrophic factors. After being labeled with enhanced green fluorescent protein, CD133+ cells were intravitreally transplanted into streptozotocin (STZ)-induced diabetic mice to assess the outcomes of visual function and retina structure and the mechanism underlying the therapeutic effect. We found that CD133+ cells co-expressed typical hematopoietic/endothelial stem/progenitor phenotypes, could differentiate to neural lineage cells, and expressed genes of robust neurotrophic factors in vitro. Functional analysis demonstrated that the transplantation of CD133+ cells prevented visual dysfunction for 56 days. Histological analysis confirmed such a functional improvement and showed that transplanted CD133+ cells survived, migrated into the inner retina (IR) over time and preserved IR degeneration, including retina ganglion cells (RGCs) and rod-on bipolar cells. In addition, a subset of transplanted CD133+ cells in the ganglion cell layer differentiated to express RGC markers in STZ-induced diabetic retina. Moreover, transplanted CD133+ cells expressed brain-derived neurotrophic factors (BDNFs) in vivo and increased the BDNF level in STZ-induced diabetic retina to support the survival of retinal cells. Based on these findings, we suggest that transplantation of bone marrow CD133+ stem cells represents a novel approach to ameliorate visual dysfunction and the underlying IR neurodegeneration at the early stage of DR.
Collapse
Affiliation(s)
- Liyuan Rong
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical
University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing,
Chongqing, China
| | - Xianliang Gu
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical
University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing,
Chongqing, China
| | - Jing Xie
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical
University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing,
Chongqing, China
| | - Yuxiao Zeng
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical
University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing,
Chongqing, China
| | - Qiyou Li
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical
University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing,
Chongqing, China
| | - Siyu Chen
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical
University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing,
Chongqing, China
| | - Ting Zou
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical
University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing,
Chongqing, China
| | - Langyue Xue
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical
University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing,
Chongqing, China
| | - Haiwei Xu
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical
University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing,
Chongqing, China
| | - Zheng Qin Yin
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical
University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing,
Chongqing, China
| |
Collapse
|
12
|
The Pleiotropic Effects of the Canonical Wnt Pathway in Early Development and Pluripotency. Genes (Basel) 2018; 9:genes9020093. [PMID: 29443926 PMCID: PMC5852589 DOI: 10.3390/genes9020093] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/30/2018] [Accepted: 01/30/2018] [Indexed: 12/20/2022] Open
Abstract
The technology to derive embryonic and induced pluripotent stem cells from early embryonic stages and adult somatic cells, respectively, emerged as a powerful resource to enable the establishment of new in vitro models, which recapitulate early developmental processes and disease. Additionally, pluripotent stem cells (PSCs) represent an invaluable source of relevant differentiated cell types with immense potential for regenerative medicine and cell replacement therapies. Pluripotent stem cells support self-renewal, potency and proliferation for extensive periods of culture in vitro. However, the core pathways that rule each of these cellular features specific to PSCs only recently began to be clarified. The Wnt signaling pathway is pivotal during early embryogenesis and is central for the induction and maintenance of the pluripotency of PSCs. Signaling by the Wnt family of ligands is conveyed intracellularly by the stabilization of β-catenin in the cytoplasm and in the nucleus, where it elicits the transcriptional activity of T-cell factor (TCF)/lymphoid enhancer factor (LEF) family of transcription factors. Interestingly, in PSCs, the Wnt/β-catenin–TCF/LEF axis has several unrelated and sometimes opposite cellular functions such as self-renewal, stemness, lineage commitment and cell cycle regulation. In addition, tight control of the Wnt signaling pathway enhances reprogramming of somatic cells to induced pluripotency. Several recent research efforts emphasize the pleiotropic functions of the Wnt signaling pathway in the pluripotent state. Nonetheless, conflicting results and unanswered questions still linger. In this review, we will focus on the diverse functions of the canonical Wnt signaling pathway on the developmental processes preceding embryo implantation, as well as on its roles in pluripotent stem cell biology such as self-renewal and cell cycle regulation and somatic cell reprogramming.
Collapse
|
13
|
Hypoxia Enhances Fusion of Oral Squamous Carcinoma Cells and Epithelial Cells Partly via the Epithelial-Mesenchymal Transition of Epithelial Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5015203. [PMID: 29581976 PMCID: PMC5822897 DOI: 10.1155/2018/5015203] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/03/2018] [Indexed: 01/06/2023]
Abstract
Increasing evidence and indications showed that cell fusion is crucial in tumor development and metastasis, and hypoxia, a closely linked factor to tumor microenvironment, which can lead to EMT, induces angiogenesis and metastasis in tumor growth. However, the relationship between hypoxia and fusion has not been reported yet. EMT will change some proteins in the epithelial cell surface and the changes of proteins in cell surface may increase cell fusion. This study found that hypoxia promotes the spontaneous cell fusion between Oral Squamous Carcinoma Cells (OSCCs) and Human Immortalized Oral Epithelial Cells (HIOECs). At the same time, Hypoxia can lead to EMT, and hypoxia-pretreated HIOECs increased fusion rate with OSCC, while the fusion rate was significantly reduced by DAPT, a kind of EMT blocker. Therefore, epithelial cells can increase spontaneously cell fusion with OSCC by EMT. Our study may provide a new insight to link among tumor microenvironment, cell fusion, and cancer.
Collapse
|
14
|
Pedone E, Olteanu VA, Marucci L, Muñoz-Martin MI, Youssef SA, de Bruin A, Cosma MP. Modeling Dynamics and Function of Bone Marrow Cells in Mouse Liver Regeneration. Cell Rep 2017; 18:107-121. [PMID: 28052241 PMCID: PMC5236012 DOI: 10.1016/j.celrep.2016.12.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/15/2016] [Accepted: 12/01/2016] [Indexed: 12/13/2022] Open
Abstract
In rodents and humans, the liver can efficiently restore its mass after hepatectomy. This is largely attributed to the proliferation and cell cycle re-entry of hepatocytes. On the other hand, bone marrow cells (BMCs) migrate into the liver after resection. Here, we find that a block of BMC recruitment into the liver severely impairs its regeneration after the surgery. Mobilized hematopoietic stem and progenitor cells (HSPCs) in the resected liver can fuse with hepatocytes, and the hybrids proliferate earlier than the hepatocytes. Genetic ablation of the hybrids severely impairs hepatocyte proliferation and liver mass regeneration. Mathematical modeling reveals a key role of bone marrow (BM)-derived hybrids to drive proliferation in the regeneration process, and predicts regeneration efficiency in experimentally non-testable conditions. In conclusion, BM-derived hybrids are essential to trigger efficient liver regeneration after hepatectomy. Bone marrow cell migration after liver hepatectomy is key for liver regeneration Migrated bone marrow cells fuse with hepatocytes Hybrids are essential for liver regeneration Mathematical modeling unveils the hybrid function for liver regeneration
Collapse
Affiliation(s)
- Elisa Pedone
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Dr Aiguader 88, 08003 Barcelona, Spain
| | - Vlad-Aris Olteanu
- Department of Engineering Mathematics, University of Bristol, Bristol BS8 1UB, UK
| | - Lucia Marucci
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, 08003 Barcelona, Spain; Department of Engineering Mathematics, University of Bristol, Bristol BS8 1UB, UK.
| | - Maria Isabel Muñoz-Martin
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, 08003 Barcelona, Spain
| | - Sameh A Youssef
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, 3584 Utrecht, the Netherlands; Department of Pathology, Alexandria Veterinary College, University of Alexandria-Egypt, 21612 Alexandria, Egypt
| | - Alain de Bruin
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, 3584 Utrecht, the Netherlands; University Medical Center Groningen, Department of Pediatrics, University of Groningen, 9713 Groningen, the Netherlands
| | - Maria Pia Cosma
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Dr Aiguader 88, 08003 Barcelona, Spain; ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|
15
|
Dudin O, Merlini L, Martin SG. Spatial focalization of pheromone/MAPK signaling triggers commitment to cell-cell fusion. Genes Dev 2017; 30:2226-2239. [PMID: 27798845 PMCID: PMC5088570 DOI: 10.1101/gad.286922.116] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/21/2016] [Indexed: 12/14/2022]
Abstract
Here, Dudin et al. show that cell fusion does not require a dedicated signal but is triggered by spatial focalization of the same pheromone–GPCR–MAPK signaling cascade that drives earlier mating events in Schizosaccharomyces pombe. Cell fusion is universal in eukaryotes for fertilization and development, but what signals this process is unknown. Here, we show in Schizosaccharomyces pombe that fusion does not require a dedicated signal but is triggered by spatial focalization of the same pheromone–GPCR (G-protein-coupled receptor)–MAPK signaling cascade that drives earlier mating events. Autocrine cells expressing the receptor for their own pheromone trigger fusion attempts independently of cell–cell contact by concentrating pheromone release at the fusion focus, a dynamic actin aster underlying the secretion of cell wall hydrolases. Pheromone receptor and MAPK cascade are similarly enriched at the fusion focus, concomitant with fusion commitment in wild-type mating pairs. This focalization promotes cell fusion by immobilizing the fusion focus, thus driving local cell wall dissolution. We propose that fusion commitment is imposed by a local increase in MAPK concentration at the fusion focus, driven by a positive feedback between fusion focus formation and focalization of pheromone release and perception.
Collapse
Affiliation(s)
- Omaya Dudin
- Department of Fundamental Microbiology, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Laura Merlini
- Department of Fundamental Microbiology, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Sophie G Martin
- Department of Fundamental Microbiology, University of Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
16
|
Vetter ML, Hitchcock PF. Report on the National Eye Institute Audacious Goals Initiative: Replacement of Retinal Ganglion Cells from Endogenous Cell Sources. Transl Vis Sci Technol 2017; 6:5. [PMID: 28316878 PMCID: PMC5354473 DOI: 10.1167/tvst.6.2.5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 12/30/2016] [Indexed: 12/22/2022] Open
Abstract
This report emerges from a workshop convened by the National Eye Institute (NEI) as part of the "Audacious Goals Initiative" (AGI). The workshop addressed the replacement of retinal ganglion cells (RGCs) from exogenous and endogenous sources, and sought to identify the gaps in our knowledge and barriers to progress in devising cellular replacement therapies for diseases where RGCs die. Here, we briefly review relevant literature regarding common diseases associated with RGC death, the genesis of RGCs in vivo, strategies for generating transplantable RGCs in vitro, and potential endogenous cellular sources to regenerate these cells. These topics provided the clinical and scientific context for the discussion among the workshop participants and are relevant to efforts that may lead to therapeutic approaches for replacing RGCs. This report also summarizes the content of the workshop discussion, which focused on: (1) cell sources for RGC replacement and regeneration, (2) optimizing integration, survival, and synaptogenesis of new RGCs, and (3) approaches for assessing the outcomes of RGC replacement therapies. We conclude this report with a summary of recommendations, based on the workshop discussions, which may guide vision scientists seeking to develop therapies for replacing RGCs in humans.
Collapse
Affiliation(s)
- Monica L Vetter
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, USA
| | - Peter F Hitchcock
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, USA ; Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
17
|
Yan TL, Wang M, Xu Z, Huang CM, Zhou XC, Jiang EH, Zhao XP, Song Y, Song K, Shao Z, Liu K, Shang ZJ. Up-regulation of syncytin-1 contributes to TNF-α-enhanced fusion between OSCC and HUVECs partly via Wnt/β-catenin-dependent pathway. Sci Rep 2017; 7:40983. [PMID: 28112190 PMCID: PMC5256027 DOI: 10.1038/srep40983] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/13/2016] [Indexed: 12/19/2022] Open
Abstract
Accumulating evidence implies that cell fusion is one of the driving forces of cancer invasion and metastasis. However, considerably less is still known about the triggering factors and underlying mechanisms associated with cancer-host cell fusion, particularly in inflammatory tumor microenvironment. In this study, we confirmed that inflammatory factor TNF-α could enhance fusion between squamous cell carcinoma cells 9 (SCC-9) and human umbilical vein endothelial cells (HUVEC). Further study revealed that TNF-α could promote up-regulation of syncytin-1 in SCC-9 and its receptor neutral amino acid transporter type 2 (ASCT-2) in HUVEC. Syncytin-1 acted as an important downstream effector in TNF-α-enhanced cancer-endothelial cell fusion. TNF-α treatment also led to the activation of Wnt/β-catenin signal pathway in SCC-9. The activation of Wnt/β-catenin signal pathway was closely associated with the up-regulation of syncytin-1 in SCC-9 and increased fusion between SCC-9 and HUVEC while blocking of Wnt/β-catenin signal pathway resulted in the corresponding down-regulation of syncytin-1 accompanied by sharp decrease of cancer-endothelial cell fusion. Taking together, our results suggest that Wnt/β-catenin signal pathway activation-dependent up-regulation of syncytin-1 contributes to the pro-inflammatory factor TNF-α-enhanced fusion between oral squamous cell carcinoma cells and endothelial cells.
Collapse
Affiliation(s)
- Ting-Lin Yan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) &Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Meng Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) &Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Zhi Xu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) &Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Chun-Ming Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) &Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Xiao-Cheng Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) &Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Er-Hui Jiang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) &Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Xiao-Ping Zhao
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Song
- Department of Stomatology, Liuzhou People's Hospital, Guangxi, China
| | - Kai Song
- Department of Oral and Maxillofacial Surgery, The Affliated Hospital of Qingdao University, Qingdao, China
| | - Zhe Shao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) &Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China.,Department of Oromaxillofacial &Head NeckOncology, School &Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ke Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) &Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China.,Department of Oromaxillofacial &Head NeckOncology, School &Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zheng-Jun Shang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) &Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China.,Department of Oromaxillofacial &Head NeckOncology, School &Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
18
|
Singh MS, Balmer J, Barnard AR, Aslam SA, Moralli D, Green CM, Barnea-Cramer A, Duncan I, MacLaren RE. Transplanted photoreceptor precursors transfer proteins to host photoreceptors by a mechanism of cytoplasmic fusion. Nat Commun 2016; 7:13537. [PMID: 27901042 PMCID: PMC5141374 DOI: 10.1038/ncomms13537] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/12/2016] [Indexed: 12/12/2022] Open
Abstract
Photoreceptor transplantation is a potential future treatment for blindness caused by retinal degeneration. Photoreceptor transplantation restores visual responses in end-stage retinal degeneration, but has also been assessed in non-degenerate retinas. In the latter scenario, subretinal transplantation places donor cells beneath an intact host outer nuclear layer (ONL) containing host photoreceptors. Here we show that host cells are labelled with the donor marker through cytoplasmic transfer—94±4.1% of apparently well-integrated donor cells containing both donor and host markers. We detect the occurrence of Cre-Lox recombination between donor and host photoreceptors, and we confirm the findings through FISH analysis of X and Y chromosomes in sex-discordant transplants. We do not find evidence of nuclear fusion of donor and host cells. The artefactual appearance of integrated donor cells in host retinas following transplantation is most commonly due to material transfer from donor cells. Understanding this novel mechanism may provide alternate therapeutic strategies at earlier stages of retinal degeneration. Previous studies have used fluorescently labelled cells to demonstrate the incorporation of transplanted photoreceptor precursors into the mouse retina. Here, the authors show that fluorescent proteins are passed between the host and transplanted cells rather than migration of donor cells into the retina.
Collapse
Affiliation(s)
- Mandeep S Singh
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, Levels 5-6 West Wing, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK
| | - Jasmin Balmer
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, Levels 5-6 West Wing, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK
| | - Alun R Barnard
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, Levels 5-6 West Wing, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK
| | - Sher A Aslam
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, Levels 5-6 West Wing, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK.,UK Ministry of Defence Army Medical Services, London SW1A 2HB, UK
| | - Daniela Moralli
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Catherine M Green
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Alona Barnea-Cramer
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, Levels 5-6 West Wing, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK
| | - Isabel Duncan
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, Levels 5-6 West Wing, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, Levels 5-6 West Wing, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK.,UK Ministry of Defence Army Medical Services, London SW1A 2HB, UK.,Moorfields Eye Hospital NHS Foundation Trust, 162 City Road, London EC1V 2PD, UK
| |
Collapse
|
19
|
Mesenchymal stem cells generate distinct functional hybrids in vitro via cell fusion or entosis. Sci Rep 2016; 6:36863. [PMID: 27827439 PMCID: PMC5101832 DOI: 10.1038/srep36863] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/20/2016] [Indexed: 02/06/2023] Open
Abstract
Homotypic and heterotypic cell-to-cell fusion are key processes during development and tissue regeneration. Nevertheless, aberrant cell fusion can contribute to tumour initiation and metastasis. Additionally, a form of cell-in-cell structure called entosis has been observed in several human tumours. Here we investigate cell-to-cell interaction between mouse mesenchymal stem cells (MSCs) and embryonic stem cells (ESCs). MSCs represent an important source of adult stem cells since they have great potential for regenerative medicine, even though they are also involved in cancer progression. We report that MSCs can either fuse forming heterokaryons, or be invaded by ESCs through entosis. While entosis-derived hybrids never share their genomes and induce degradation of the target cell, fusion-derived hybrids can convert into synkaryons. Importantly we show that hetero-to-synkaryon transition occurs through cell division and not by nuclear membrane fusion. Additionally, we also observe that the ROCK-actin/myosin pathway is required for both fusion and entosis in ESCs but only for entosis in MSCs. Overall, we show that MSCs can undergo fusion or entosis in culture by generating distinct functional cellular entities. These two processes are profoundly different and their outcomes should be considered given the beneficial or possible detrimental effects of MSC-based therapeutic applications.
Collapse
|
20
|
Altarche-Xifro W, di Vicino U, Muñoz-Martin MI, Bortolozzi A, Bové J, Vila M, Cosma MP. Functional Rescue of Dopaminergic Neuron Loss in Parkinson's Disease Mice After Transplantation of Hematopoietic Stem and Progenitor Cells. EBioMedicine 2016; 8:83-95. [PMID: 27428421 PMCID: PMC4919540 DOI: 10.1016/j.ebiom.2016.04.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 04/14/2016] [Indexed: 01/24/2023] Open
Abstract
Parkinson's disease is a common neurodegenerative disorder, which is due to the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and for which no definitive cure is currently available. Cellular functions in mouse and human tissues can be restored after fusion of bone marrow (BM)-derived cells with a variety of somatic cells. Here, after transplantation of hematopoietic stem and progenitor cells (HSPCs) in the SNpc of two different mouse models of Parkinson's disease, we significantly ameliorated the dopaminergic neuron loss and function. We show fusion of transplanted HSPCs with neurons and with glial cells in the ventral midbrain of Parkinson's disease mice. Interestingly, the hybrids can undergo reprogramming in vivo and survived up to 4 weeks after transplantation, while acquiring features of mature astroglia. These newly generated astroglia produced Wnt1 and were essential for functional rescue of the dopaminergic neurons. Our data suggest that glial-derived hybrids produced upon fusion of transplanted HSPCs in the SNpc can rescue the Parkinson's disease phenotype via a niche-mediated effect, and can be exploited as an efficient cell-therapy approach. Transplantation of HSPCs into the substantia nigra of PD mice ameliorates dopaminergic neuron loss and function. Hybrids generated after fusion of transplanted HSPCs undergo reprogramming in vivo and acquire features of mature astroglia. Newly generated astroglia produced Wnt1 and can functionally rescue the dopaminergic neuron loss.
A definitive therapy for Parkinson's disease is not available. Here, we transplanted hematopoietic stem and progenitor cells into the substantia nigra of brains of two different mouse models of Parkinson's disease. These transplanted cells fused with neurons and glial cells of the recipient mice. Four weeks after transplantation, the hybrids acquired features of mature astroglia, secreted Wnt1, and functionally ameliorated dopaminergic neuron loss. Current cell therapy approaches are being pursued in the striatum with the aim to increase dopamine levels. Here we show that the loss of dopaminergic neurons can be protected against by direct actions in the substantia nigra.
Collapse
Affiliation(s)
- Wassim Altarche-Xifro
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Dr Aiguader 88, 08003 Barcelona, Spain
| | - Umberto di Vicino
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Dr Aiguader 88, 08003 Barcelona, Spain
| | - Maria Isabel Muñoz-Martin
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Dr Aiguader 88, 08003 Barcelona, Spain
| | - Analía Bortolozzi
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain; Department of Neurochemistry and Neuropharmacology, IIBB-CSIC (Consejo Superior de Investigaciones Científicas), Barcelona, Spain
| | - Jordi Bové
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute and Centre for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Miquel Vila
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute and Centre for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain; Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona (UAB), Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Maria Pia Cosma
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Dr Aiguader 88, 08003 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain.
| |
Collapse
|
21
|
Wang R, Chen S, Li C, Ng KTP, Kong CW, Cheng J, Cheng SH, Li RA, Lo CM, Man K, Sun D. Fusion with stem cell makes the hepatocellular carcinoma cells similar to liver tumor-initiating cells. BMC Cancer 2016; 16:56. [PMID: 26846780 PMCID: PMC4743091 DOI: 10.1186/s12885-016-2094-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 01/29/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cell fusion is a fast and highly efficient technique for cells to acquire new properties. The fusion of somatic cells with stem cells can reprogram somatic cells to a pluripotent state. Our research on the fusion of stem cells and cancer cells demonstrates that the fused cells can exhibit stemness and cancer cell-like characteristics. Thus, tumor-initiating cell-like cells are generated. METHODS We employed laser-induced single-cell fusion technique to fuse the hepatocellular carcinoma cells and human embryonic stem cells (hESC). Real-time RT-PCR, flow cytometry and in vivo tumorigenicity assay were adopted to identify the gene expression difference. RESULTS We successfully produced a fused cell line that coalesces the gene expression information of hepatocellular carcinoma cells and stem cells. Experimental results showed that the fused cells expressed cancer and stemness markers as well as exhibited increased resistance to drug treatment and enhanced tumorigenesis. CONCLUSIONS Fusion with stem cells transforms liver cancer cells into tumor initiating-like cells. Results indicate that fusion between cancer cell and stem cell may generate tumor initiating-like cells.
Collapse
Affiliation(s)
- Ran Wang
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China.
| | - Shuxun Chen
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China.
| | - Changxian Li
- Department of Surgery, Li Ka-Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.
| | - Kevin Tak Pan Ng
- Department of Surgery, Li Ka-Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.
| | - Chi-wing Kong
- Stem Cell and Regenerative Medicine Consortium, and Departments of Medicine and Physiology, Li Ka-Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.
| | - Jinping Cheng
- Environmental Science Program, School of Science, Hong Kong University of Science and Technology, Hong Kong, China.
| | - Shuk Han Cheng
- Department of Biomedical Science, City University of Hong Kong, Hong Kong, China.
| | - Ronald A Li
- Stem Cell and Regenerative Medicine Consortium, and Departments of Medicine and Physiology, Li Ka-Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.
| | - Chung Mau Lo
- Department of Surgery, Li Ka-Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.
| | - Kwan Man
- Department of Surgery, Li Ka-Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.
| | - Dong Sun
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
22
|
Shinn-Thomas JH, del Campo JJ, Wang J, Mohler WA. The EFF-1A Cytoplasmic Domain Influences Hypodermal Cell Fusions in C. elegans But Is Not Dependent on 14-3-3 Proteins. PLoS One 2016; 11:e0146874. [PMID: 26800457 PMCID: PMC4723337 DOI: 10.1371/journal.pone.0146874] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 12/25/2015] [Indexed: 12/19/2022] Open
Abstract
Background Regulatory and biophysical mechanisms of cell-cell fusion are largely unknown despite the fundamental requirement for fused cells in eukaryotic development. Only two cellular fusogens that are not of clear recent viral origin have been identified to date, both in nematodes. One of these, EFF-1, is necessary for most cell fusions in Caenorhabditis elegans. Unregulated EFF-1 expression causes lethality due to ectopic fusion between cells not developmentally programmed to fuse, highlighting the necessity of tight fusogen regulation for proper development. Identifying factors that regulate EFF-1 and its paralog AFF-1 could lead to discovery of molecular mechanisms that control cell fusion upstream of the action of a membrane fusogen. Bioinformatic analysis of the EFF-1A isoform’s predicted cytoplasmic domain (endodomain) previously revealed two motifs that have high probabilities of interacting with 14-3-3 proteins when phosphorylated. Mutation of predicted phosphorylation sites within these motifs caused measurable loss of eff-1 gene function in cell fusion in vivo. Moreover, a human 14-3-3 isoform bound to EFF-1::GFP in vitro. We hypothesized that the two 14-3-3 proteins in C. elegans, PAR-5 and FTT-2, may regulate either localization or fusion-inducing activity of EFF-1. Methodology/Principal Findings Timing of fusion events was slightly but significantly delayed in animals unable to produce full-length EFF-1A. Yet, mutagenesis and live imaging showed that phosphoserines in putative 14-3-3 binding sites are not essential for EFF-1::GFP accumulation at the membrane contact between fusion partner cells. Moreover, although the EFF-1A endodomain was required for normal rates of eff-1-dependent epidermal cell fusions, reduced levels of FTT-2 and PAR-5 did not visibly affect the function of wild-type EFF-1 in the hypodermis. Conclusions/Significance Deletion of the EFF-1A endodomain noticeably affects the timing of hypodermal cell fusions in vivo. However, prohibiting phosphorylation of candidate 14-3-3-binding sites does not impact localization of the fusogen. Hypodermal membrane fusion activity persists when 14-3-3 expression levels are reduced.
Collapse
Affiliation(s)
- Jessica H. Shinn-Thomas
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, MC-6403, 263 Farmington Avenue, Farmington, CT 06030–6403, United States of America
- * E-mail: (WAM); (JHST)
| | - Jacob J. del Campo
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, MC-6403, 263 Farmington Avenue, Farmington, CT 06030–6403, United States of America
| | - Jianjun Wang
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, MC-6403, 263 Farmington Avenue, Farmington, CT 06030–6403, United States of America
| | - William A. Mohler
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, MC-6403, 263 Farmington Avenue, Farmington, CT 06030–6403, United States of America
- * E-mail: (WAM); (JHST)
| |
Collapse
|
23
|
Macrophage and Multinucleated Giant Cell Classification. CURRENT TOPICS IN ENVIRONMENTAL HEALTH AND PREVENTIVE MEDICINE 2016. [DOI: 10.1007/978-4-431-55732-6_1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
24
|
Zeng C, Zhang Y, Park SC, Eun JR, Nguyen NT, Tschudy-Seney B, Jung YJ, Theise ND, Zern MA, Duan Y. CD34(+) Liver Cancer Stem Cells Were Formed by Fusion of Hepatobiliary Stem/Progenitor Cells with Hematopoietic Precursor-Derived Myeloid Intermediates. Stem Cells Dev 2015; 24:2467-78. [PMID: 26192559 DOI: 10.1089/scd.2015.0202] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
A large number of cancer stem cells (CSCs) were identified and characterized; however, the origins and formation of CSCs remain elusive. In this study, we examined the origination of the newly identified CD34(+) liver CSC (LCSC). We found that CD34(+) LCSC coexpressed liver stem cell and myelomonocytic cell markers, showing a mixed phenotype, a combination of hepatobiliary stem/progenitor cells (HSPCs) and myelomonocytic cells. Moreover, human xenografts produced by CD34(+) LCSCs and the parental cells, which CD34(+) LCSC was isolated from, coexpressed liver cancer and myelomonocytic markers, also demonstrating mixed phenotypes. The xenografts and the parental cells secreted albumin demonstrating their hepatocyte origin and also expressed cytokines [interleukin (IL)-1b, IL-6, IL-12A, IL-18, tumor necrosis factor-alpha (TNF-α), and CSF1] and chemokines (IL-8, CCL2, and CCL5). Expression of these cytokines and chemokines responded to the stimuli [interferon-γ (INF-γ), IL-4, and lipopolysaccharide (LPS)]. Furthermore, human xenografts and the parental cells phagocytized Escherichia coli. CD34(+) LCSC coexpressed CD45, demonstrating that its origin appears to be from a hematopoietic precursor. The percentage of cells positive for OV6, CD34, and CD31, presenting the markers of HSPC, hematopoietic, and myelomonocytic cells, increased under treatment of CD34(+) LCSC with a drug. Cytogenetic analysis showed that CD34(+) LCSC contained a greater number of chromosomes. HBV DNA integrations and mutations in CD34(+) LCSC and the parental cells were identical to those in the literature or the database. Thus, these results demonstrated that CD34(+) LCSCs were formed by fusion of HSPC with CD34(+) hematopoietic precursor-derived myeloid intermediates; it appears that this is the first report that human CSCs have been formed by the fusion. Therefore, it represents a significant step toward better understanding of the formation of human CSC and the diverse origins of liver cancers.
Collapse
Affiliation(s)
- Changjun Zeng
- 1 Department of Internal Medicine, University of California Davis Medical Center , Sacramento, California.,2 Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California.,3 College of Animal Science and Technology, Sichuan Agricultural University , Ya'an, China
| | - Yanling Zhang
- 1 Department of Internal Medicine, University of California Davis Medical Center , Sacramento, California.,2 Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California.,4 School of Biotechnology, Southern Medical University , Guangzhou, China
| | - Su Cheol Park
- 1 Department of Internal Medicine, University of California Davis Medical Center , Sacramento, California.,2 Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California.,5 Department of Internal Medicine, Korea Institute of Radiological & Medical Sciences , Seoul, Korea
| | - Jong Ryeol Eun
- 1 Department of Internal Medicine, University of California Davis Medical Center , Sacramento, California.,2 Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California.,6 Department of Internal Medicine, Yeungnam University College of Medicine , Daegu, Korea
| | - Ngoc Tue Nguyen
- 1 Department of Internal Medicine, University of California Davis Medical Center , Sacramento, California.,2 Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California
| | - Benjamin Tschudy-Seney
- 1 Department of Internal Medicine, University of California Davis Medical Center , Sacramento, California.,2 Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California
| | - Yong Jin Jung
- 1 Department of Internal Medicine, University of California Davis Medical Center , Sacramento, California.,2 Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California.,7 Department of Internal Medicine, Seoul National University College of Medicine , Seoul, Korea
| | - Neil D Theise
- 8 Department of Pathology and Medicine, Beth Israel Medical Center , Albert Einstein College of Medicine, New York, New York
| | - Mark A Zern
- 1 Department of Internal Medicine, University of California Davis Medical Center , Sacramento, California.,2 Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California
| | - Yuyou Duan
- 1 Department of Internal Medicine, University of California Davis Medical Center , Sacramento, California.,2 Institute for Regenerative Cures, University of California Davis Medical Center , Sacramento, California.,9 Department of Dermatology, University of California Davis Medical Center , Sacramento, California
| |
Collapse
|
25
|
Wei HJ, Nickoloff JA, Chen WH, Liu HY, Lo WC, Chang YT, Yang PC, Wu CW, Williams DF, Gelovani JG, Deng WP. FOXF1 mediates mesenchymal stem cell fusion-induced reprogramming of lung cancer cells. Oncotarget 2015; 5:9514-29. [PMID: 25237908 PMCID: PMC4253450 DOI: 10.18632/oncotarget.2413] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Several reports suggest that malignant cells generate phenotypic diversity through fusion with various types of stromal cells within the tumor microenvironment. Mesenchymal stem cell (MSC) is one of the critical components in the tumor microenvironment and a promising fusogenic candidate, but the underlying functions of MSC fusion with malignant cell have not been fully examined. Here, we demonstrate that MSCs fuse spontaneously with lung cancer cells, and the latter is reprogrammed to slow growth and stem-like state. Transcriptome profiles reveal that lung cancer cells are reprogrammed to a more benign state upon MSC fusion. We further identified FOXF1 as a reprogramming mediator that contributes not only to the reprogramming toward stemness but also to the p21-regulated growth suppression in fusion progeny. Collectively, MSC fusion does not enhance the intrinsic malignancy of lung cancer cells. The anti-malignant effects of MSC fusion-induced reprogramming on lung cancer cells were accomplished by complementation of tumorigenic defects, including restoration of p21 function and normal terminal differentiation pathways as well as up-regulation of FOXF1, a putative tumor suppressor. Such fusion process raises the therapeutic potential that MSC fusion can be utilized to reverse cellular phenotypes in cancer.
Collapse
Affiliation(s)
- Hong-Jian Wei
- Graduate Institute of Biomedical Materials and Engineering, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan. Stem Cell Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Jac A Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Wei-Hong Chen
- Stem Cell Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Hen-Yu Liu
- Stem Cell Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Cheng Lo
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 110, Taiwan. School of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Ya-Ting Chang
- Graduate Institute of Biomedical Materials and Engineering, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Pan-Chyr Yang
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Cheng-Wen Wu
- Institute of Biochemistry and Molecular Biology, National Yang Ming University, Taipei 112, Taiwan
| | - David F Williams
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC 27157, USA
| | - Juri G Gelovani
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI 48201, USA Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Win-Ping Deng
- Graduate Institute of Biomedical Materials and Engineering, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan. Stem Cell Research Center, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
26
|
Parris GE. Cell-Cell Fusion, Chemotaxis and Metastasis. INTERCELLULAR COMMUNICATION IN CANCER 2015:227-254. [DOI: 10.1007/978-94-017-7380-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
27
|
Lazova R, Laberge GS, Duvall E, Spoelstra N, Klump V, Sznol M, Cooper D, Spritz RA, Chang JT, Pawelek JM. A Melanoma Brain Metastasis with a Donor-Patient Hybrid Genome following Bone Marrow Transplantation: First Evidence for Fusion in Human Cancer. PLoS One 2013; 8:e66731. [PMID: 23840523 PMCID: PMC3694119 DOI: 10.1371/journal.pone.0066731] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 05/09/2013] [Indexed: 12/12/2022] Open
Abstract
Background Tumor cell fusion with motile bone marrow-derived cells (BMDCs) has long been posited as a mechanism for cancer metastasis. While there is much support for this from cell culture and animal studies, it has yet to be confirmed in human cancer, as tumor and marrow-derived cells from the same patient cannot be easily distinguished genetically. Methods We carried out genotyping of a metastatic melanoma to the brain that arose following allogeneic bone-marrow transplantation (BMT), using forensic short tandem repeat (STR) length-polymorphisms to distinguish donor and patient genomes. Tumor cells were isolated free of leucocytes by laser microdissection, and tumor and pre-transplant blood lymphocyte DNAs were analyzed for donor and patient alleles at 14 autosomal STR loci and the sex chromosomes. Results All alleles in the donor and patient pre-BMT lymphocytes were found in tumor cells. The alleles showed disproportionate relative abundances in similar patterns throughout the tumor, indicating the tumor was initiated by a clonal fusion event. Conclusions Our results strongly support fusion between a BMDC and a tumor cell playing a role in the origin of this metastasis. Depending on the frequency of such events, the findings could have important implications for understanding the generation of metastases, including the origins of tumor initiating cells and the cancer epigenome.
Collapse
Affiliation(s)
- Rossitza Lazova
- Deptartment of Dermatology, Yale School of Medicine, New Haven, Connecticut, United States of America ; The Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, United States of America
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Chan L, Terashima T, Urabe H, Lin F, Kojima H. Pathogenesis of diabetic neuropathy: bad to the bone. Ann N Y Acad Sci 2012; 1240:70-6. [PMID: 22172042 DOI: 10.1111/j.1749-6632.2011.06309.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Insulin and proinsulin are normally produced only by the pancreas and thymus. We detected in diabetic rodents the presence of extra pancreatic proinsulin-producing bone marrow-derived cells (PI-BMDCs) in the BM, liver, and fat. In mice and rats with diabetic neuropathy, we also found proinsulin-producing cells in the sciatic nerve and neurons of the dorsal root ganglion (DRG). BM transplantation experiments using genetically marked donor and recipient mice showed that the proinsulin-producing cells in the DRG, which morphologically resemble neurons, are actually polyploid proinsulin-producing fusion cells formed between neurons and PI-BMDCs. Additional experiments indicate that diabetic neuropathy is not simply the result of nerve cells being damaged directly by hyperglycemia. Rather, hyperglycemia induces fusogenic PI-BMDCs that travel to the peripheral nervous system, where they fuse with Schwann cells and DRG neurons, causing neuronal dysfunction and death, the sine qua non for diabetic neuropathy. Poorly controlled diabetes is indeed bad to the bone.
Collapse
Affiliation(s)
- Lawrence Chan
- Diabetes and Endocrinology Research Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, Texas 77030, USA.
| | | | | | | | | |
Collapse
|
29
|
Goupille O, Pallafacchina G, Relaix F, Conway SJ, Cumano A, Robert B, Montarras D, Buckingham M. Characterization of Pax3-expressing cells from adult blood vessels. J Cell Sci 2011; 124:3980-8. [PMID: 22159413 DOI: 10.1242/jcs.085373] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
We report expression of Pax3, an important regulator of skeletal muscle stem cell behaviour, in the brachial and femoral arteries of adult mice. In these contractile arteries of the limb, but not in the elastic arteries of the trunk, bands of GFP-positive cells were observed in Pax3(GFP/+) mice. Histological and biochemical examination of the vessels, together with clonal analysis after purification of Pax3-GFP-positive cells by flow cytometry, established their vascular smooth muscle identity. These blood-vessel-derived cells do not respond to inducers of other mesodermal cell types, such as bone, however, they can contribute to muscle fibre formation when co-cultured with skeletal muscle cells. This myogenic conversion depends on the expression of Pax3, but is rare and non-cell autonomous as it requires cell fusion. Myocardin, which promotes acquisition of a mature smooth muscle phenotype in these Pax3-GFP-positive cells, antagonises their potential for skeletal muscle differentiation. Genetic manipulation shows that myocardin is, however, positively regulated by Pax3, unlike genes for other myocardin-related factors, MRTFA, MRTFB or SRF. Expression of Pax3 overlaps with that reported for Msx2, which is required for smooth muscle differentiation of blood vessel-derived multipotent mesoangioblasts. These observations are discussed with respect to the origin and function of Pax3-expressing cells in blood vessels, and more general questions of cell fate determination and adult cell plasticity and reprogramming.
Collapse
Affiliation(s)
- Olivier Goupille
- Molecular Genetics of Morphogenesis Unit, Department of Developmental Biology, URA CNRS 2578, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Arno A, Smith AH, Blit PH, Shehab MA, Gauglitz GG, Jeschke MG. Stem Cell Therapy: A New Treatment for Burns? Pharmaceuticals (Basel) 2011; 4:1355-1380. [PMID: 27721328 PMCID: PMC4060129 DOI: 10.3390/ph4101355] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 09/21/2011] [Accepted: 10/10/2011] [Indexed: 12/12/2022] Open
Abstract
Stem cell therapy has emerged as a promising new approach in almost every medicine specialty. This vast, heterogeneous family of cells are now both naturally (embryonic and adult stem cells) or artificially obtained (induced pluripotent stem cells or iPSCs) and their fates have become increasingly controllable, thanks to ongoing research in this passionate new field. We are at the beginning of a new era in medicine, with multiple applications for stem cell therapy, not only as a monotherapy, but also as an adjunct to other strategies, such as organ transplantation or standard drug treatment. Regrettably, serious preclinical concerns remain and differentiation, cell fusion, senescence and signalling crosstalk with growth factors and biomaterials are still challenges for this promising multidisciplinary therapeutic modality. Severe burns have several indications for stem cell therapy, including enhancement of wound healing, replacement of damaged skin and perfect skin regeneration - incorporating skin appendages and reduced fibrosis -, as well as systemic effects, such as inflammation, hypermetabolism and immunosuppression. The aim of this review is to describe well established characteristics of stem cells and to delineate new advances in the stem cell field, in the context of burn injury and wound healing.
Collapse
Affiliation(s)
- Anna Arno
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, University of Toronto, 2075 Bayview Avenue, Toronto, Ontario M4N 3M5, Canada
- Plastic Surgery Department and Burn Unit, Vall d'Hebron University Hospital, Autonomous University of Barcelona, Passeig de la Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Alexandra H Smith
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, University of Toronto, 2075 Bayview Avenue, Toronto, Ontario M4N 3M5, Canada
| | - Patrick H Blit
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, University of Toronto, 2075 Bayview Avenue, Toronto, Ontario M4N 3M5, Canada
| | - Mohammed Al Shehab
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, University of Toronto, 2075 Bayview Avenue, Toronto, Ontario M4N 3M5, Canada
| | - Gerd G Gauglitz
- Department of Dermatology and Allergology, Ludwig Maximilians University, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
| | - Marc G Jeschke
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, University of Toronto, 2075 Bayview Avenue, Toronto, Ontario M4N 3M5, Canada.
| |
Collapse
|
31
|
Terashima T, Kojima H, Chan L. Bone marrow expression of poly(ADP-ribose) polymerase underlies diabetic neuropathy via hematopoietic-neuronal cell fusion. FASEB J 2011; 26:295-308. [PMID: 21978940 DOI: 10.1096/fj.11-186262] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diabetic neuropathy is the most common diabetic complication. The pathogenetic pathways include oxidative stress, advanced glycation end product (AGE) formation, protein kinase C, and NF-κB activation, as well as increased polyol flux. These metabolic perturbations affect neurons, Schwann cells, and vasa nervorum, which are held to be the primary cell types involved. We hypothesize that diabetes induces the appearance of abnormal bone marrow-derived cells (BMDCs) that fuse with neurons in the dorsal root ganglia (DRG) of mice, leading to diabetic neuropathy. Neuronal poly(ADP-ribose) polymerase-1 (PARP-1) activation in diabetes is known to generate free radical and oxidant-induced injury and poly(ADP-ribose) polymer formation, resulting in neuronal death and dysfunction, culminating in neuropathy. We further hypothesize that BM-specific PARP expression plays a determining role in disease pathogenesis. Here we show that bone marrow transplantation (BMT) of PARP-knockout (PARPKO) cells to wild-type mice protects against, whereas BMT of wild-type cells to PARPKO mice, which are normally "neuropathy-resistant," confers susceptibility to, diabetic neuropathy. The pathogenetic process involving hyperglycemia, BMDCs, and BMDC-neuron fusion can be recapitulated in vitro. Incubation in high, but not low, glucose confers fusogenicity to BMDCs, which are characterized by proinsulin (PI) and TNF-α coexpression; coincubation of isolated DRG neurons with PI-BMDCs in high glucose leads to spontaneous fusion between the 2 cell types, while the presence of a PARP inhibitor or use of PARPKO BMDCs in the incubation protects against BMDC-neuron fusion. These complementary in vivo and in vitro experiments indicate that BMDC-PARP expression promotes diabetic neuropathy via BMDC-neuron fusion.
Collapse
Affiliation(s)
- Tomoya Terashima
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | | |
Collapse
|
32
|
Lluis F, Pedone E, Pepe S, Cosma MP. The Wnt/β-catenin signaling pathway tips the balance between apoptosis and reprograming of cell fusion hybrids. Stem Cells 2010; 28:1940-9. [PMID: 20827748 PMCID: PMC3003905 DOI: 10.1002/stem.515] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 08/20/2010] [Indexed: 12/26/2022]
Abstract
Cell-cell fusion contributes to cell differentiation and developmental processes. We have previously showed that activation of Wnt/β-catenin enhances somatic cell reprograming after polyethylene glycol (PEG)-mediated fusion. Here, we show that neural stem cells and ESCs can fuse spontaneously in cocultures, although with very low efficiency (about 2%), as the hybrids undergo apoptosis. In contrast, when Wnt/β-catenin signaling is activated in ESCs and leads to accumulation of low amounts of β-catenin in the nucleus, activated ESCs can reprogram somatic cells with very high efficiency after spontaneous fusion. Furthermore, we also show that different levels of β-catenin accumulation in the ESC nuclei can modulate cell proliferation, although in our experimental setting, cell proliferation does not modulate the reprograming efficiency per se. Overall, the present study provides evidence that spontaneous fusion occurs, while the survival of the reprogramed clones is strictly dependent on induction of a Wnt-mediated reprograming pathway.
Collapse
Affiliation(s)
- Frederic Lluis
- Telethon Institute of Genetics and Medicine CNR, Naples, Italy
| | | | | | | |
Collapse
|