1
|
Stevens BT, Hatley ME. Developmental Heterogeneity of Rhabdomyosarcoma. Cold Spring Harb Perspect Med 2025; 15:a041583. [PMID: 38772705 PMCID: PMC11694754 DOI: 10.1101/cshperspect.a041583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Rhabdomyosarcoma (RMS) is a pediatric embryonal solid tumor and the most common pediatric soft tissue sarcoma. The histology and transcriptome of RMS resemble skeletal muscle progenitor cells that have failed to terminally differentiate. Thus, RMS is typically thought to arise from corrupted skeletal muscle progenitor cells during development. However, RMS can occur in body regions devoid of skeletal muscle, suggesting the potential for nonmyogenic cells of origin. Here, we discuss the interplay between RMS driver mutations and cell(s) of origin with an emphasis on driving location specificity. Additionally, we discuss the mechanisms governing RMS transformation events and tumor heterogeneity through the lens of transcriptional networks and epigenetic control. Finally, we reimagine Waddington's developmental landscape to include a plane of transformation connecting distinct lineage landscapes to more accurately reflect the phenomena observed in pediatric cancers.
Collapse
Affiliation(s)
- Bradley T Stevens
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
- St. Jude Graduate School of Biomedical Sciences, Memphis, Tennessee 38105, USA
| | - Mark E Hatley
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| |
Collapse
|
2
|
Yang X, Zhao Y, Liu W, Gao Z, Wang C, Wang C, Li S, Zhang X. Single-cell transcriptomics reveals neural stem cell trans-differentiation and cell subpopulations in whole heart decellularized extracellular matrix. BIOPHYSICS REPORTS 2024; 10:241-253. [PMID: 39281200 PMCID: PMC11399890 DOI: 10.52601/bpr.2024.240011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/16/2024] [Indexed: 09/18/2024] Open
Abstract
The whole heart decellularized extracellular matrix (ECM) has become a promising scaffold material for cardiac tissue engineering. Our previous research has shown that the whole heart acellular matrix possesses the memory function regulating neural stem cells (NSCs) trans-differentiating to cardiac lineage cells. However, the cell subpopulations and phenotypes in the trans-differentiation of NSCs have not been clearly identified. Here, we performed single-cell RNA sequencing and identified 2,765 cells in the recellularized heart with NSCs revealing the cellular diversity of cardiac and neural lineage, confirming NSCs were capable of trans-differentiating into the cardiac lineage while maintaining the original ability to differentiate into the neural lineage. Notably, the trans-differentiated heart-like cells have dual signatures of neuroectoderm and cardiac mesoderm. This study unveils an in-depth mechanism underlying the trans-differentiation of NSCs and provides a new opportunity and theoretical basis for cardiac regeneration.
Collapse
Affiliation(s)
- Xiaoning Yang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yuwei Zhao
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Wei Liu
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Zhongbao Gao
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Chunlan Wang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Changyong Wang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Siwei Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Xiao Zhang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| |
Collapse
|
3
|
Bakhmet EI, Tomilin AN. Key features of the POU transcription factor Oct4 from an evolutionary perspective. Cell Mol Life Sci 2021; 78:7339-7353. [PMID: 34698883 PMCID: PMC11072838 DOI: 10.1007/s00018-021-03975-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/16/2021] [Accepted: 10/12/2021] [Indexed: 01/06/2023]
Abstract
Oct4, a class V POU-domain protein that is encoded by the Pou5f1 gene, is thought to be a key transcription factor in the early development of mammals. This transcription factor plays indispensable roles in pluripotent stem cells as well as in the acquisition of pluripotency during somatic cell reprogramming. Oct4 has also been shown to play a role as a pioneer transcription factor during zygotic genome activation (ZGA) from zebrafish to human. However, during the past decade, several studies have brought these conclusions into question. It was clearly shown that the first steps in mouse development are not affected by the loss of Oct4. Subsequently, the role of Oct4 as a genome activator was brought into doubt. It was also found that the reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) could proceed without Oct4. In this review, we summarize recent findings, reassess the role of Oct4 in reprogramming and ZGA, and point to structural features that may underlie this role. We speculate that pluripotent stem cells resemble neural stem cells more closely than previously thought. Oct4 orthologs within the POUV class hold key roles in genome activation during early development of species with late ZGA. However, in Placentalia, eutherian-specific proteins such as Dux overtake Oct4 in ZGA and endow them with the formation of an evolutionary new tissue-the placenta.
Collapse
Affiliation(s)
- Evgeny I Bakhmet
- Laboratory of the Molecular Biology of Stem Cells, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia.
| | - Alexey N Tomilin
- Laboratory of the Molecular Biology of Stem Cells, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
4
|
Langdon CG, Gadek KE, Garcia MR, Evans MK, Reed KB, Bush M, Hanna JA, Drummond CJ, Maguire MC, Leavey PJ, Finkelstein D, Jin H, Schreiner PA, Rehg JE, Hatley ME. Synthetic essentiality between PTEN and core dependency factor PAX7 dictates rhabdomyosarcoma identity. Nat Commun 2021; 12:5520. [PMID: 34535684 PMCID: PMC8448747 DOI: 10.1038/s41467-021-25829-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 09/01/2021] [Indexed: 02/08/2023] Open
Abstract
PTEN promoter hypermethylation is nearly universal and PTEN copy number loss occurs in ~25% of fusion-negative rhabdomyosarcoma (FN-RMS). Here we show Pten deletion in a mouse model of FN-RMS results in less differentiated tumors more closely resembling human embryonal RMS. PTEN loss activated the PI3K pathway but did not increase mTOR activity. In wild-type tumors, PTEN was expressed in the nucleus suggesting loss of nuclear PTEN functions could account for these phenotypes. Pten deleted tumors had increased expression of transcription factors important in neural and skeletal muscle development including Dbx1 and Pax7. Pax7 deletion completely rescued the effects of Pten loss. Strikingly, these Pten;Pax7 deleted tumors were no longer FN-RMS but displayed smooth muscle differentiation similar to leiomyosarcoma. These data highlight how Pten loss in FN-RMS is connected to a PAX7 lineage-specific transcriptional output that creates a dependency or synthetic essentiality on the transcription factor PAX7 to maintain tumor identity.
Collapse
Affiliation(s)
- Casey G Langdon
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Katherine E Gadek
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Matthew R Garcia
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Myron K Evans
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Kristin B Reed
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Rhodes College, Memphis, TN, 38112, USA
| | - Madeline Bush
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- St. Jude Graduate School of Biomedical Sciences, Memphis, TN, 38105, USA
| | - Jason A Hanna
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Purdue Center for Cancer Research, Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Catherine J Drummond
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Pathology, University of Otago, Dunedin, Otago, New Zealand
| | - Matthew C Maguire
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Patrick J Leavey
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Hongjian Jin
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Patrick A Schreiner
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jerold E Rehg
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Mark E Hatley
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
5
|
Fang J, Sia J, Soto J, Wang P, Li LK, Hsueh YY, Sun R, Francis Faull K, Tidball JG, Li S. Skeletal muscle regeneration via the chemical induction and expansion of myogenic stem cells in situ or in vitro. Nat Biomed Eng 2021; 5:864-879. [PMID: 33737730 PMCID: PMC8387336 DOI: 10.1038/s41551-021-00696-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 02/04/2021] [Indexed: 02/05/2023]
Abstract
Muscle loss and impairment resulting from traumatic injury can be alleviated by therapies using muscle stem cells. However, collecting sufficient numbers of autologous myogenic stem cells and expanding them efficiently has been challenging. Here we show that myogenic stem cells (predominantly Pax7+ cells)-which were selectively expanded from readily obtainable dermal fibroblasts or skeletal muscle stem cells using a specific cocktail of small molecules and transplanted into muscle injuries in adult, aged or dystrophic mice-led to functional muscle regeneration in the three animal models. We also show that sustained release of the small-molecule cocktail in situ through polymer nanoparticles led to muscle repair by inducing robust activation and expansion of resident satellite cells. Chemically induced stem cell expansion in vitro and in situ may prove to be advantageous for stem cell therapies that aim to regenerate skeletal muscle and other tissues.
Collapse
Affiliation(s)
- Jun Fang
- Department of Bioengineering, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Junren Sia
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jennifer Soto
- Department of Bioengineering, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Pingping Wang
- Department of Bioengineering, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA,Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - LeeAnn K. Li
- Department of Bioengineering, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA,David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Yuan-Yu Hsueh
- Department of Bioengineering, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA,Division of Plastic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70456, Taiwan
| | - Raymond Sun
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kym Francis Faull
- Pasarow Mass Spectrometry Laboratory, Jane and Terry Semel Institute for Neuroscience and Human Behavior and Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - James G. Tidball
- Department of Integrative Biology and Physiology, Molecular, Cellular & Integrative Physiology Program, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Song Li
- Department of Bioengineering, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA,Correspondence and requests for materials should be addressed to S. L.,
| |
Collapse
|
6
|
Rosińska S, Gavard J. Tumor Vessels Fuel the Fire in Glioblastoma. Int J Mol Sci 2021; 22:6514. [PMID: 34204510 PMCID: PMC8235363 DOI: 10.3390/ijms22126514] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma, a subset of aggressive brain tumors, deploy several means to increase blood vessel supply dedicated to the tumor mass. This includes typical program borrowed from embryonic development, such as vasculogenesis and sprouting angiogenesis, as well as unconventional processes, including co-option, vascular mimicry, and transdifferentiation, in which tumor cells are pro-actively engaged. However, these neo-generated vascular networks are morphologically and functionally abnormal, suggesting that the vascularization processes are rather inefficient in the tumor ecosystem. In this review, we reiterate the specificities of each neovascularization modality in glioblastoma, and, how they can be hampered mechanistically in the perspective of anti-cancer therapies.
Collapse
Affiliation(s)
- Sara Rosińska
- CRCINA, Inserm, CNRS, Université de Nantes, 44000 Nantes, France;
| | - Julie Gavard
- CRCINA, Inserm, CNRS, Université de Nantes, 44000 Nantes, France;
- Integrated Center for Oncology, ICO, 44800 St. Herblain, France
| |
Collapse
|
7
|
Ausems CRM, van Engelen BGM, van Bokhoven H, Wansink DG. Systemic cell therapy for muscular dystrophies : The ultimate transplantable muscle progenitor cell and current challenges for clinical efficacy. Stem Cell Rev Rep 2021; 17:878-899. [PMID: 33349909 PMCID: PMC8166694 DOI: 10.1007/s12015-020-10100-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 01/07/2023]
Abstract
The intrinsic regenerative capacity of skeletal muscle makes it an excellent target for cell therapy. However, the potential of muscle tissue to renew is typically exhausted and insufficient in muscular dystrophies (MDs), a large group of heterogeneous genetic disorders showing progressive loss of skeletal muscle fibers. Cell therapy for MDs has to rely on suppletion with donor cells with high myogenic regenerative capacity. Here, we provide an overview on stem cell lineages employed for strategies in MDs, with a focus on adult stem cells and progenitor cells resident in skeletal muscle. In the early days, the potential of myoblasts and satellite cells was explored, but after disappointing clinical results the field moved to other muscle progenitor cells, each with its own advantages and disadvantages. Most recently, mesoangioblasts and pericytes have been pursued for muscle cell therapy, leading to a handful of preclinical studies and a clinical trial. The current status of (pre)clinical work for the most common forms of MD illustrates the existing challenges and bottlenecks. Besides the intrinsic properties of transplantable cells, we discuss issues relating to cell expansion and cell viability after transplantation, optimal dosage, and route and timing of administration. Since MDs are genetic conditions, autologous cell therapy and gene therapy will need to go hand-in-hand, bringing in additional complications. Finally, we discuss determinants for optimization of future clinical trials for muscle cell therapy. Joined research efforts bring hope that effective therapies for MDs are on the horizon to fulfil the unmet clinical need in patients.
Collapse
Affiliation(s)
- C Rosanne M Ausems
- Donders lnstitute for Brain Cognition and Behavior, Department of Human Genetics, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands
- Donders lnstitute for Brain Cognition and Behavior, Department of Neurology, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Department of Cell Biology, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands
| | - Baziel G M van Engelen
- Donders lnstitute for Brain Cognition and Behavior, Department of Neurology, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands
| | - Hans van Bokhoven
- Donders lnstitute for Brain Cognition and Behavior, Department of Human Genetics, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands.
| | - Derick G Wansink
- Radboud Institute for Molecular Life Sciences, Department of Cell Biology, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands.
| |
Collapse
|
8
|
Wang C, Yang X, Zhang X, Liu B, Liu W, Shen Y, Gao Z, Yin Q, Wang C, Zhou J. TMT-based quantitative proteome profiles reveal the memory function of a whole heart decellularized matrix for neural stem cell trans-differentiation into the cardiac lineage. Biomater Sci 2021; 9:3692-3704. [PMID: 34008595 DOI: 10.1039/d0bm01287d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Whole organ or tissue decellularized matrices are a promising scaffold for tissue engineering because they maintain the specific memory of the original organ or tissue. A whole organ or tissue decellularized matrix contains extracellular matrix (ECM) components, and exhibits ultrastructural and mechanical properties, which could significantly regulate the fate of stem cells. To better understand the memory function of whole organ decellularized matrices, we constructed a heart decellularized matrix and seeded cross-embryonic layer stem cells - neural stem cells (NSCs) to repopulate the matrix, engineering cardiac tissue, in which a large number of NSCs differentiated into the neural lineage, but besides that, NSCs showed an obvious tendency of trans-differentiating into cardiac lineage cells. The results demonstrated that the whole heart decellularized microenvironment possesses memory function. To reveal the underlying mechanism, TMT-based quantitative proteomics analysis was used to identify the differently expressed proteins in the whole heart decellularized matrix compared with a brain decellularized matrix. 937 of the proteins changed over 1.5 fold, with 573 of the proteins downregulated and 374 of the proteins upregulated, among which integrin ligands in the ECM serve as key signals in regulating NSC fate. The findings here provide a novel insight into the memory function of tissue-specific microenvironments and pave the way for the therapeutic application of personalized tissues.
Collapse
Affiliation(s)
- Changyong Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing 100850, PR China.
| | - Xiaoning Yang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing 100850, PR China.
| | - Xiao Zhang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing 100850, PR China.
| | - Baijun Liu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing 100850, PR China.
| | - Wei Liu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing 100850, PR China.
| | - Yuan Shen
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing 100850, PR China.
| | - Zhongbao Gao
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing 100850, PR China.
| | - Qi Yin
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing 100850, PR China.
| | - Chunlan Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing 100850, PR China.
| | - Jin Zhou
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing 100850, PR China.
| |
Collapse
|
9
|
He R, Li H, Wang L, Li Y, Zhang Y, Chen M, Zhu Y, Zhang C. Engraftment of human induced pluripotent stem cell-derived myogenic progenitors restores dystrophin in mice with duchenne muscular dystrophy. Biol Res 2020; 53:22. [PMID: 32430065 PMCID: PMC7238630 DOI: 10.1186/s40659-020-00288-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a devastating genetic muscular disorder with no effective treatment that is caused by the loss of dystrophin. Human induced pluripotent stem cells (hiPSCs) offer a promising unlimited resource for cell-based therapies of muscular dystrophy. However, their clinical applications are hindered by inefficient myogenic differentiation, and moreover, the engraftment of non-transgene hiPSC-derived myogenic progenitors has not been examined in the mdx mouse model of DMD. Methods We investigated the muscle regenerative potential of myogenic progenitors derived from hiPSCs in mdx mice. The hiPSCs were transfected with enhanced green fluorescent protein (EGFP) vector and defined as EGFP hiPSCs. Myogenic differentiation was performed on EGFP hiPSCs with supplementary of basic fibroblast growth factor, forskolin, 6-bromoindirubin-3′-oxime as well as horse serum. EGFP hiPSCs-derived myogenic progenitors were engrafted into mdx mice via both intramuscular and intravenous injection. The restoration of dystrophin expression, the ratio of central nuclear myofibers, and the transplanted cells-derived satellite cells were accessed after intramuscular and systemic transplantation. Results We report that abundant myogenic progenitors can be generated from hiPSCs after treatment with these three small molecules, with consequent terminal differentiation giving rise to mature myotubes in vitro. Upon intramuscular or systemic transplantation into mdx mice, these myogenic progenitors engrafted and contributed to human-derived myofiber regeneration in host muscles, restored dystrophin expression, ameliorated pathological lesions, and seeded the satellite cell compartment in dystrophic muscles. Conclusions This study demonstrates the muscle regeneration potential of myogenic progenitors derived from hiPSCs using non-transgenic induction methods. Engraftment of hiPSC-derived myogenic progenitors could be a potential future therapeutic strategy to treat DMD in a clinical setting.
Collapse
Affiliation(s)
- Ruojie He
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, Guangdong, China
| | - Huan Li
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, Guangdong, China
| | - Liang Wang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, Guangdong, China
| | - Yaqin Li
- Department of Neurology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yu Zhang
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Menglong Chen
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Yuling Zhu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China. .,Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, Guangdong, China.
| | - Cheng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China. .,Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Multi-Spheroid-Loaded Human Acellular Dermal Matrix Carrier Preserves Its Spheroid Shape and Improves In Vivo Adipose-Derived Stem Cell Delivery and Engraftment. Tissue Eng Regen Med 2020; 17:271-283. [PMID: 32314311 DOI: 10.1007/s13770-020-00252-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/09/2020] [Accepted: 03/15/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Current in vivo adult stem cell delivery presents limited clinical effects due to poor engraftment and survival. To overcome current challenges in cell delivery and promote surgical cell delivery for soft tissue repair, a multi-spheroid-loaded thin sectioned acellular dermal matrix (tsADM) carrier which preserves loaded spheroids' three-dimensional (3D) structure, was developed. METHODS Adipose-derived stem cells (ASCs) were used for spheroid delivery. After generating spheroids in 3D cell culture dishes, spheroid plasticity and survival in-between coverslips were evaluated. Spheroids were loaded onto tsADM, their shape changes were followed up for 14 days, and then imaged. Spheroid adhesion stability to tsADM against shear stress was also evaluated. Finally, cell delivery efficacy was compared with cell-seeded tsADM by in vivo implantation and histological evaluation. RESULTS Spheroids withstood cyclic compression stress and maintained their 3D shape without fusion after 48 h of culture in-between coverslips. Cell survival improved when spheroids were cultured on tsADM in-between the coverslips. Spheroid-loaded tsADM with coverslips maintained their spheroid outline for 14 days of culture whereas without coverslips, the group lost their outline due to spreading after 4 days in culture. Spheroids loaded onto tsADMs were more stable after six rather than 3 days in culture. Spheroid-loaded tsADMs showed about a 2.96-fold higher ASCs transplantation efficacy than cell-seeded tsADMs after 2 weeks of in vivo transplantation. CONCLUSION These results indicate that transplantation of spheroid-loaded tsADMs significantly improved cell delivery. These findings suggest that a combined approach with other cells, drugs, and nanoparticles may improve cell delivery and therapeutic efficacy.
Collapse
|
11
|
Murphy AR, Haynes JM, Laslett AL, Cameron NR, O'Brien CM. Three-dimensional differentiation of human pluripotent stem cell-derived neural precursor cells using tailored porous polymer scaffolds. Acta Biomater 2020; 101:102-116. [PMID: 31610339 DOI: 10.1016/j.actbio.2019.10.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/30/2019] [Accepted: 10/07/2019] [Indexed: 02/07/2023]
Abstract
This study investigates the utility of a tailored poly(ethylene glycol) diacrylate-crosslinked porous polymeric tissue engineering scaffold, with mechanical properties specifically optimised to be comparable to that of mammalian brain tissue for 3D human neural cell culture. Results obtained here demonstrate the attachment, proliferation and terminal differentiation of both human induced pluripotent stem cell- and embryonic stem cell-derived neural precursor cells (hPSC-NPCs) throughout the interconnected porous network within laminin-coated scaffolds. Phenotypic data and functional analyses are presented demonstrating that this material supports terminal in vitro neural differentiation of hPSC-NPCs to a mixed population of viable neuronal and glial cells for periods of up to 49 days. This is evidenced by the upregulation of TUBB3, MAP2, SYP and GFAP gene expression, as well as the presence of the proteins βIII-TUBULIN, NEUN, MAP2 and GFAP. Functional maturity of neural cells following 49 days 3D differentiation culture was tested via measurement of intracellular calcium. These analyses revealed spontaneously active, synchronous and rhythmic calcium flux, as well as response to the neurotransmitter glutamate. This tailored construct has potential application as an improved in vitro human neurogenesis model with utility in platform drug discovery programs. STATEMENT OF SIGNIFICANCE: The interconnected porosity of polyHIPE scaffolds exhibits the ability to support three-dimensional neural cell network formation due to limited resistance to cellular migration and re-organisation. The previously developed scaffold material displays mechanical properties similar to that of the mammalian brain. This research also employs the utility of pluripotent stem cell-derived neural cells which are of greater clinical relevance than primary neural cell lines. This scaffold material has future potential in better mimicking three-dimensional neural networks found in the human brain and may result in improved in vitro models for disease modelling and drug screening applications.
Collapse
|
12
|
Affiliation(s)
| | - Donald S Sakaguchi
- Department of Genetics, Development and Cell Biology; Neuroscience Program; Nanovaccine Institute, Iowa State University, Ames, IA, USA
| |
Collapse
|
13
|
André LM, Ausems CRM, Wansink DG, Wieringa B. Abnormalities in Skeletal Muscle Myogenesis, Growth, and Regeneration in Myotonic Dystrophy. Front Neurol 2018; 9:368. [PMID: 29892259 PMCID: PMC5985300 DOI: 10.3389/fneur.2018.00368] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/07/2018] [Indexed: 12/16/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) and 2 (DM2) are autosomal dominant degenerative neuromuscular disorders characterized by progressive skeletal muscle weakness, atrophy, and myotonia with progeroid features. Although both DM1 and DM2 are characterized by skeletal muscle dysfunction and also share other clinical features, the diseases differ in the muscle groups that are affected. In DM1, distal muscles are mainly affected, whereas in DM2 problems are mostly found in proximal muscles. In addition, manifestation in DM1 is generally more severe, with possible congenital or childhood-onset of disease and prominent CNS involvement. DM1 and DM2 are caused by expansion of (CTG•CAG)n and (CCTG•CAGG)n repeats in the 3' non-coding region of DMPK and in intron 1 of CNBP, respectively, and in overlapping antisense genes. This critical review will focus on the pleiotropic problems that occur during development, growth, regeneration, and aging of skeletal muscle in patients who inherited these expansions. The current best-accepted idea is that most muscle symptoms can be explained by pathomechanistic effects of repeat expansion on RNA-mediated pathways. However, aberrations in DNA replication and transcription of the DM loci or in protein translation and proteome homeostasis could also affect the control of proliferation and differentiation of muscle progenitor cells or the maintenance and physiological integrity of muscle fibers during a patient's lifetime. Here, we will discuss these molecular and cellular processes and summarize current knowledge about the role of embryonic and adult muscle-resident stem cells in growth, homeostasis, regeneration, and premature aging of healthy and diseased muscle tissue. Of particular interest is that also progenitor cells from extramuscular sources, such as pericytes and mesoangioblasts, can participate in myogenic differentiation. We will examine the potential of all these types of cells in the application of regenerative medicine for muscular dystrophies and evaluate new possibilities for their use in future therapy of DM.
Collapse
Affiliation(s)
- Laurène M André
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - C Rosanne M Ausems
- Department of Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Derick G Wansink
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Bé Wieringa
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
14
|
Sandquist EJ, Essner JJ, Sakaguchi DS. Xenotransplantation of adult hippocampal neural progenitors into the developing zebrafish for assessment of stem cell plasticity. PLoS One 2018; 13:e0198025. [PMID: 29795671 PMCID: PMC5967829 DOI: 10.1371/journal.pone.0198025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/12/2018] [Indexed: 12/26/2022] Open
Abstract
Adult stem cells are considered multipotent, restricted to differentiate into a few tissue-specific cell types. With the advent of technologies which can dedifferentiate and transdifferentiate cell types, assumptions about the process of cell fate determination must be reconsidered, including the role of extrinsic versus intrinsic factors. To determine the plasticity of adult neural progenitors, rat hippocampal progenitor cells were xenotransplanted into embryonic zebrafish. These animals allow for easy detection of transplanted cells due to their external development and transparency at early stages. Adult neural progenitors were observed throughout the zebrafish for the duration of the experiment (at least five days post-transplantation). While the majority of transplanted cells were observed in the central nervous system, a large percentage of cells were located in superficial tissues. However, approximately one-third of these cells retained neural morphology and expression of the neuronal marker, Class III β-tubulin, indicating that the transplanted adult neural progenitors did not adapt alternate fates. A very small subset of cells demonstrated unique, non-neural flattened morphology, suggesting that adult neural progenitors may exhibit plasticity in this model, though at a very low rate. These findings demonstrate that the developing zebrafish may be an efficient model to explore plasticity of a variety of adult stem cell types and the role of external factors on cell fate.
Collapse
Affiliation(s)
- Elizabeth J. Sandquist
- Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States of America
- * E-mail: (EJS); (DSS)
| | - Jeffrey J. Essner
- Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States of America
| | - Donald S. Sakaguchi
- Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States of America
- Neuroscience Program, Iowa State University, Ames, Iowa, United States of America
- * E-mail: (EJS); (DSS)
| |
Collapse
|
15
|
Hollands P, Aboyeji D, Orcharton M. Dental pulp stem cells in regenerative medicine. Br Dent J 2018; 224:sj.bdj.2018.348. [PMID: 29725075 DOI: 10.1038/sj.bdj.2018.348] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2018] [Indexed: 12/22/2022]
Abstract
The mesenchymal stem cells (dental pulp stem cells; DPSC) found inside teeth represent a significant future source of stem cells for regenerative medicine procedures. This review describes the ontogeny of DPSC; the laboratory processing and collection of DPSC; the immuno-cytochemical characterisation of DPSC; the differentiation between adult DPSC and DPSC obtained from exfoliated deciduous teeth (SHED) and their potential use in regenerative medicine procedures in the future both in dental and general medical applications.
Collapse
Affiliation(s)
- P Hollands
- WideCells Institute of Stem Cell Technology (ISCT), University of Manchester Innovation Centre, Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT
| | - D Aboyeji
- WideCells Institute of Stem Cell Technology (ISCT), University of Manchester Innovation Centre, Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT
| | - M Orcharton
- WideCells Institute of Stem Cell Technology (ISCT), University of Manchester Innovation Centre, Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT
| |
Collapse
|
16
|
Lizier M, Castelli A, Montagna C, Lucchini F, Vezzoni P, Faggioli F. Cell fusion in the liver, revisited. World J Hepatol 2018; 10:213-221. [PMID: 29527257 PMCID: PMC5838440 DOI: 10.4254/wjh.v10.i2.213] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/28/2017] [Accepted: 02/06/2018] [Indexed: 02/06/2023] Open
Abstract
There is wide agreement that cell fusion is a physiological process in cells in mammalian bone, muscle and placenta. In other organs, such as the cerebellum, cell fusion is controversial. The liver contains a considerable number of polyploid cells: They are commonly believed to originate by genome endoreplication, although the contribution of cell fusion to polyploidization has not been excluded. Here, we address the topic of cell fusion in the liver from a historical point of view. We discuss experimental evidence clearly supporting the hypothesis that cell fusion occurs in the liver, specifically when bone marrow cells were injected into mice and shown to rescue genetic hepatic degenerative defects. Those experiments-carried out in the latter half of the last century-were initially interpreted to show “transdifferentiation”, but are now believed to demonstrate fusion between donor macrophages and host hepatocytes, raising the possibility that physiologically polyploid cells, such as hepatocytes, could originate, at least partially, through homotypic cell fusion. In support of the homotypic cell fusion hypothesis, we present new data generated using a chimera-based model, a much simpler model than those previously used. Cell fusion as a road to polyploidization in the liver has not been extensively investigated, and its contribution to a variety of conditions, such as viral infections, carcinogenesis and aging, remains unclear.
Collapse
Affiliation(s)
- Michela Lizier
- Istituto di Ricerca Genetica e Biomedica, CNR, Milan 20138, Italy
- Human Genome Laboratory, Humanitas Clinical and Research Center, IRCCS, Milan 20089, Italy
| | - Alessandra Castelli
- Istituto di Ricerca Genetica e Biomedica, CNR, Milan 20138, Italy
- Human Genome Laboratory, Humanitas Clinical and Research Center, IRCCS, Milan 20089, Italy
| | - Cristina Montagna
- Department of Genetics and Pathology Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Franco Lucchini
- Centro Ricerche Biotecnologiche, Università Cattolica del Sacro Cuore, Cremona 26100, Italy
| | - Paolo Vezzoni
- Istituto di Ricerca Genetica e Biomedica, CNR, Milan 20138, Italy
- Human Genome Laboratory, Humanitas Clinical and Research Center, IRCCS, Milan 20089, Italy
| | - Francesca Faggioli
- Istituto di Ricerca Genetica e Biomedica, CNR, Milan 20138, Italy
- Human Genome Laboratory, Humanitas Clinical and Research Center, IRCCS, Milan 20089, Italy
| |
Collapse
|
17
|
Ku J, El-Hashash A. Stem Cell Roles and Applications in Genetic Neurodegenerative Diseases. STEM CELLS IN CLINICAL APPLICATIONS 2018. [DOI: 10.1007/978-3-319-98065-2_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
18
|
Tang Y, Yu P, Cheng L. Current progress in the derivation and therapeutic application of neural stem cells. Cell Death Dis 2017; 8:e3108. [PMID: 29022921 PMCID: PMC5682670 DOI: 10.1038/cddis.2017.504] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/28/2017] [Accepted: 08/31/2017] [Indexed: 12/13/2022]
Abstract
Neural stem cells (NSCs) have a unique role in neural regeneration. Cell therapy based on NSC transplantation is a promising tool for the treatment of nervous system diseases. However, there are still many issues and controversies associated with the derivation and therapeutic application of these cells. In this review, we summarize the different sources of NSCs and their derivation methods, including direct isolation from primary tissues, differentiation from pluripotent stem cells and transdifferentiation from somatic cells. We also review the current progress in NSC implantation for the treatment of various neural defects and injuries in animal models and clinical trials. Finally, we discuss potential optimization strategies for NSC derivation and propose urgent challenges to the clinical translation of NSC-based therapies in the near future.
Collapse
Affiliation(s)
- Yuewen Tang
- National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Shanghai Institute of Haematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei Yu
- Department of Orthopaedics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Cheng
- National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Shanghai Institute of Haematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Sam TN, Xiao J, Roehrich H, Low WC, Gregerson DS. Engrafted Neural Progenitor Cells Express a Tissue-Restricted Reporter Gene Associated with Differentiated Retinal Photoreceptor Cells. Cell Transplant 2017; 15:147-60. [PMID: 16719048 DOI: 10.3727/000000006783982098] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Neural progenitor cells (NPCs) have shown ability to repair injured CNS, and might provide precursors to retinal neurons. NPCs were isolated from the brains of 14 day murine embryos of transgenic mice that express β-galactosidase (β-gal) on the arrestin promoter, which specifically directs expression to retinal photoreceptor cells. NPCs were transferred to adult, syngeneic mice via inoculation into the anterior chamber of the eye, the peritoneal cavity, or the brain. At 14 weeks postgrafting, tissues were collected and examined to determine if differentiated NPC progeny were present in retina based on histochemical detection of β-gal. Four of six anterior chamber-inoculated recipients showed Bluo-gal-stained cells in retina, indicating the presence of transferred NPCs or their progeny. Because the progenitor cells do not express β-gal, positive staining indicates differentiation leading to activation of the arrestin promoter. Two recipients inoculated by the intraperitoneal route also exhibited Bluo-gal staining in retina. The NPCs did not express β-gal if inoculated into brain, but survived and dispersed. Most recipients, regardless of inoculation route, were PCR positive for β-gal DNA in extraocular tissues, but no Bluo-gal staining was found outside of the retina. Injury to the retina promoted, but was not required, for progenitor cell engraftment. β-Gal-positive cells were concentrated in the outer layers of the retina. In summary, a reporter gene specifically expressed in differentiated retinal photoreceptor cells due to the activity of the arrestin promoter was expressed in recipient mouse retina following transfer of NPCs prepared from the β-gal transgenic mice. The presence of β-gal DNA, but not Bluo-gal staining, in spleen and other tissues revealed that the cells also migrated elsewhere and took up residence in other organs, but did not undergo differentiation that led to β-gal expression.
Collapse
Affiliation(s)
- Thien N Sam
- Department of Ophthalmology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|
20
|
Bacou F, Andalousi RBE, Daussin PA, Micallef JP, Levin JM, Chammas M, Casteilla L, Reyne Y, Nouguès J. Transplantation of Adipose Tissue-Derived Stromal Cells Increases Mass and Functional Capacity of Damaged Skeletal Muscle. Cell Transplant 2017; 13:103-111. [PMID: 28853933 DOI: 10.3727/000000004773301771] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The regenerating skeletal muscle environment is capable of inducing uncommitted progenitors to terminally differentiate. The aim of this work was to determine whether adipose tissue-derived stromal cells were able to participate in muscle regeneration and to characterize the effect on muscle mass and functional capacities after transplantation of these cells. Adipose tissue stromal cells labeled with Adv cyto LacZ from 3-day-old primary cultures (SVF1) were autotransplanted into damaged tibialis anterior muscles. Fifteen days later, β-galactosidase staining of regenerated fibers was detected, showing participation of these cells in muscle regeneration. Two months after SVF1 cell transfer, muscles were heavier, showed a significantly larger fiber section area, and developed a significantly higher maximal force compared with damaged control muscles. These results are similar to those previously obtained after satellite cell transplantation. However, SVF1 transfer also generated a small amount of adipose tissue localized along the needle course. To minimize these adipose contaminants, we transferred cells from 7-day-old secondary cultures of the SVF1, containing only a small proportion of already engaged preadipocytes (SVF2). Under these conditions, no adipose tissue was observed in regenerated muscle but there was also no effect on muscle performances compared with damaged control muscles. This result provides further evidence for the existence of progenitor cells in the stromal fraction of freshly isolated adipose tissue cells, which, under our conditions, keep some of their pluripotent properties in primary cultures.
Collapse
Affiliation(s)
- Francis Bacou
- UMR 866 Différenciation cellulaire et Croissance, INRA, Montpellier Cedex 1, France
| | | | - Paul-André Daussin
- UMR 866 Différenciation cellulaire et Croissance, INRA, Montpellier Cedex 1, France.,Service de Chirurgie Orthopédique 2 et Chirurgie de la Main, Hôpital Lapeyronie, CHU Montpellier, France
| | - Jean-Paul Micallef
- Service de Chirurgie Orthopédique 2 et Chirurgie de la Main, Hôpital Lapeyronie, CHU Montpellier, France
| | - Jonathan M Levin
- UMR 866 Différenciation cellulaire et Croissance, INRA, Montpellier Cedex 1, France
| | - Michel Chammas
- Service de Chirurgie Orthopédique 2 et Chirurgie de la Main, Hôpital Lapeyronie, CHU Montpellier, France
| | | | - Yves Reyne
- UMR 866 Différenciation cellulaire et Croissance, INRA, Montpellier Cedex 1, France
| | - Jean Nouguès
- UMR 866 Différenciation cellulaire et Croissance, INRA, Montpellier Cedex 1, France
| |
Collapse
|
21
|
In Utero Stem Cell Transplantation: Potential Therapeutic Application for Muscle Diseases. Stem Cells Int 2017; 2017:3027520. [PMID: 28596791 PMCID: PMC5450178 DOI: 10.1155/2017/3027520] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/26/2017] [Indexed: 01/07/2023] Open
Abstract
Muscular dystrophies, myopathies, and traumatic muscle injury and loss encompass a large group of conditions that currently have no cure. Myoblast transplantations have been investigated as potential cures for these conditions for decades. However, current techniques lack the ability to generate cell numbers required to produce any therapeutic benefit. In utero stem cell transplantation into embryos has been studied for many years mainly in the context of hematopoietic cells and has shown to have experimental advantages and therapeutic applications. Moreover, patient-derived cells can be used for experimental transplantation into nonhuman animal embryos via in utero injection as the immune response is absent at such early stages of development. We therefore propose in utero transplantation as a potential method to generate patient-derived humanized skeletal muscle as well as muscle stem cells in animals for therapeutic purposes as well as patient-specific drug screening.
Collapse
|
22
|
Lee SE, Uhm SJ, Son YJ, Park YG, Kim EY, Park SP. Intermediate Reprogramming of Mouse Skin Fibroblasts into Stem-Like Cells by Bone Morphogenetic Protein 4. Cell Reprogram 2017; 19:107-115. [PMID: 28170287 DOI: 10.1089/cell.2016.0037] [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: 11/12/2022] Open
Abstract
Specific transcription factors are sufficient to reprogram fully induced pluripotent stem cells or other types of cells. These findings raise the question of whether chemical molecules or proteins can replace transcription factors to alter the defined cell fate. In this study, we treated mouse skin fibroblasts (MSFs) with bone morphogenetic protein 4 (BMP4) and examined intermediate reprogramming of MSFs into stem-like cells. Putative epidermal stem cells isolated from the ventral skin epidermis of an adult mouse were used to confirm the reprogramming activity of BMP4, which increased the proliferation of these cells. After these cells formed spheroids, they were treated with BMP4 and cultured for 5 days. Following BMP4 treatment, the characteristics of these cells changed, and they expressed Oct-4 and its target transcripts Nanog, Sox2, and alkaline phosphatase. To confirm the stem cell potency of these cells, we induced their differentiation into cardiomyocytes. Stem-like cell-derived cardiomyocytes exhibited mRNA expression of cardiac mesoderm markers such as Nk2 transcription factor-related locus 5 and connexin 40, and the cardiomyocyte marker troponin T. These differentiated cells exhibited contracting masses. These results suggest that BMP4-mediated somatic stem cell reprogramming may become an alternative approach for cell therapy.
Collapse
Affiliation(s)
- Seung-Eun Lee
- 1 Stem Cell Research Center, Jeju National University , Jeju, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju, Korea
| | - Sang-Jun Uhm
- 3 Department of Animal Science and Biotechnology, Sangji Youngseo College , Wonju, South Korea
| | - Yeo-Jin Son
- 1 Stem Cell Research Center, Jeju National University , Jeju, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju, Korea
| | - Yun-Gwi Park
- 1 Stem Cell Research Center, Jeju National University , Jeju, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju, Korea
| | - Eun-Young Kim
- 1 Stem Cell Research Center, Jeju National University , Jeju, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju, Korea.,4 Mirae Cell Bio, Seoul, Korea
| | - Se-Pill Park
- 1 Stem Cell Research Center, Jeju National University , Jeju, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju, Korea.,4 Mirae Cell Bio, Seoul, Korea
| |
Collapse
|
23
|
Xin ZC, Xu YD, Lin G, Lue TF, Guo YL. Recruiting endogenous stem cells: a novel therapeutic approach for erectile dysfunction. Asian J Androl 2016; 18:10-5. [PMID: 25926601 PMCID: PMC4736335 DOI: 10.4103/1008-682x.150040] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Transplanted stem cells (SCs), owing to their regenerative capacity, represent one of the most promising methods to restore erectile dysfunction (ED). However, insufficient source, invasive procedures, ethical and regulatory issues hamper their use in clinical applications. The endogenous SCs/progenitor cells resident in organ and tissues play critical roles for organogenesis during development and for tissue homeostasis in adulthood. Even without any therapeutic intervention, human body has a robust self-healing capability to repair the damaged tissues or organs. Therefore, SCs-for-ED therapy should not be limited to a supply-side approach. The resident endogenous SCs existing in patients could also be a potential target for ED therapy. The aim of this review was to summarize contemporary evidence regarding: (1) SC niche and SC biological features in vitro; (2) localization and mobilization of endogenous SCs; (3) existing evidence of penile endogenous SCs and their possible mode of mobilization. We performed a search on PubMed for articles related to these aspects in a wide range of basic studies. Together, numerous evidences hold the promise that endogenous SCs would be a novel therapeutic approach for the therapy of ED.
Collapse
Affiliation(s)
- Zhong-Cheng Xin
- Andrology Center, Peking University First Hospital, Peking University, Beijing 100034, USA
| | | | | | | | | |
Collapse
|
24
|
Vascular Transdifferentiation in the CNS: A Focus on Neural and Glioblastoma Stem-Like Cells. Stem Cells Int 2016; 2016:2759403. [PMID: 27738435 PMCID: PMC5055959 DOI: 10.1155/2016/2759403] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 09/05/2016] [Indexed: 01/12/2023] Open
Abstract
Glioblastomas are devastating and extensively vascularized brain tumors from which glioblastoma stem-like cells (GSCs) have been isolated by many groups. These cells have a high tumorigenic potential and the capacity to generate heterogeneous phenotypes. There is growing evidence to support the possibility that these cells are derived from the accumulation of mutations in adult neural stem cells (NSCs) as well as in oligodendrocyte progenitors. It was recently reported that GSCs could transdifferentiate into endothelial-like and pericyte-like cells both in vitro and in vivo, notably under the influence of Notch and TGFβ signaling pathways. Vascular cells derived from GBM cells were also observed directly in patient samples. These results could lead to new directions for designing original therapeutic approaches against GBM neovascularization but this specific reprogramming requires further molecular investigations. Transdifferentiation of nontumoral neural stem cells into vascular cells has also been described and conversely vascular cells may generate neural stem cells. In this review, we present and discuss these recent data. As some of them appear controversial, further validation will be needed using new technical approaches such as high throughput profiling and functional analyses to avoid experimental pitfalls and misinterpretations.
Collapse
|
25
|
Xiao C, Qian M, Yin C, Zhang Y, Hu H, Yao S. A zebrafish mosaic assay to study mammalian stem cells in real time in vivo. J Mol Histol 2016; 47:437-44. [PMID: 27554369 DOI: 10.1007/s10735-016-9688-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/30/2016] [Indexed: 02/05/2023]
Abstract
The differentiation potentials of stem cells have been evaluated by various in vivo and in vitro assays. However, these assays have different limitations hindering efficient study of mammalian stem cells. Here we describe a rapid and powerful mosaic assay to study the differentiation potentials of stem cells in real time in vivo by using zebrafish embryo. We transplanted mouse neural stem cells into zebrafish embryos at different developmental stages and found that they mainly formed neural tissues while occasionally trans-differentiated into mesoderm- and endoderm-derived tissues. Because zebrafish embryo is transparent, the behaviors of transplanted mouse stem cells can be easily tracked in a real-time manner and at single-cell resolution. We expect that this assay may be widely applied to explore the in vivo behaviors of any stem cells available.
Collapse
Affiliation(s)
- Chun Xiao
- College of life Science and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Meilin Qian
- College of life Science and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chaoran Yin
- College of life Science and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yonggang Zhang
- College of life Science and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Huozhen Hu
- College of life Science and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Shaohua Yao
- College of life Science and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Ledesma-Martínez E, Mendoza-Núñez VM, Santiago-Osorio E. Mesenchymal Stem Cells Derived from Dental Pulp: A Review. Stem Cells Int 2015; 2016:4709572. [PMID: 26779263 PMCID: PMC4686712 DOI: 10.1155/2016/4709572] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/09/2015] [Indexed: 01/09/2023] Open
Abstract
The mesenchymal stem cells of dental pulp (DPSCs) were isolated and characterized for the first time more than a decade ago as highly clonogenic cells that were able to generate densely calcified colonies. Now, DPSCs are considered to have potential as stem cell source for orthopedic and oral maxillofacial reconstruction, and it has been suggested that they may have applications beyond the scope of the stomatognathic system. To date, most studies have shown that, regardless of their origin in third molars, incisors, or exfoliated deciduous teeth, DPSCs can generate mineralized tissue, an extracellular matrix and structures type dentin, periodontal ligament, and dental pulp, as well as other structures. Different groups worldwide have designed and evaluated new efficient protocols for the isolation, expansion, and maintenance of clinically safe human DPSCs in sufficient numbers for various therapeutics protocols and have discussed the most appropriate route of administration, the possible contraindications to their clinical use, and the parameters to be considered for monitoring their clinical efficacy and proper biological source. At present, DPSC-based therapy is promising but because most of the available evidence was obtained using nonhuman xenotransplants, it is not a mature technology.
Collapse
Affiliation(s)
- Edgar Ledesma-Martínez
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, MEX, Mexico
| | - Víctor Manuel Mendoza-Núñez
- Research Unit on Gerontology, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, MEX, Mexico
| | - Edelmiro Santiago-Osorio
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, 09230 Mexico City, MEX, Mexico
| |
Collapse
|
27
|
La Colla A, Pronsato L, Milanesi L, Vasconsuelo A. 17β-Estradiol and testosterone in sarcopenia: Role of satellite cells. Ageing Res Rev 2015; 24:166-77. [PMID: 26247846 DOI: 10.1016/j.arr.2015.07.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 07/24/2015] [Accepted: 07/30/2015] [Indexed: 12/25/2022]
Abstract
The loss of muscle mass and strength with aging, referred to as sarcopenia, is a prevalent condition among the elderly. Although the molecular mechanisms underlying sarcopenia are unclear, evidence suggests that an age-related acceleration of myocyte loss via apoptosis might be responsible for muscle perfomance decline. Interestingly, sarcopenia has been associated to a deficit of sex hormones which decrease upon aging. The skeletal muscle ability to repair and regenerate itself would not be possible without satellite cells, a subpopulation of cells that remain quiescent throughout life. They are activated in response to stress, enabling them to guide skeletal muscle regeneration. Thus, these cells could be a key factor to overcome sarcopenia. Of importance, satellite cells are 17β-estradiol (E2) and testosterone (T) targets. In this review, we summarize potential mechanisms through which these hormones regulate satellite cells activation during skeletal muscle regeneration in the elderly. The advance in its understanding will help to the development of potential therapeutic agents to alleviate and treat sarcopenia and other related myophaties.
Collapse
|
28
|
Sienkiewicz D, Kulak W, Okurowska-Zawada B, Paszko-Patej G, Kawnik K. Duchenne muscular dystrophy: current cell therapies. Ther Adv Neurol Disord 2015; 8:166-77. [PMID: 26136844 PMCID: PMC4480531 DOI: 10.1177/1756285615586123] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Duchenne muscular dystrophy is a genetically determined X-linked disease and the most common, progressive pediatric muscle disorder. For decades, research has been conducted to find an effective therapy. This review presents current therapeutic methods for Duchenne muscular dystrophy, based on scientific articles in English published mainly in the period 2000 to 2014. We used the PubMed database to identify and review the most important studies. An analysis of contemporary studies of stem cell therapy and the use of granulocyte colony-stimulating factor (G-CSF) in muscular dystrophy was performed.
Collapse
Affiliation(s)
- Dorota Sienkiewicz
- Department of Pediatric Rehabilitation Medical University, Bialystok, Poland
| | - Wojciech Kulak
- Department of Pediatric Rehabilitation, Medical University of Białystok, 15-274 Bialystok, 17 Waszyngtona street, Poland
| | | | | | - Katarzyna Kawnik
- Department of Pediatric Rehabilitation Medical University, Bialystok, Poland
| |
Collapse
|
29
|
Videla Richardson GA, Garcia CP, Roisman A, Slavutsky I, Fernandez Espinosa DD, Romorini L, Miriuka SG, Arakaki N, Martinetto H, Scassa ME, Sevlever GE. Specific Preferences in Lineage Choice and Phenotypic Plasticity of Glioma Stem Cells Under BMP4 and Noggin Influence. Brain Pathol 2015; 26:43-61. [PMID: 25808628 DOI: 10.1111/bpa.12263] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 03/12/2015] [Indexed: 12/23/2022] Open
Abstract
Although BMP4-induced differentiation of glioma stem cells (GSCs) is well recognized, details of the cellular responses triggered by this morphogen are still poorly defined. In this study, we established several GSC-enriched cell lines (GSC-ECLs) from high-grade gliomas. The expansion of these cells as adherent monolayers, and not as floating neurospheres, enabled a thorough study of the phenotypic changes that occurred during their differentiation. Herein, we evaluated GSC-ECLs' behavior toward differentiating conditions by depriving them of growth factors and/or by adding BMP4 at different concentrations. After analyzing cellular morphology, proliferation and lineage marker expression, we determined that GSC-ECLs have distinct preferences in lineage choice, where some of them showed an astrocyte fate commitment and others a neuronal one. We found that this election seems to be dictated by the expression pattern of BMP signaling components present in each GSC-ECL. Additionally, treatment of GSC-ECLs with the BMP antagonist, Noggin, also led to evident phenotypic changes. Interestingly, under certain conditions, some GSC-ECLs adopted an unexpected smooth muscle-like phenotype. As a whole, our findings illustrate the wide differentiation potential of GSCs, highlighting their molecular complexity and paving a way to facilitate personalized differentiating therapies.
Collapse
Affiliation(s)
- Guillermo Agustín Videla Richardson
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Escobar, Provincia de Buenos Aires, Argentina
| | - Carolina Paola Garcia
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Escobar, Provincia de Buenos Aires, Argentina
| | - Alejandro Roisman
- Laboratorio de Neuropatología, Departamento de Neuropatología y Biología Molecular, Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | - Irma Slavutsky
- Laboratorio de Neuropatología, Departamento de Neuropatología y Biología Molecular, Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | - Damián Darío Fernandez Espinosa
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Escobar, Provincia de Buenos Aires, Argentina
| | - Leonardo Romorini
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Escobar, Provincia de Buenos Aires, Argentina
| | - Santiago Gabriel Miriuka
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Escobar, Provincia de Buenos Aires, Argentina
| | - Naomi Arakaki
- Laboratorio de Biología Molecular, Departamento de Neuropatología y Biología Molecular, Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | - Horacio Martinetto
- Laboratorio de Genética de Neoplasias Linfoides, Instituto de Medicina Experimental, CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - María Elida Scassa
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Escobar, Provincia de Buenos Aires, Argentina
| | - Gustavo Emilio Sevlever
- Laboratorio de Biología Molecular, Departamento de Neuropatología y Biología Molecular, Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| |
Collapse
|
30
|
Pan S, Chen W, Liu X, Xiao J, Wang Y, Liu J, Du Y, Wang Y, Zhang Y. Application of a novel population of multipotent stem cells derived from skin fibroblasts as donor cells in bovine SCNT. PLoS One 2015; 10:e0114423. [PMID: 25602959 PMCID: PMC4300223 DOI: 10.1371/journal.pone.0114423] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 11/10/2014] [Indexed: 01/26/2023] Open
Abstract
Undifferentiated stem cells are better donor cells for somatic cell nuclear transfer (SCNT), resulting in more offspring than more differentiated cells. While various stem cell populations have been confirmed to exist in the skin, progress has been restricted due to the lack of a suitable marker for their prospective isolation. To address this fundamental issue, a marker is required that could unambiguously prove the differentiation state of the donor cells. We therefore utilized magnetic activated cell sorting (MACS) to separate a homogeneous population of small SSEA-4+ cells from a heterogeneous population of bovine embryonic skin fibroblasts (BEF). SSEA-4+ cells were 8-10 μm in diameter and positive for alkaline phosphatase (AP). The percentage of SSEA-4+ cells within the cultured BEF population was low (2-3%). Immunocytochemistry and PCR analyses revealed that SSEA-4+ cells expressed pluripotency-related markers, and could differentiate into cells comprising all three germ layers in vitro. They remained undifferentiated over 20 passages in suspension culture. In addition, cloned embryos derived from SSEA-4 cells showed significant differences in cleavage rate and blastocyst development when compared with those from BEF and SSEA-4− cells. Moreover, blastocysts derived from SSEA-4+ cells showed a higher total cell number and lower apoptotic index as compared to BEF and SSEA-4– derived cells. It is well known that nuclei from pluripotent stem cells yield a higher cloning efficiency than those from adult somatic cells, however, pluripotent stem cells are relatively difficult to obtain from bovine. The SSEA-4+ cells described in the current study provide an attractive candidate for SCNT and a promising platform for the generation of transgenic cattle.
Collapse
Affiliation(s)
- Shaohui Pan
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Wuju Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Xu Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Jiajia Xiao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Yanqin Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Jun Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Yue Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Yongsheng Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail: (YW); (YZ)
| | - Yong Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail: (YW); (YZ)
| |
Collapse
|
31
|
Multistage Adipose-Derived Stem Cell Myogenesis: An Experimental and Modeling Study. Cell Mol Bioeng 2014. [DOI: 10.1007/s12195-014-0362-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
32
|
Fox IJ, Daley GQ, Goldman SA, Huard J, Kamp TJ, Trucco M. Stem cell therapy. Use of differentiated pluripotent stem cells as replacement therapy for treating disease. Science 2014; 345:1247391. [PMID: 25146295 PMCID: PMC4329726 DOI: 10.1126/science.1247391] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pluripotent stem cells (PSCs) directed to various cell fates holds promise as source material for treating numerous disorders. The availability of precisely differentiated PSC-derived cells will dramatically affect blood component and hematopoietic stem cell therapies and should facilitate treatment of diabetes, some forms of liver disease and neurologic disorders, retinal diseases, and possibly heart disease. Although an unlimited supply of specific cell types is needed, other barriers must be overcome. This review of the state of cell therapies highlights important challenges. Successful cell transplantation will require optimizing the best cell type and site for engraftment, overcoming limitations to cell migration and tissue integration, and occasionally needing to control immunologic reactivity, as well as a number of other challenges. Collaboration among scientists, clinicians, and industry is critical for generating new stem cell-based therapies.
Collapse
Affiliation(s)
- Ira J Fox
- Department of Surgery, Children's Hospital of Pittsburgh and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - George Q Daley
- Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA. Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Broad Institute, Cambridge, MA, USA. Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, The University of Rochester Medical Center, Rochester, NY, USA. Center for Basic and Translational Neuroscience, University of Copenhagen, Denmark
| | - Johnny Huard
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Timothy J Kamp
- Stem Cell and Regenerative Medicine Center, Cellular and Molecular Arrhythmia Research Program, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Massimo Trucco
- Division of Immunogenetics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
33
|
Abstract
Perinatal brain injuries are a leading cause of cerebral palsy worldwide. The potential of stem cell therapy to prevent or reduce these impairments has been widely discussed within the medical and scientific communities and an increasing amount of research is being conducted in this field. Animal studies support the idea that a number of stem cells types, including cord blood and mesenchymal stem cells have a neuroprotective effect in neonatal hypoxia-ischemia. Both these cell types are readily available in a clinical setting. The mechanisms of action appear to be diverse, including immunomodulation, activation of endogenous stem cells, release of growth factors, and anti-apoptotic effects. Here, we review the different types of stem cells and progenitor cells that are potential candidates for therapeutic strategies in perinatal brain injuries, and summarize recent preclinical and clinical studies.
Collapse
Affiliation(s)
- Andre W Phillips
- The Hugo W. Moser Research Institute at Kennedy Krieger Institute Johns Hopkins University, Baltimore, Maryland, USA ; Department of Neurology Johns Hopkins University, Baltimore, Maryland, USA
| | | | | |
Collapse
|
34
|
Shoshani O, Zipori D. Stress as a fundamental theme in cell plasticity. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:371-7. [PMID: 25038585 DOI: 10.1016/j.bbagrm.2014.07.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/08/2014] [Accepted: 07/09/2014] [Indexed: 01/16/2023]
Abstract
Over a decade of intensive investigation of the possible plasticity of mammalian cells has eventually substantiated that mammalian species are endowed with a remarkable capacity to change mature cell fates. We review below the evidence for the occurrence of processes such as dedifferentiation and transdifferentiation within mammalian tissues in vivo, and in cells removed from their protective microenvironment and seeded in culture under conditions poorly resembling their physiological state in situ. Overall, these studies point to one major conclusion: stressful conditions, whether due to in vivo tissue damage or otherwise to isolation of cells from their in vivo restrictive niches, lead to extreme fate changes. Some examples of dedifferentiation are discussed in detail showing that rare cells within the population tend to turn back into less mature ones due to severe cell damage. It is proposed that cell stress, mechanistically sensed by isolation from neighboring cells, leads to dedifferentiation, in an attempt to build a new stem cell reservoir for subsequent regeneration of the damaged tissue. This article is part of a Special Issue entitled: Stress as a fundamental theme in cell plasticity.
Collapse
Affiliation(s)
- Ofer Shoshani
- Department of Cellular and Molecular Medicine, Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA, USA
| | - Dov Zipori
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
35
|
Nguyen TD, Widera D, Greiner J, Müller J, Martin I, Slotta C, Hauser S, Kaltschmidt C, Kaltschmidt B. Prolonged cultivation of hippocampal neural precursor cells shifts their differentiation potential and selects for aneuploid cells. Biol Chem 2014; 394:1623-36. [PMID: 24084358 DOI: 10.1515/hsz-2013-0191] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 09/27/2013] [Indexed: 11/15/2022]
Abstract
Neural precursor cells (NPCs) are lineage-restricted neural stem cells with limited self-renewal, giving rise to a broad range of neural cell types such as neurons, astrocytes, and oligodendrocytes. Despite this developmental potential, the differentiation capacity of NPCs has been controversially discussed concerning the trespassing lineage boundaries, for instance resulting in hematopoietic competence. Assessing their in vitro plasticity, we isolated nestin+/Sox2+, NPCs from the adult murine hippocampus. In vitro-expanded adult NPCs were able to form neurospheres, self-renew, and differentiate into neuronal, astrocytic, and oligodendrocytic cells. Although NPCs cultivated in early passage efficiently gave rise to neuronal cells in a directed differentiation assay, extensively cultivated NPCs revealed reduced potential for ectodermal differentiation. We further observed successful differentiation of long-term cultured NPCs into osteogenic and adipogenic cell types, suggesting that NPCs underwent a fate switch during culture. NPCs cultivated for more than 12 passages were aneuploid (abnormal chromosome numbers such as 70 chromosomes). Furthermore, they showed growth factor-independent proliferation, a hallmark of tumorigenic transformation. In conclusion, our findings substantiate the lineage restriction of NPCs from adult mammalian hippocampus. Prolonged cultivation results, however, in enhanced differentiation potential, which may be attributed to transformation events leading to aneuploid cells.
Collapse
|
36
|
Is carcinoma a mesenchymal disease? The role of the stromal microenvironment in carcinogenesis. Pathology 2013; 45:371-81. [PMID: 23594691 DOI: 10.1097/pat.0b013e328360b600] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Most research into the biology of carcinoma has focused on the epithelial cells therein; the inherent assumption has been that the tumour arises from epithelial cells 'gone bad', and that the surrounding stroma is simply an 'innocent bystander'. However, there is increasing evidence that there is a complex interplay between tumour cells and their surrounding microenvironment, and that the latter may be just as important in determining the development and clinical behaviour of a given tumour. Similarly, traditional oncological practice has been predominantly aimed at a perceived ideal goal of killing all the tumour epithelial cells, with only a few recently developed therapies seeking to affect other components (such as tumour vasculature); but identifying stromal factors involved in tumour growth and survival may well lead to the development of novel therapies. This review examines current understanding of the interplay between tumour epithelial cells and their microenvironment, and enumerates various stromal factors which appear to play a role in tumour progression and/or metastasis.
Collapse
|
37
|
Simon L, Cooke PS, Berry SE. Aorta-derived mesoangioblasts can be differentiated into functional uterine epithelium, but not prostatic epithelium or epidermis, by instructive mesenchymes. Cells Tissues Organs 2013; 198:169-78. [PMID: 24192012 DOI: 10.1159/000354900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2013] [Indexed: 11/19/2022] Open
Abstract
Mesoangiobasts are blood vessel-derived stem cells that differentiate into smooth, skeletal, and cardiac muscle cells. We have reported that postnatal aorta-derived mesoangioblasts (ADM) regenerate skeletal muscle and prevent onset of dilated cardiomyopathy in animal models of Duchenne muscular dystrophy. ADM also differentiate into myelinating glial cells, suggesting they are multipotent and capable of generating mesodermal or ectodermal derivatives. Mesenchyme of some fetal organs is a potent instructive inducer. Here we examined whether ADM can differentiate into prostatic, uterine, and skin epithelium by recombining ADM with fetal or neonatal mesenchyme from these organs and grafting them under the renal capsule of syngeneic hosts. In tissue recombinants of uterine mesenchyme (UtM) and ADM, ADM formed histologically normal simple columnar uterine epithelium that expressed estrogen receptor 1 and in response to estrogen showed increased mitogenesis and downregulation of progesterone receptor. In contrast, ADM did not differentiate into prostatic epithelium or epidermis when recombined with urogenital sinus mesenchyme or fetal dermis, respectively. These results indicate that ADM can respond to cues from neonatal UtM and differentiate into morphologically and functionally normal uterine epithelial cells, and support previous reports that ADM can differentiate into a variety of tissues of the mesodermal lineage. However, these data indicate that ADM are restricted in their capacity to differentiate into endodermal and ectodermal derivatives such as prostatic and skin epithelial cells, respectively.
Collapse
Affiliation(s)
- Liz Simon
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, La., USA
| | | | | |
Collapse
|
38
|
Isolation of multilineage progenitors from mouse brain. In Vitro Cell Dev Biol Anim 2013; 49:307-14. [PMID: 23636940 DOI: 10.1007/s11626-013-9625-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 04/18/2013] [Indexed: 10/26/2022]
Abstract
Stem cells are unique cell populations with the ability to undergo self-renewal and differentiation. These cells have been identified in a wide range of tissues and possess varied differentiation potentials. Tissue-specific stem cells have typically been thought to have limited differentiation capabilities. We show here that fibroblast-like cells isolated from mouse brain possess cross-germ layer differentiation abilities. These cells were found to express typical mesenchymal stem cell markers (CD44, CD29, and CD105) and were able to be passaged more than 50 times. When treated under defined conditions, the brain-derived cells were able to generate many different cell types including adipocytes, osteocytes, astrocytes, neurons, and even hepatocyte-like cells. The hepatocyte-like cells not only expressed liver cell-specific markers, but also exhibited the capacity for glycogen storage and low-density lipoprotein uptake. These results demonstrate the existence of cells in the brain with three-germ-layer differentiation potential.
Collapse
|
39
|
Salerno M, Avnet S, Bonuccelli G, Eramo A, De Maria R, Gambarotti M, Gamberi G, Baldini N. Sphere-forming cell subsets with cancer stem cell properties in human musculoskeletal sarcomas. Int J Oncol 2013; 43:95-102. [PMID: 23636271 DOI: 10.3892/ijo.2013.1927] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 03/04/2013] [Indexed: 11/05/2022] Open
Abstract
Musculoskeletal sarcomas are aggressive malignancies often characterized by an adverse prognosis despite the use of intense multiagent chemotherapy or molecular targeted therapy in combination to surgery and radiotherapy. Stem-like cells identified within solid tumors have been recently implicated in drug resistance, metastasis and local relapse. Here, we report the identification of putative cancer stem cells (CSCs) in sarcomas using a sphere culture system. These sarcospheres, able to grow in anchorage-independent and serum-starved conditions, express the pluripotent embryonic stem cell marker genes OCT3/4, Nanog and SOX2. Expression levels of these genes were greater in sarcospheres than in the parental tumor cultures. Importantly, the isolated tumor spheres transplanted into mice were tumorigenic and capable of recapitulating the human disease. Finally, we demonstrated that low (1%) O2 conditions, reproducing those found within the tumor microenvironment, significantly increase the number and the size of sarcospheres. The sphere formation assay is, therefore, a valuable method for the isolation of putative CSCs from human sarcomas and its efficiency is improved by controlling oxygen availability. This method provides a reliable preclinical model that can be used for future studies aimed at investigating crucial aspects of sarcoma biology, such as resistance to treatments and relapse.
Collapse
Affiliation(s)
- Manuela Salerno
- Department of Orthopaedic Pathophysiology and Regenerative Medicine, Istituto Ortopedico Rizzoli, Bologna, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Manzar N, Manzar B, Hussain N, Hussain MFA, Raza S. The ethical dilemma of embryonic stem cell research. SCIENCE AND ENGINEERING ETHICS 2013; 19:97-106. [PMID: 22038063 DOI: 10.1007/s11948-011-9326-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 10/18/2011] [Indexed: 05/31/2023]
Abstract
To determine the knowledge, attitude, and ethical concerns of medical students and graduates with regard to Embryonic Stem Cell (ESC) research. This questionnaire based descriptive study was conducted at the Civil Hospital Karachi (CHK), Pakistan from February to July 2008. A well structured questionnaire was administered to medical students and graduate doctors, which included their demographic profile as well as questions in line with the study objective. Informed consent was taken and full confidentiality was assured to the participants. Data were entered in a Statistical Package for Social Sciences (SPSS version.12) and analyzed. A total of 204 male and 216 female medical students and doctors were administered questionnaires out of which 105 males (51.4%) and 108 females (50%) were aware of the embryonic stem cell research and its ethical implications. Forty percent males and 47% of females were of the opinion that life begins at conception. Forty-six percent males and 39% females were in favor of stem cell research while only 31% males and 28% females supported the ESC research. Less than 1/3 of students supported using frozen embryos for research purposes while more than 2/3 indicated that they were unlikely to support abortion for stem cell research purposes. The majority of the students were in favor of stem cell research with some reservations regarding ESC research. A sizeable number of students withheld their views, reflecting their poor understanding of medical ethics. The result of the study indicates a need for incorporating bioethics into the medical curriculum.
Collapse
Affiliation(s)
- Nabeel Manzar
- Dow University of Health Sciences, Karachi, Pakistan.
| | | | | | | | | |
Collapse
|
41
|
Masaki T, Qu J, Cholewa-Waclaw J, Burr K, Raaum R, Rambukkana A. Reprogramming adult Schwann cells to stem cell-like cells by leprosy bacilli promotes dissemination of infection. Cell 2013; 152:51-67. [PMID: 23332746 PMCID: PMC4314110 DOI: 10.1016/j.cell.2012.12.014] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 10/31/2012] [Accepted: 12/10/2012] [Indexed: 01/09/2023]
Abstract
Differentiated cells possess a remarkable genomic plasticity that can be manipulated to reverse or change developmental commitments. Here, we show that the leprosy bacterium hijacks this property to reprogram adult Schwann cells, its preferred host niche, to a stage of progenitor/stem-like cells (pSLC) of mesenchymal trait by downregulating Schwann cell lineage/differentiation-associated genes and upregulating genes mostly of mesoderm development. Reprogramming accompanies epigenetic changes and renders infected cells highly plastic, migratory, and immunomodulatory. We provide evidence that acquisition of these properties by pSLC promotes bacterial spread by two distinct mechanisms: direct differentiation to mesenchymal tissues, including skeletal and smooth muscles, and formation of granuloma-like structures and subsequent release of bacteria-laden macrophages. These findings support a model of host cell reprogramming in which a bacterial pathogen uses the plasticity of its cellular niche for promoting dissemination of infection and provide an unexpected link between cellular reprogramming and host-pathogen interaction.
Collapse
Affiliation(s)
- Toshihiro Masaki
- MRC Center for Regenerative Medicine, University of Edinburgh, Little France Campus, Edinburgh, EH16 4SB, Scotland, UK,Center for Neuroregeneration, University of Edinburgh, Little France Campus, Edinburgh, EH16 4SB, Scotland, UK,The Rockefeller University, York Avenue, New York, NY 10065, USA
| | - Jinrong Qu
- The Rockefeller University, York Avenue, New York, NY 10065, USA
| | - Justyna Cholewa-Waclaw
- MRC Center for Regenerative Medicine, University of Edinburgh, Little France Campus, Edinburgh, EH16 4SB, Scotland, UK,Center for Neuroregeneration, University of Edinburgh, Little France Campus, Edinburgh, EH16 4SB, Scotland, UK
| | - Karen Burr
- Center for Neuroregeneration, University of Edinburgh, Little France Campus, Edinburgh, EH16 4SB, Scotland, UK
| | - Ryan Raaum
- The Rockefeller University, York Avenue, New York, NY 10065, USA
| | - Anura Rambukkana
- MRC Center for Regenerative Medicine, University of Edinburgh, Little France Campus, Edinburgh, EH16 4SB, Scotland, UK,Center for Neuroregeneration, University of Edinburgh, Little France Campus, Edinburgh, EH16 4SB, Scotland, UK,Center for Infectious Diseases, University of Edinburgh, Little France Campus, Edinburgh, EH16 4SB, Scotland, UK,The Rockefeller University, York Avenue, New York, NY 10065, USA,Correspondence: (A.R), Telephone: +44(0) 131-651-9565, Fax: +44(0) 131-651-9501
| |
Collapse
|
42
|
Gullapalli VK, Khodair MA, Wang H, Sugino IK, Madreperla S, Zarbin MA. Transplantation Frontiers. Retina 2013. [DOI: 10.1016/b978-1-4557-0737-9.00125-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
43
|
Kazanis I. Neurogenesis in the adult mammalian brain: how much do we need, how much do we have? Curr Top Behav Neurosci 2013; 15:3-29. [PMID: 22976273 DOI: 10.1007/7854_2012_227] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The last two decades cytogenic processes (both neurogenic and gliogenic) driven by neural stem cells surviving within the adult mammalian brain have been extensively investigated. It is now well established that within at least two cytogenic niches, the subependymal zone of the lateral ventricles and the subgranular zone in the dentate gyrus, new neurons are born everyday with a fraction of them being finally incorporated into established neuronal networks in the olfactory bulb and the hippocampus, respectively. But how significant is adult neurogenesis in the context of the mature brain and what are the possibilities that these niches can contribute significantly in tissue repair after degenerative insults, or in the restoration of normal hippocampal function in the context of mental and cognitive disorders? Here, we summarise the available data on the normal behaviour of adult neural stem cells in the young and the aged brain and on their response to degeneration. Focus will be given, whenever possible, to numbers: how many stem cells survive in the adult brain, how many cells they can generate and at what ratios do they produce neurons and glia?
Collapse
Affiliation(s)
- Ilias Kazanis
- MRC Cambridge Centre for Stem cell Biology and Regenerative Medicine and Department of Veterinary Medicine, University of Cambridge, Madingley Road, CB3 0ES, Cambridge, UK,
| |
Collapse
|
44
|
Microenvironment-evoked cell lineage conversion: Shifting the focus from internal reprogramming to external forcing. Ageing Res Rev 2013; 12:29-38. [PMID: 22561469 DOI: 10.1016/j.arr.2012.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Revised: 03/28/2012] [Accepted: 04/18/2012] [Indexed: 01/02/2023]
Abstract
Seeking possible ways to create replacement cells for the faded ones with deficits in functionality or quantity inspires comprehensive needs for cell lineage conversion. To fulfill this promise, reprogramming and microenvironment direction have been used to manipulate abundant cell fates. We briefly describe the evolution and fundamental insights of these two major strategies applied for lineage specification, comment generally on their current limitations, and analyze the orchestral interplay between them. We also present several future directions and discuss the potential clinical uses. Based on the relatively slight safety and technical issues, we conclude that microenvironment-evoked cell lineage conversion, instead of reprogramming, will be the shifting focus in regenerative medicine.
Collapse
|
45
|
Ulrich H, Abbracchio MP, Burnstock G. Extrinsic purinergic regulation of neural stem/progenitor cells: implications for CNS development and repair. Stem Cell Rev Rep 2012; 8:755-67. [PMID: 22544361 DOI: 10.1007/s12015-012-9372-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
There has been tremendous progress in understanding neural stem cell (NSC) biology, with genetic and cell biological methods identifying sequential gene expression and molecular interactions guiding NSC specification into distinct neuronal and glial populations during development. Data has emerged on the possible exploitation of NSC-based strategies to repair adult diseased brain. However, despite increased information on lineage specific transcription factors, cell-cycle regulators and epigenetic factors involved in the fate and plasticity of NSCs, understanding of extracellular cues driving the behavior of embryonic and adult NSCs is still very limited. Knowledge of factors regulating brain development is crucial in understanding the pathogenetic mechanisms of brain dysfunction. Since injury-activated repair mechanisms in adult brain often recapitulate ontogenetic events, the identification of these players will also reveal novel regenerative strategies. Here, we highlight the purinergic system as a key emerging player in the endogenous control of NSCs. Purinergic signalling molecules (ATP, UTP and adenosine) act with growth factors in regulating the synchronized proliferation, migration, differentiation and death of NSCs during brain and spinal cord development. At early stages of development, transient and time-specific release of ATP is critical for initiating eye formation; once anatomical CNS structures are defined, purinergic molecules participate in calcium-dependent neuron-glia communication controlling NSC behaviour. When development is complete, some purinergic mechanisms are silenced, but can be re-activated in adult brain after injury, suggesting a role in regeneration and self-repair. Targeting the purinergic system to develop new strategies for neurodevelopmental disorders and neurodegenerative diseases will be also discussed.
Collapse
Affiliation(s)
- Henning Ulrich
- Departamento de Bioquimica, Instituto de Quimica, Universidade de São Paulo, Av. Prof. Lineu Prestes, 748, São Paulo 05508-900, SP, Brazil.
| | | | | |
Collapse
|
46
|
Transdifferentiation of glioblastoma stem-like cells into mural cells drives vasculogenic mimicry in glioblastomas. J Neurosci 2012; 32:12950-60. [PMID: 22973019 DOI: 10.1523/jneurosci.2017-12.2012] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Recent evidence has shown that glioblastoma stem-like cells (GSCs) can transdifferentiate into endothelial cells and vascular-like tumor cells. The latter pattern of vascularization indicates an alternative microvascular circulation known as vasculogenic mimicry (VM). However, it remains to be clarified how the GSC-driven VM makes a significant contribution to tumor vasculature. Here, we investigated 11 cases of glioblastomas and found that most of them consisted of blood-perfused vascular channels that coexpress mural cell markers smooth muscle α-actin and platelet-derived growth factor receptor β, epidermal growth factor receptor, and vascular endothelial growth factor receptor 2 (Flk-1), but not CD31 or VE-cadherin. This microvasculature coexisted with endothelial cell-associated vessels. GSCs derived from patients with glioblastomas developed vigorous mural cell-associated vascular channels but few endothelial cell vessels in orthotopic animal models. Suppression of Flk-1 activity and gene expression abrogated GSC transdifferentiation and vascularization in vitro, and inhibited VM in animal models. This study establishes mural-like tumor cells differentiated from GSCs as a significant contributor to microvasculature of glioblastoma and points to Flk-1 as a potential target for therapeutic intervention that could complement current anti-angiogenic treatment.
Collapse
|
47
|
Zeng X, Zhang L, Sun L, Zhang D, Zhao H, Jia J, Wang W. Recovery from rat sciatic nerve injury in vivo through the use of differentiated MDSCs in vitro. Exp Ther Med 2012; 5:193-196. [PMID: 23251266 PMCID: PMC3523948 DOI: 10.3892/etm.2012.785] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 09/18/2012] [Indexed: 01/13/2023] Open
Abstract
In this study, muscle-derived stem cells (MDSCs) whose differentiation into neuron-like cells was induced by ciliary neurotrophic factor (CNTF) and Salvia (Salvia miltiorrhiza) in vitro were used to repair rat sciatic nerve injuries in vivo, in order to investigate their multifunctional characteristics as pluripotent stem cells. The sciatic nerve in the right side of the lower limb was exposed under the anesthetized condition of 10% chloral hydrate (0.3 ml/100 g) injection into the abdominal cavity. The tissue which was 0.5 cm above the sciatic nerve bifurcation was broken using a hemostat. After induction, MDSCs were transferred in sodium hyaluronate gel and were placed into the damaged area. An untreated control group was also included in this study. The surgical area was sutured after washing with gentamycin sulfate solution. Sciatic nerve function index (SFI) was calculated, electrophysiological tests were performed and the recovery rate of gastrocnemius muscle wet weight was also calculated. Four weeks post-surgery, the SFI and the recovery rate of gastrocnemius muscle wet weight in the MDSC group were significantly higher than those in the control group (P<0.05). MDSCs whose differentiation is induced by CNTF and Salvia play an active role in the repair of peripheral nerve injury.
Collapse
Affiliation(s)
- Xiangyi Zeng
- The Third Affiliated Hospital of Liaoning Medical University
| | | | | | | | | | | | | |
Collapse
|
48
|
Bai RY, Staedtke V, Lidov HG, Eberhart CG, Riggins GJ. OTX2 represses myogenic and neuronal differentiation in medulloblastoma cells. Cancer Res 2012; 72:5988-6001. [PMID: 22986744 DOI: 10.1158/0008-5472.can-12-0614] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The brain development transcription factor OTX2 is overexpressed and/or genomically amplified in most medulloblastomas, but the mechanistic basis for its contributions in this setting are not understood. In this study, we identified OTX2 as a transcriptional repressor and a gatekeeper of myogenic and neuronal differentiation in medulloblastoma cells. OTX2 binds to the MyoD1 core enhancer through its homeobox domain, and the remarkable repressor activity exhibited by the homeobox domain renders OTX2 transcriptionally repressive. RNA interference-mediated attenuation of OTX2 expression triggered myogenic and neuronal differentiation in vitro and prolonged the survival in an orthotopic medulloblastoma mouse model. Conversely, inducing myogenic conversion of medulloblastoma cells led to the loss of OTX2 expression. In medullomyoblastoma, a medulloblastoma subtype containing muscle elements, myogenic cells share cytogenetic signatures with the primitive tumor cells and OTX2 expression was lost in the differentiated myogenic cells. Thus, OTX2 functions via its homeobox domain as a suppressor of differentiation, and the loss of OTX2 expression is linked to the myogenesis in medullomyoblastoma. Together, our findings illustrate the origin of muscle cells in medullomyoblastomas and the oncogenic mechanism of OTX2 as a repressor of diverse differentiating potential.
Collapse
Affiliation(s)
- Ren-Yuan Bai
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA.
| | | | | | | | | |
Collapse
|
49
|
Zhang R, Jack GS, Rao N, Zuk P, Ignarro LJ, Wu B, Rodríguez LV. Nuclear fusion-independent smooth muscle differentiation of human adipose-derived stem cells induced by a smooth muscle environment. Stem Cells 2012; 30:481-90. [PMID: 22213158 DOI: 10.1002/stem.1023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human adipose-derived stem cells hASC have been isolated and were shown to have multilineage differentiation capacity. Although both plasticity and cell fusion have been suggested as mechanisms for cell differentiation in vivo, the effect of the local in vivo environment on the differentiation of adipose-derived stem cells has not been evaluated. We previously reported the in vitro capacity of smooth muscle differentiation of these cells. In this study, we evaluate the effect of an in vivo smooth muscle environment in the differentiation of hASC. We studied this by two experimental designs: (a) in vivo evaluation of smooth muscle differentiation of hASC injected into a smooth muscle environment and (b) in vitro evaluation of smooth muscle differentiation capacity of hASC exposed to bladder smooth muscle cells. Our results indicate a time-dependent differentiation of hASC into mature smooth muscle cells when these cells are injected into the smooth musculature of the urinary bladder. Similar findings were seen when the cells were cocultured in vitro with primary bladder smooth muscle cells. Chromosomal analysis demonstrated that microenvironment cues rather than nuclear fusion are responsible for this differentiation. We conclude that cell plasticity is present in hASCs, and their differentiation is accomplished in the absence of nuclear fusion.
Collapse
Affiliation(s)
- Rong Zhang
- Department of Urology, Sichuan University, Chengdu, Sichuan, China
| | | | | | | | | | | | | |
Collapse
|
50
|
Ma X, Zhang S, Zhou J, Chen B, Shang Y, Gao T, Wang X, Xie H, Chen F. Clone-derived human AF-amniotic fluid stem cells are capable of skeletal myogenic differentiation in vitro and in vivo. J Tissue Eng Regen Med 2012; 6:598-613. [PMID: 22396316 DOI: 10.1002/term.462] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 05/02/2011] [Accepted: 07/01/2011] [Indexed: 12/14/2022]
Affiliation(s)
| | | | | | | | | | | | - Xue Wang
- Department of Urology; Shanghai Children's Hospital; Shanghai Jiao Tong University School of Medicine; Shanghai; People's Republic of China
| | - Hua Xie
- Department of Urology; Shanghai Children's Hospital; Shanghai Jiao Tong University School of Medicine; Shanghai; People's Republic of China
| | | |
Collapse
|