1
|
Singh J, Singh S. Review on kidney diseases: types, treatment and potential of stem cell therapy. RENAL REPLACEMENT THERAPY 2023; 9:21. [PMID: 37131920 PMCID: PMC10134709 DOI: 10.1186/s41100-023-00475-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 04/11/2023] [Indexed: 05/04/2023] Open
Abstract
Renal disorders are an emerging global public health issue with a higher growth rate despite progress in supportive therapies. In order to find more promising treatments to stimulate renal repair, stem cell-based technology has been proposed as a potentially therapeutic option. The self-renewal and proliferative nature of stem cells raised the hope to fight against various diseases. Similarly, it opens a new path for the treatment and repair of damaged renal cells. This review focuses on the types of renal diseases; acute and chronic kidney disease-their statistical data, and the conventional drugs used for treatment. It includes the possible stem cell therapy mechanisms involved and outcomes recorded so far, the limitations of using these regenerative medicines, and the progressive improvement in stem cell therapy by adopting approaches like PiggyBac, Sleeping Beauty, and the Sendai virus. Specifically, about the paracrine activities of amniotic fluid stem cells, renal stem cells, embryonic stem cells, mesenchymal stem cell, induced pluripotent stem cells as well as other stem cells.
Collapse
Affiliation(s)
- Jaspreet Singh
- School of Bioengineering & Biosciences, Lovely Professional University, 15935, Block 56, Room No 202, Phagwara, Punjab 144411 India
| | - Sanjeev Singh
- School of Bioengineering & Biosciences, Lovely Professional University, 15935, Block 56, Room No 202, Phagwara, Punjab 144411 India
| |
Collapse
|
2
|
Sharp B, Rallabandi R, Devaux P. Advances in RNA Viral Vector Technology to Reprogram Somatic Cells: The Paramyxovirus Wave. Mol Diagn Ther 2022; 26:353-367. [PMID: 35763161 DOI: 10.1007/s40291-022-00599-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 11/24/2022]
Abstract
Ethical issues are a significant barrier to the use of embryonic stem cells in patients due to their origin: human embryos. To further the development of stem cells in a patient application, alternative sources of cells were sought. A process referred to as reprogramming was established to create induced pluripotent stem cells from somatic cells, resolving the ethical issues, and vectors were developed to deliver the reprogramming factors to generate induced pluripotent stem cells. Early viral vectors used integrating retroviruses and lentiviruses as delivery vehicles for the transcription factors required to initiate reprogramming. However, because of the inherent risk associated with vectors that integrate into the host genome, non-integrating approaches were explored. The development of non-integrating viral vectors offers a safer alternative, and these modern vectors are reliable, efficient, and easy to use to achieve induced pluripotent stem cells suitable for direct patient application in the growing field of individualized medicine. This review summarizes all the RNA viral vectors in the field of reprogramming with a special focus on the emerging delivery vectors based on non-integrating Paramyxoviruses, Sendai and measles viruses. We discuss their design and evolution towards being safe and efficient reprogramming vectors in generating induced pluripotent stem cells from somatic cells.
Collapse
Affiliation(s)
- Brenna Sharp
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ramya Rallabandi
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, MN, USA.,Regenerative Sciences Program, Mayo Clinic, Rochester, MN, USA
| | - Patricia Devaux
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA. .,Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, MN, USA. .,Regenerative Sciences Program, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
Metzler E, Escobar H, Sunaga-Franze DY, Sauer S, Diecke S, Spuler S. Generation of hiPSC-Derived Skeletal Muscle Cells: Exploiting the Potential of Skeletal Muscle-Derived hiPSCs. Biomedicines 2022; 10:1204. [PMID: 35625941 PMCID: PMC9138862 DOI: 10.3390/biomedicines10051204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 12/28/2022] Open
Abstract
Cell therapies for muscle wasting disorders are on the verge of becoming a realistic clinical perspective. Muscle precursor cells derived from human induced pluripotent stem cells (hiPSCs) represent the key to unrestricted cell numbers indispensable for the treatment of generalized muscle wasting such as cachexia or intensive care unit (ICU)-acquired weakness. We asked how the cell of origin influences efficacy and molecular properties of hiPSC-derived muscle progenitor cells. We generated hiPSCs from primary muscle stem cells and from peripheral blood mononuclear cells (PBMCs) of the same donors (n = 4) and compared their molecular profiles, myogenic differentiation potential, and ability to generate new muscle fibers in vivo. We show that reprogramming into hiPSCs from primary muscle stem cells was faster and 35 times more efficient than from blood cells. Global transcriptome comparison revealed significant differences, but differentiation into induced myogenic cells using a directed transgene-free approach could be achieved with muscle- and PBMC-derived hiPSCs, and both cell types generated new muscle fibers in vivo. Differences in myogenic differentiation efficiency were identified with hiPSCs generated from individual donors. The generation of muscle-stem-cell-derived hiPSCs is a fast and economic method to obtain unrestricted cell numbers for cell-based therapies in muscle wasting disorders, and in this aspect are superior to blood-derived hiPSCs.
Collapse
Affiliation(s)
- Eric Metzler
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; (H.E.); (D.Y.S.-F.); (S.S.); (S.D.)
- Experimental and Clinical Research Center, a Cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and Charité—Universitätsmedizin Berlin, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Helena Escobar
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; (H.E.); (D.Y.S.-F.); (S.S.); (S.D.)
- Experimental and Clinical Research Center, a Cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and Charité—Universitätsmedizin Berlin, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Daniele Yumi Sunaga-Franze
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; (H.E.); (D.Y.S.-F.); (S.S.); (S.D.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Genomics Platform, Hannoversche Straße 28, 10115 Berlin, Germany
| | - Sascha Sauer
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; (H.E.); (D.Y.S.-F.); (S.S.); (S.D.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Genomics Platform, Hannoversche Straße 28, 10115 Berlin, Germany
| | - Sebastian Diecke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; (H.E.); (D.Y.S.-F.); (S.S.); (S.D.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Pluripotent Stem Cells Platform, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Simone Spuler
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; (H.E.); (D.Y.S.-F.); (S.S.); (S.D.)
- Experimental and Clinical Research Center, a Cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and Charité—Universitätsmedizin Berlin, Lindenberger Weg 80, 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Lindenberger Weg 80, 13125 Berlin, Germany
| |
Collapse
|
4
|
Borgohain MP, Haridhasapavalan KK, Dey C, Adhikari P, Thummer RP. An Insight into DNA-free Reprogramming Approaches to Generate Integration-free Induced Pluripotent Stem Cells for Prospective Biomedical Applications. Stem Cell Rev Rep 2020; 15:286-313. [PMID: 30417242 DOI: 10.1007/s12015-018-9861-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
More than a decade ago, a pioneering study reported generation of induced Pluripotent Stem Cells (iPSCs) by ectopic expression of a cocktail of reprogramming factors in fibroblasts. This study has revolutionized stem cell research and has garnered immense interest from the scientific community globally. iPSCs hold tremendous potential for understanding human developmental biology, disease modeling, drug screening and discovery, and personalized cell-based therapeutic applications. The seminal study identified Oct4, Sox2, Klf4 and c-Myc as a potent combination of genes to induce reprogramming. Subsequently, various reprogramming factors were identified by numerous groups. Most of these studies have used integrating viral vectors to overexpress reprogramming factors in somatic cells to derive iPSCs. However, these techniques restrict the clinical applicability of these cells as they may alter the genome due to random viral integration resulting in insertional mutagenesis and tumorigenicity. To circumvent this issue, alternative integration-free reprogramming approaches are continuously developed that eliminate the risk of genomic modifications and improve the prospects of iPSCs from lab to clinic. These methods establish that integration of transgenes into the genome is not essential to induce pluripotency in somatic cells. This review provides a comprehensive overview of the most promising DNA-free reprogramming techniques that have the potential to derive integration-free iPSCs without genomic manipulation, such as sendai virus, recombinant proteins, microRNAs, synthetic messenger RNA and small molecules. The understanding of these approaches shall pave a way for the generation of clinical-grade iPSCs. Subsequently, these iPSCs can be differentiated into desired cell type(s) for various biomedical applications.
Collapse
Affiliation(s)
- Manash P Borgohain
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Krishna Kumar Haridhasapavalan
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Chandrima Dey
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Poulomi Adhikari
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India.
| |
Collapse
|
5
|
Zeng J, Li Y, Ma Z, Hu M. Advances in Small Molecules in Cellular Reprogramming: Effects, Structures, and Mechanisms. Curr Stem Cell Res Ther 2020; 16:115-132. [PMID: 32564763 DOI: 10.2174/1574888x15666200621172042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 11/22/2022]
Abstract
The method of cellular reprogramming using small molecules involves the manipulation of somatic cells to generate desired cell types under chemically limited conditions, thus avoiding the ethical controversy of embryonic stem cells and the potential hazards of gene manipulation. The combinations of small molecules and their effects on mouse and human somatic cells are similar. Several small molecules, including CHIR99021, 616452, A83-01, SB431542, forskolin, tranylcypromine and valproic acid [VPA], have been frequently used in reprogramming of mouse and human somatic cells. This indicated that the reprogramming approaches related to these compounds were essential. These approaches were mainly divided into four classes: epigenetic modification, signal modulation, metabolic modulation and senescent suppression. The structures and functions of small molecules involved in these reprogramming approaches have been studied extensively. Molecular docking gave insights into the mechanisms and structural specificities of various small molecules in the epigenetic modification. The binding modes of RG108, Bix01294, tranylcypromine and VPA with their corresponding proteins clearly illustrated the interactions between these compounds and the active sites of the proteins. Glycogen synthase kinase 3β [CHIR99021], transforming growth factor β [616452, A83-01 and SB431542] and protein kinase A [forskolin] signaling pathway play important roles in signal modulation during reprogramming, however, the mechanisms and structural specificities of these inhibitors are still unknown. Further, the numbers of small molecules in the approaches of metabolic modulation and senescent suppression were too few to compare. This review aims to serve as a reference for reprogramming through small molecules in order to benefit future regenerative medicine and clinical drug discovery.
Collapse
Affiliation(s)
- Jun Zeng
- Yunnan Key laboratory for Basic Research on Bone and Joint Diseases & Yunnan Stem Cell Translational Research Center, Kunming University, Kunming 650214, China
| | - Yanjiao Li
- Yunnan Key laboratory for Basic Research on Bone and Joint Diseases & Yunnan Stem Cell Translational Research Center, Kunming University, Kunming 650214, China
| | - Zhaoxia Ma
- Yunnan Key laboratory for Basic Research on Bone and Joint Diseases & Yunnan Stem Cell Translational Research Center, Kunming University, Kunming 650214, China
| | - Min Hu
- Yunnan Key laboratory for Basic Research on Bone and Joint Diseases & Yunnan Stem Cell Translational Research Center, Kunming University, Kunming 650214, China
| |
Collapse
|
6
|
Abbey D, Singh G, Verma I, Derebail S, Kolkundkar U, Chandrashekar DS, Acharya KK, Vemuri MC, Seshagiri PB. Successful Derivation of an Induced Pluripotent Stem Cell Line from a Genetically Nonpermissive Enhanced Green Fluorescent Protein-Transgenic FVB/N Mouse Strain. Cell Reprogram 2019; 21:270-284. [PMID: 31596624 DOI: 10.1089/cell.2019.0019] [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: 11/13/2022] Open
Abstract
The embryonic stem cell line derivation from nonpermissive mouse strains is a challenging and highly inefficient process. The cellular reprogramming strategy provides an alternative route for generating pluripotent stem cell (PSC) lines from such strains. In this study, we successfully derived an enhanced green fluorescent protein (EGFP)-transgenic "N9" induced pluripotent stem cell (iPS cell, iPSC) line from the FVB/N strain-derived mouse embryonic fibroblasts (MEFs). The exposure of MEFs to human OCT4, SOX2, KLF4, and c-MYC (OSKM) transgenes via lentiviral transduction resulted in complete reprogramming. The N9 iPS cell line demonstrated all the criteria of a typical mouse PSC line, including normal colony morphology and karyotype (40,XY), high replication and propagation efficiencies, expression of the pluripotency-associated genes, spontaneous differentiation to three germ lineage-derived cell types, and robust potential of chimeric blastocyst formation. Taken together, using human OSKM genes for transduction, we report, for the first time, the successful derivation of an EGFP-expressing iPS cell line from a genetically nonpermissive transgenic FVB/N mouse. This cell line could provide opportunities for designing protocols for efficient derivation of PSC lines from other nonpermissive strains and developing mouse models of human diseases.
Collapse
Affiliation(s)
- Deepti Abbey
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Gurbind Singh
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India.,Present address: Centre for Stem Cell Research, Christian Medical College Campus, Bagayam, Vellore, India
| | - Isha Verma
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | | | | | | | | | | | - Polani B Seshagiri
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| |
Collapse
|
7
|
Vila OF, Uzel SG, Ma SP, Williams D, Pak J, Kamm RD, Vunjak-Novakovic G. Quantification of human neuromuscular function through optogenetics. Am J Cancer Res 2019; 9:1232-1246. [PMID: 30867827 PMCID: PMC6401498 DOI: 10.7150/thno.25735] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 12/23/2018] [Indexed: 01/15/2023] Open
Abstract
The study of human neuromuscular diseases has traditionally been performed in animal models, due to the difficulty of performing studies in human subjects. Despite the unquestioned value of animal models, inter-species differences hamper the translation of these findings to clinical trials. Tissue-engineered models of the neuromuscular junction (NMJ) allow for the recapitulation of the human physiology in tightly controlled in vitro settings. Methods: Here we report the first human patient-specific tissue-engineered model of the neuromuscular junction (NMJ) that combines stem cell technology with tissue engineering, optogenetics, microfabrication and image processing. The combination of custom-made hardware and software allows for repeated, quantitative measurements of NMJ function in a user-independent manner. Results: We demonstrate the utility of this model for basic and translational research by characterizing in real time the functional changes during physiological and pathological processes. Principal Conclusions: This system holds great potential for the study of neuromuscular diseases and drug screening, allowing for the extraction of quantitative functional data from a human, patient-specific system.
Collapse
|
8
|
Haridhasapavalan KK, Borgohain MP, Dey C, Saha B, Narayan G, Kumar S, Thummer RP. An insight into non-integrative gene delivery approaches to generate transgene-free induced pluripotent stem cells. Gene 2018; 686:146-159. [PMID: 30472380 DOI: 10.1016/j.gene.2018.11.069] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/11/2018] [Accepted: 11/20/2018] [Indexed: 02/06/2023]
Abstract
Over a decade ago, a landmark study that reported derivation of induced Pluripotent Stem Cells (iPSCs) by reprogramming fibroblasts has transformed stem cell research attracting the interest of the scientific community worldwide. These cells circumvent the ethical and immunological concerns associated with embryonic stem cells, and the limited self-renewal ability and restricted differentiation potential linked to adult stem cells. iPSCs hold great potential for understanding basic human biology, in vitro disease modeling, high-throughput drug testing and discovery, and personalized regenerative medicine. The conventional reprogramming methods involving retro- and lenti-viral vectors to deliver reprogramming factors in somatic cells to generate iPSCs nullify the clinical applicability of these cells. Although these gene delivery systems are efficient and robust, they carry an enormous risk of permanent genetic modifications and are potentially tumorigenic. To evade these safety concerns and derive iPSCs for human therapy, tremendous technological advancements have resulted in the development of non-integrating viral- and non-viral approaches. These gene delivery techniques curtail or eliminate the risk of any genomic alteration and enhance the prospects of iPSCs from bench-to-bedside. The present review provides a comprehensive overview of non-integrating viral (adenoviral vectors, adeno-associated viral vectors, and Sendai virus vectors) and DNA-based, non-viral (plasmid transfection, minicircle vectors, transposon vectors, episomal vectors, and liposomal magnetofection) approaches that have the potential to generate transgene-free iPSCs. The understanding of these techniques could pave the way for the use of iPSCs for various biomedical applications.
Collapse
Affiliation(s)
- Krishna Kumar Haridhasapavalan
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute Technology Guwahati, Guwahati 781039, Assam, India.
| | - Manash P Borgohain
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute Technology Guwahati, Guwahati 781039, Assam, India.
| | - Chandrima Dey
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute Technology Guwahati, Guwahati 781039, Assam, India.
| | - Bitan Saha
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute Technology Guwahati, Guwahati 781039, Assam, India
| | - Gloria Narayan
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute Technology Guwahati, Guwahati 781039, Assam, India.
| | - Sachin Kumar
- Viral Immunology Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
9
|
Direct phenotypic conversion of human fibroblasts into functional osteoblasts triggered by a blockade of the transforming growth factor-β signal. Sci Rep 2018; 8:8463. [PMID: 29855543 PMCID: PMC5981640 DOI: 10.1038/s41598-018-26745-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 05/18/2018] [Indexed: 02/06/2023] Open
Abstract
A procedure to generate functional osteoblasts from human somatic cells may pave the way to a novel and effective transplantation therapy in bone disorders. Here, we report that human fibroblasts were induced to show osteoblast phenotypes by culturing with ALK5 i II, which is a specific inhibitor for activin-like kinase 5 (ALK5) (tumor growth factor-β receptor 1 (TGF-β R1)). Cells cultured with ALK5 i II expressed osteoblast-specific genes and massively produced calcified bone matrix, similar to the osteoblasts induced from mesenchymal stem cells (MSC-OBs). Treatment with vitamin D3 in addition to ALK5 i II induced more osteoblast-like characters, and the efficiency of the conversion reached approximately 90%. The chemical compound-mediated directly converted osteoblasts (cOBs) were similar to human primary osteoblasts in terms of expression profiles of osteoblast-related genes. The cOBs abundantly produced bone matrix in vivo and facilitated bone healing after they were transplanted into immunodeficient mice at an artificially induced defect lesion in femoral bone. The present procedure realizes a highly efficient direct conversion of human fibroblasts into transgene-free and highly functional osteoblasts, which might be applied in a novel strategy of bone regeneration therapy in bone diseases.
Collapse
|
10
|
Establishment of integration-free induced pluripotent stem cells from human recessive dystrophic epidermolysis bullosa keratinocytes. J Dermatol Sci 2017; 89:263-271. [PMID: 29229433 DOI: 10.1016/j.jdermsci.2017.11.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/15/2017] [Accepted: 11/27/2017] [Indexed: 11/21/2022]
Abstract
BACKGROUND Induced pluripotent stem cell (iPSC) technology enables patient-specific pluripotent stem cells to be derived from adult somatic cells without the use of an embryonic cell source. To date, recessive dystrophic epidermolysis bullosa (RDEB)-specific iPSCs have been generated from patients using integrating retroviral vectors. However, vector integration into the host genome can endanger the biosafety and differentiation propensities of iPSCs. Although various integration-free reprogramming systems have been reported, their utility in reprogramming somatic cells from patients remains largely undetermined. OBJECTIVE Our study aims to establish safe iPSCs from keratinocytes of RDEB patients using non-integration vector. METHOD We optimized and infected non-integrating Sendai viral vectors to reprogram keratinocytes from healthy volunteers and RDEB patients. RESULTS Sendai vector infection led to the reproducible generation of genomic modification-free iPSCs from these keratinocytes, which was proved by immunohistochemistry, reverse transcription polymerase chain reaction, methylation assay, teratoma assay and embryoid body formation assay. Furthermore, we confirmed that these iPSCs have the potential to differentiate into dermal fibroblasts and epidermal keratinocytes. CONCLUSION This is the first report to prove that the Sendai vector system facilitates the reliable reprogramming of patient keratinocytes into transgene-free iPSCs, providing another pluripotent platform for personalized diagnostic and therapeutic approaches to RDEB.
Collapse
|
11
|
Baranek M, Belter A, Naskręt-Barciszewska MZ, Stobiecki M, Markiewicz WT, Barciszewski J. Effect of small molecules on cell reprogramming. MOLECULAR BIOSYSTEMS 2017; 13:277-313. [PMID: 27918060 DOI: 10.1039/c6mb00595k] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The essential idea of regenerative medicine is to fix or replace tissues or organs with alive and patient-specific implants. Pluripotent stem cells are able to indefinitely self-renew and differentiate into all cell types of the body which makes them a potent substantial player in regenerative medicine. The easily accessible source of induced pluripotent stem cells may allow obtaining and cultivating tissues in vitro. Reprogramming refers to regression of mature cells to its initial pluripotent state. One of the approaches affecting pluripotency is the usage of low molecular mass compounds that can modulate enzymes and receptors leading to the formation of pluripotent stem cells (iPSCs). It would be great to assess the general character of such compounds and reveal their new derivatives or modifications to increase the cell reprogramming efficiency. Many improvements in the methods of pluripotency induction have been made by various groups in order to limit the immunogenicity and tumorigenesis, increase the efficiency and accelerate the kinetics. Understanding the epigenetic changes during the cellular reprogramming process will extend the comprehension of stem cell biology and lead to potential therapeutic approaches. There are compounds which have been already proven to be or for now only putative inducers of the pluripotent state that may substitute for the classic reprogramming factors (Oct3/4, Sox2, Klf4, c-Myc) in order to improve the time and efficiency of pluripotency induction. The effect of small molecules on gene expression is dosage-dependent and their application concentration needs to be strictly determined. In this review we analysed the role of small molecules in modulations leading to pluripotency induction, thereby contributing to our understanding of stem cell biology and uncovering the major mechanisms involved in that process.
Collapse
Affiliation(s)
- M Baranek
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - A Belter
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - M Z Naskręt-Barciszewska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - M Stobiecki
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - W T Markiewicz
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| | - J Barciszewski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego str. 12/14, 61-704 Poznań, Poland.
| |
Collapse
|
12
|
Uusi-Rauva K, Blom T, von Schantz-Fant C, Blom T, Jalanko A, Kyttälä A. Induced Pluripotent Stem Cells Derived from a CLN5 Patient Manifest Phenotypic Characteristics of Neuronal Ceroid Lipofuscinoses. Int J Mol Sci 2017; 18:E955. [PMID: 28468312 PMCID: PMC5454868 DOI: 10.3390/ijms18050955] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/12/2017] [Accepted: 04/26/2017] [Indexed: 01/19/2023] Open
Abstract
Neuronal ceroid lipofuscinoses (NCLs) are autosomal recessive progressive encephalopathies caused by mutations in at least 14 different genes. Despite extensive studies performed in different NCL animal models, the molecular mechanisms underlying neurodegeneration in NCLs remain poorly understood. To model NCL in human cells, we generated induced pluripotent stem cells (iPSCs) by reprogramming skin fibroblasts from a patient with CLN5 (ceroid lipofuscinosis, neuronal, 5) disease, the late infantile variant form of NCL. These CLN5 patient-derived iPSCs (CLN5Y392X iPSCs) harbouring the most common CLN5 mutation, c.1175_1176delAT (p.Tyr392X), were further differentiated into neural lineage cells, the most affected cell type in NCLs. The CLN5Y392X iPSC-derived neural lineage cells showed accumulation of autofluorescent storage material and subunit C of the mitochondrial ATP synthase, both representing the hallmarks of many forms of NCLs, including CLN5 disease. In addition, we detected abnormalities in the intracellular organelles and aberrations in neuronal sphingolipid transportation, verifying the previous findings obtained from Cln5-deficient mouse macrophages. Therefore, patient-derived iPSCs provide a suitable model to study the mechanisms of NCL diseases.
Collapse
Affiliation(s)
- Kristiina Uusi-Rauva
- National Institute for Health and Welfare, Genomics and Biomarkers Unit, P.O. Box 104, 00251 Helsinki, Finland.
- Folkhälsan Institute of Genetics, P.O. Box 63, University of Helsinki, 00014 Helsinki, Finland.
| | - Tea Blom
- National Institute for Health and Welfare, Genomics and Biomarkers Unit, P.O. Box 104, 00251 Helsinki, Finland.
| | | | - Tomas Blom
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland.
| | - Anu Jalanko
- National Institute for Health and Welfare, Genomics and Biomarkers Unit, P.O. Box 104, 00251 Helsinki, Finland.
| | - Aija Kyttälä
- National Institute for Health and Welfare, Genomics and Biomarkers Unit, P.O. Box 104, 00251 Helsinki, Finland.
| |
Collapse
|
13
|
Smith AS, Passey SL, Martin NR, Player DJ, Mudera V, Greensmith L, Lewis MP. Creating Interactions between Tissue-Engineered Skeletal Muscle and the Peripheral Nervous System. Cells Tissues Organs 2016; 202:143-158. [PMID: 27825148 PMCID: PMC5175300 DOI: 10.1159/000443634] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2015] [Indexed: 12/22/2022] Open
Abstract
Effective models of mammalian tissues must allow and encourage physiologically (mimetic) correct interactions between co-cultured cell types in order to produce culture microenvironments as similar as possible to those that would normally occur in vivo. In the case of skeletal muscle, the development of such a culture model, integrating multiple relevant cell types within a biomimetic scaffold, would be of significant benefit for investigations into the development, functional performance, and pathophysiology of skeletal muscle tissue. Although some work has been published regarding the behaviour of in vitro muscle models co-cultured with organotypic slices of CNS tissue or with stem cell-derived neurospheres, little investigation has so far been made regarding the potential to maintain isolated motor neurons within a 3D biomimetic skeletal muscle culture platform. Here, we review the current state of the art for engineering neuromuscular contacts in vitro and provide original data detailing the development of a 3D collagen-based model for the co-culture of primary muscle cells and motor neurons. The devised culture system promotes increased myoblast differentiation, forming arrays of parallel, aligned myotubes on which areas of nerve-muscle contact can be detected by immunostaining for pre- and post-synaptic proteins. Quantitative RT-PCR results indicate that motor neuron presence has a positive effect on myotube maturation, suggesting neural incorporation influences muscle development and maturation in vitro. The importance of this work is discussed in relation to other published neuromuscular co-culture platforms along with possible future directions for the field.
Collapse
Affiliation(s)
- Alec S.T. Smith
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, London, UK
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM) England, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
- Department of Bioengineering, University of Washington, Seattle, Wash., USA
| | - Samantha L. Passey
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM) England, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Vic., Australia
| | - Neil R.W. Martin
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM) England, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Darren J. Player
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM) England, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Vivek Mudera
- Division of Surgery and Interventional Science, UCL Institute of Orthopaedics and Musculoskeletal Science, London, UK
| | - Linda Greensmith
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, London, UK
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, London, UK
| | - Mark P. Lewis
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM) England, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
- *Prof. Mark P. Lewis, School of Sport, Exercise and Health Sciences, Loughborough University, Ashby Road, Loughborough LE11 3TU (UK), E-Mail
| |
Collapse
|
14
|
Chemically Induced Reprogramming of Somatic Cells to Pluripotent Stem Cells and Neural Cells. Int J Mol Sci 2016; 17:226. [PMID: 26861316 PMCID: PMC4783958 DOI: 10.3390/ijms17020226] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 02/01/2016] [Indexed: 12/31/2022] Open
Abstract
The ability to generate transplantable neural cells in a large quantity in the laboratory is a critical step in the field of developing stem cell regenerative medicine for neural repair. During the last few years, groundbreaking studies have shown that cell fate of adult somatic cells can be reprogrammed through lineage specific expression of transcription factors (TFs)-and defined culture conditions. This key concept has been used to identify a number of potent small molecules that could enhance the efficiency of reprogramming with TFs. Recently, a growing number of studies have shown that small molecules targeting specific epigenetic and signaling pathways can replace all of the reprogramming TFs. Here, we provide a detailed review of the studies reporting the generation of chemically induced pluripotent stem cells (ciPSCs), neural stem cells (ciNSCs), and neurons (ciN). We also discuss the main mechanisms of actions and the pathways that the small molecules regulate during chemical reprogramming.
Collapse
|
15
|
Histone H4 acetylation and the epigenetic reader Brd4 are critical regulators of pluripotency in embryonic stem cells. BMC Genomics 2016; 17:95. [PMID: 26847871 PMCID: PMC4740988 DOI: 10.1186/s12864-016-2414-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/26/2016] [Indexed: 12/14/2022] Open
Abstract
Background Pluripotent cells can be differentiated into many different cell types in vitro. Successful differentiation is guided in large part by epigenetic reprogramming and regulation of critical gene expression patterns. Recent genome-wide studies have identified the distribution of different histone-post-translational modifications (PTMs) in various conditions and during cellular differentiation. However, our understanding of the abundance of histone PTMs and their regulatory mechanisms still remain unknown. Results Here, we present a quantitative and comprehensive study of the abundance levels of histone PTMs during the differentiation of mouse embryonic stem cells (ESCs) using mass spectrometry (MS). We observed dynamic changes of histone PTMs including increased H3K9 methylation levels in agreement with previously reported results. More importantly, we found a global decrease of multiply acetylated histone H4 peptides. Brd4 targets acetylated H4 with a strong affinity to multiply modified H4 acetylation sites. We observed that the protein levels of Brd4 decreased upon differentiation together with global histone H4 acetylation. Inhibition of Brd4:histone H4 interaction by the BET domain inhibitor (+)-JQ1 in ESCs results in enhanced differentiation to the endodermal lineage, by disrupting the protein abundance dynamics. Genome-wide ChIP-seq mapping showed that Brd4 and H4 acetylation are co-occupied in the genome, upstream of core pluripotency genes such as Oct4 and Nanog in ESCs and lineage-specific genes in embryoid bodies (EBs). Conclusions Together, our data demonstrate the fundamental role of Brd4 in monitoring cell differentiation through its interaction with acetylated histone marks and disruption of Brd4 may cause aberrant differentiation. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2414-y) contains supplementary material, which is available to authorized users.
Collapse
|
16
|
Kyttälä A, Moraghebi R, Valensisi C, Kettunen J, Andrus C, Pasumarthy KK, Nakanishi M, Nishimura K, Ohtaka M, Weltner J, Van Handel B, Parkkonen O, Sinisalo J, Jalanko A, Hawkins RD, Woods NB, Otonkoski T, Trokovic R. Genetic Variability Overrides the Impact of Parental Cell Type and Determines iPSC Differentiation Potential. Stem Cell Reports 2016; 6:200-12. [PMID: 26777058 PMCID: PMC4750096 DOI: 10.1016/j.stemcr.2015.12.009] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/08/2015] [Accepted: 12/14/2015] [Indexed: 12/18/2022] Open
Abstract
Reports on the retention of somatic cell memory in induced pluripotent stem cells (iPSCs) have complicated the selection of the optimal cell type for the generation of iPSC biobanks. To address this issue we compared transcriptomic, epigenetic, and differentiation propensities of genetically matched human iPSCs derived from fibroblasts and blood, two tissues of the most practical relevance for biobanking. Our results show that iPSC lines derived from the same donor are highly similar to each other. However, genetic variation imparts a donor-specific expression and methylation profile in reprogrammed cells that leads to variable functional capacities of iPSC lines. Our results suggest that integration-free, bona fide iPSC lines from fibroblasts and blood can be combined in repositories to form biobanks. Due to the impact of genetic variation on iPSC differentiation, biobanks should contain cells from large numbers of donors. Isogenic iPSC from fibroblasts and blood have similar differentiation propensities Donor-dependent variability affects molecular and differentiation propensities of iPSCs Impact of donor variability exceeds source-cell-specific differences in iPSC lines Bona fide iPSC lines from different tissues can be combined in the repositories
Collapse
Affiliation(s)
- Aija Kyttälä
- Genomics and Biomarkers Unit, National Institute for Health and Welfare (THL), THL Biobank, 00290 Helsinki, Finland
| | - Roksana Moraghebi
- Department of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, BMC A12, 221 84 Lund, Sweden
| | - Cristina Valensisi
- Division of Medical Genetics, Departments of Medicine and Genome Sciences, University of Washington, Seattle, WA 98195-7720, USA; Turku Centre for Biotechnology, Turku 20520, Finland
| | - Johannes Kettunen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare (THL), THL Biobank, 00290 Helsinki, Finland; Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu 90014, Finland; NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio 70210, Finland; Biocenter Oulu, University of Oulu, 90014 Oulu, Finland
| | - Colin Andrus
- Division of Medical Genetics, Departments of Medicine and Genome Sciences, University of Washington, Seattle, WA 98195-7720, USA
| | | | - Mahito Nakanishi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8565, Japan
| | - Ken Nishimura
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8565, Japan; Laboratory of Gene Regulation, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Manami Ohtaka
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8565, Japan
| | - Jere Weltner
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, University of Helsinki, 00290 Helsinki, Finland
| | | | - Olavi Parkkonen
- Heart and Lung Center, Helsinki University Central Hospital and University of Helsinki, 00029 HUS Helsinki, Finland
| | - Juha Sinisalo
- Heart and Lung Center, Helsinki University Central Hospital and University of Helsinki, 00029 HUS Helsinki, Finland
| | - Anu Jalanko
- Genomics and Biomarkers Unit, National Institute for Health and Welfare (THL), THL Biobank, 00290 Helsinki, Finland
| | - R David Hawkins
- Division of Medical Genetics, Departments of Medicine and Genome Sciences, University of Washington, Seattle, WA 98195-7720, USA; Turku Centre for Biotechnology, Turku 20520, Finland
| | - Niels-Bjarne Woods
- Department of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, BMC A12, 221 84 Lund, Sweden
| | - Timo Otonkoski
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, University of Helsinki, 00290 Helsinki, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, 00029 HUS Helsinki, Finland.
| | - Ras Trokovic
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, University of Helsinki, 00290 Helsinki, Finland.
| |
Collapse
|
17
|
Pini J, Rouleau M, Desnuelle C, Sacconi S, Bendahhou S. Modeling Andersen's Syndrome in Human Induced Pluripotent Stem Cells. Stem Cells Dev 2015; 25:151-9. [PMID: 26573604 DOI: 10.1089/scd.2015.0258] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Andersen's syndrome (AS) is a rare disorder characterized by a triad of symptoms: periodic paralysis, cardiac arrhythmia, and bone developmental defects. Most of the patients carry mutations on the inward rectifier potassium channel Kir2.1 encoded by the KCNJ2 gene. kcnj2 knockout mice are lethal at birth preventing, hence, thorough investigations of the physiological and pathophysiological events. We have generated induced pluripotent stem (iPS) cells from healthy as well as from AS patient muscular biopsies using the four-gene cassette required for cellular reprogramming (Oct4, Sox2, Klf4, and c-Myc). The generated AS-iPS cells exhibited the gold standard requirement for iPS cells: expression of genetics and surface pluripotent markers, strong alkaline phosphatase activity, self-renewal, and could be differentiated by the formation of embryoid bodies (EBs) into the three germ layers. Sequencing of the entire coding sequence of the KCNJ2 gene, in AS-iPS cells, revealed that the reprogramming process did not revert the Andersen's syndrome-associated mutation. Moreover, no difference was observed between control and AS-iPS cells in terms of pluripotent markers' expression, self-renewal, and three germ layer differentiation. Interestingly, expression of osteogenic markers are lower in EB-differentiated AS-iPS compared to control iPS cells. Our results showed that the Kir2.1 channel is not important for the reprogramming process and the early step of the development in vitro. However, the osteogenic machinery appears to be hastened in AS-iPS cells, strongly indicating that the generated AS-iPS cells could be a good model to better understand the AS pathophysiology.
Collapse
Affiliation(s)
- Jonathan Pini
- 1 UMR7370 CNRS, LP2M, Labex ICST, Faculté de Médecine, University Nice Sophia Antipolis , Nice, France
| | - Matthieu Rouleau
- 1 UMR7370 CNRS, LP2M, Labex ICST, Faculté de Médecine, University Nice Sophia Antipolis , Nice, France
| | - Claude Desnuelle
- 2 INSERM, U1081, Institute for Research on Cancer and Aging of Nice (IRCAN) , Nice, France .,3 CNRS, UMR 7284, Institute for Research on Cancer and Aging of Nice (IRCAN) , Nice, France .,4 Faculty of Medicine, Neuromuscular Diseases and ALS Specialized Center, University of Nice-Sophia-Antipolis , Nice, France
| | - Sabrina Sacconi
- 2 INSERM, U1081, Institute for Research on Cancer and Aging of Nice (IRCAN) , Nice, France .,3 CNRS, UMR 7284, Institute for Research on Cancer and Aging of Nice (IRCAN) , Nice, France .,4 Faculty of Medicine, Neuromuscular Diseases and ALS Specialized Center, University of Nice-Sophia-Antipolis , Nice, France
| | - Saïd Bendahhou
- 1 UMR7370 CNRS, LP2M, Labex ICST, Faculté de Médecine, University Nice Sophia Antipolis , Nice, France
| |
Collapse
|
18
|
Trokovic R, Weltner J, Noisa P, Raivio T, Otonkoski T. Combined negative effect of donor age and time in culture on the reprogramming efficiency into induced pluripotent stem cells. Stem Cell Res 2015; 15:254-62. [DOI: 10.1016/j.scr.2015.06.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/01/2015] [Accepted: 06/05/2015] [Indexed: 01/17/2023] Open
|
19
|
Rony IK, Baten A, Bloomfield JA, Islam ME, Billah MM, Islam KD. Inducing pluripotency in vitro: recent advances and highlights in induced pluripotent stem cells generation and pluripotency reprogramming. Cell Prolif 2015; 48:140-56. [PMID: 25643745 DOI: 10.1111/cpr.12162] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 10/05/2014] [Indexed: 12/12/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are considered patient-specific counterparts of embryonic stem cells as they originate from somatic cells after forced expression of pluripotency reprogramming factors Oct4, Sox2, Klf4 and c-Myc. iPSCs offer unprecedented opportunity for personalized cell therapies in regenerative medicine. In recent years, iPSC technology has undergone substantial improvement to overcome slow and inefficient reprogramming protocols, and to ensure clinical-grade iPSCs and their functional derivatives. Recent developments in iPSC technology include better reprogramming methods employing novel delivery systems such as non-integrating viral and non-viral vectors, and characterization of alternative reprogramming factors. Concurrently, small chemical molecules (inhibitors of specific signalling or epigenetic regulators) have become crucial to iPSC reprogramming; they have the ability to replace putative reprogramming factors and boost reprogramming processes. Moreover, common dietary supplements, such as vitamin C and antioxidants, when introduced into reprogramming media, have been found to improve genomic and epigenomic profiles of iPSCs. In this article, we review the most recent advances in the iPSC field and potent application of iPSCs, in terms of cell therapy and tissue engineering.
Collapse
Affiliation(s)
- I K Rony
- Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| | | | | | | | | | | |
Collapse
|
20
|
Davies SG, Kennewell PD, Russell AJ, Seden PT, Westwood R, Wynne GM. Stemistry: the control of stem cells in situ using chemistry. J Med Chem 2015; 58:2863-94. [PMID: 25590360 DOI: 10.1021/jm500838d] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A new paradigm for drug research has emerged, namely the deliberate search for molecules able to selectively affect the proliferation, differentiation, and migration of adult stem cells within the tissues in which they exist. Recently, there has been significant interest in medicinal chemistry toward the discovery and design of low molecular weight molecules that affect stem cells and thus have novel therapeutic activity. We believe that a successful agent from such a discover program would have profound effects on the treatment of many long-term degenerative disorders. Among these conditions are examples such as cardiovascular decay, neurological disorders including Alzheimer's disease, and macular degeneration, all of which have significant unmet medical needs. This perspective will review evidence from the literature that indicates that discovery of such agents is achievable and represents a worthwhile pursuit for the skills of the medicinal chemist.
Collapse
Affiliation(s)
- Stephen G Davies
- †Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, U.K
| | - Peter D Kennewell
- †Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, U.K
| | - Angela J Russell
- †Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, U.K.,‡Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, U.K
| | - Peter T Seden
- †Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, U.K
| | - Robert Westwood
- †Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, U.K
| | - Graham M Wynne
- †Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, U.K
| |
Collapse
|
21
|
Li M, Li L, Zhang J, Verma V, Liu Q, Shi D, Huang B. An Insight on Small Molecule Induced Foot-Print Free Naive Pluripotent Stem Cells in Livestock. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/scd.2015.51001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Park HS, Hwang I, Choi KA, Jeong H, Lee JY, Hong S. Generation of induced pluripotent stem cells without genetic defects by small molecules. Biomaterials 2014; 39:47-58. [PMID: 25477171 DOI: 10.1016/j.biomaterials.2014.10.055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 10/19/2014] [Indexed: 12/21/2022]
Abstract
The generation of induced pluripotent stem cells (iPSCs) often causes genetic and epigenetic defects, which may limit their clinical applications. Here, we show that reprogramming in the presence of small molecules preserved the genomic stability of iPSCs by inhibiting DNA double-strand breaks (DSBs) and activating Zscan4 gene. Surprisingly, the small molecules protected normal karyotype by facilitating repair of the DSBs that occurred during the early reprogramming process and long-term culture of iPSCs. The stemness and cell growth of iPSCs(+) were normally sustained with high expression of pluripotency genes compared that of iPSCs(-). Moreover, small molecules maintained the differentiation potential of iPSCs(+) for the three germ layers, whereas it was lost in iPSCs(-). Our results demonstrate that the defined small molecules are potent factors for generation of high quality iPSCs with preservation of genomic integrity by facilitating the reprogramming process.
Collapse
Affiliation(s)
- Hang-Soo Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Jeongneung-dong, Sungbuk-gu, Seoul 136-703, Republic of Korea
| | - Insik Hwang
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Jeongneung-dong, Sungbuk-gu, Seoul 136-703, Republic of Korea
| | - Kyung-Ah Choi
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Jeongneung-dong, Sungbuk-gu, Seoul 136-703, Republic of Korea
| | - Hyesun Jeong
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Jeongneung-dong, Sungbuk-gu, Seoul 136-703, Republic of Korea
| | - Ji-Yun Lee
- Department of Pathology, College of Medicine, Korea University, Anam-dong, Sungbuk-gu, Seoul 136-701, Republic of Korea
| | - Sunghoi Hong
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Jeongneung-dong, Sungbuk-gu, Seoul 136-703, Republic of Korea.
| |
Collapse
|
23
|
Trokovic R, Weltner J, Nishimura K, Ohtaka M, Nakanishi M, Salomaa V, Jalanko A, Otonkoski T, Kyttälä A. Advanced feeder-free generation of induced pluripotent stem cells directly from blood cells. Stem Cells Transl Med 2014; 3:1402-9. [PMID: 25355732 DOI: 10.5966/sctm.2014-0113] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Generation of validated human induced pluripotent stem cells (iPSCs) for biobanking is essential for exploring the full potential of iPSCs in disease modeling and drug discovery. Peripheral blood mononuclear cells (PBMCs) are attractive targets for reprogramming, because blood is collected by a routine clinical procedure and is a commonly stored material in biobanks. Generation of iPSCs from blood cells has previously been reported using integrative retroviruses, episomal Sendai viruses, and DNA plasmids. However, most of the published protocols require expansion and/or activation of a specific cell population from PBMCs. We have recently collected a PBMC cohort from the Finnish population containing more than 2,000 subjects. Here we report efficient generation of iPSCs directly from PBMCs in feeder-free conditions in approximately 2 weeks. The produced iPSC clones are pluripotent and transgene-free. Together, these properties make this novel method a powerful tool for large-scale reprogramming of PBMCs and for iPSC biobanking.
Collapse
Affiliation(s)
- Ras Trokovic
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, University of Helsinki, Helsinki, Finland; Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Chronic Disease Prevention and Public Health Genomics Unit, THL Biobank, National Institute for Health and Welfare (THL), Helsinki, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Jere Weltner
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, University of Helsinki, Helsinki, Finland; Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Chronic Disease Prevention and Public Health Genomics Unit, THL Biobank, National Institute for Health and Welfare (THL), Helsinki, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Ken Nishimura
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, University of Helsinki, Helsinki, Finland; Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Chronic Disease Prevention and Public Health Genomics Unit, THL Biobank, National Institute for Health and Welfare (THL), Helsinki, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Manami Ohtaka
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, University of Helsinki, Helsinki, Finland; Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Chronic Disease Prevention and Public Health Genomics Unit, THL Biobank, National Institute for Health and Welfare (THL), Helsinki, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Mahito Nakanishi
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, University of Helsinki, Helsinki, Finland; Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Chronic Disease Prevention and Public Health Genomics Unit, THL Biobank, National Institute for Health and Welfare (THL), Helsinki, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Veikko Salomaa
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, University of Helsinki, Helsinki, Finland; Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Chronic Disease Prevention and Public Health Genomics Unit, THL Biobank, National Institute for Health and Welfare (THL), Helsinki, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Anu Jalanko
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, University of Helsinki, Helsinki, Finland; Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Chronic Disease Prevention and Public Health Genomics Unit, THL Biobank, National Institute for Health and Welfare (THL), Helsinki, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Timo Otonkoski
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, University of Helsinki, Helsinki, Finland; Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Chronic Disease Prevention and Public Health Genomics Unit, THL Biobank, National Institute for Health and Welfare (THL), Helsinki, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Aija Kyttälä
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, University of Helsinki, Helsinki, Finland; Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Chronic Disease Prevention and Public Health Genomics Unit, THL Biobank, National Institute for Health and Welfare (THL), Helsinki, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
24
|
Feltes BC, Bonatto D. Combining small molecules for cell reprogramming through an interatomic analysis. MOLECULAR BIOSYSTEMS 2014; 9:2741-63. [PMID: 24056910 DOI: 10.1039/c3mb70159j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The knowledge available about the application and generation of induced pluripotent stem cells (iPSC) has grown since their discovery, and new techniques to enhance the reprogramming process have been described. Among the new approaches to induce iPSC that have gained great attention is the use of small molecules for reprogramming. The application of small molecules, unlike genetic manipulation, provides for control of the reprogramming process through the shifting of concentrations and the combination of different molecules. However, different researchers have reported the use of "reprogramming cocktails" with variable results and drug combinations. Thus, the proper combination of small molecules for successful and enhanced reprogramming is a matter for discussion. However, testing all potential drug combinations in different cell lineages is very costly and time-consuming. Therefore, in this article, we discuss the use of already employed molecules for iPSC generation, followed by the application of systems chemo-biology tools to create different data sets of protein-protein (PPI) and chemical-protein (CPI) interaction networks based on the knowledge of already used and new reprogramming cocktail combinations. We further analyzed the biological processes associated with PPI-CPI networks and provided new potential protein targets to be inhibited or expressed for stem cell reprogramming. In addition, we applied a new interference analysis to prospective targets that could negatively affect the classical pluripotency-associated factors (SOX2, NANOG, KLF4 and OCT4) and thus potentially improve reprogramming protocols.
Collapse
Affiliation(s)
- Bruno César Feltes
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves 9500 - Prédio 43421 - Sala 219, Porto Alegre, Caixa Postal 15005, RS - Brazil.
| | | |
Collapse
|
25
|
Ifkovits JL, Addis RC, Epstein JA, Gearhart JD. Inhibition of TGFβ signaling increases direct conversion of fibroblasts to induced cardiomyocytes. PLoS One 2014; 9:e89678. [PMID: 24586958 PMCID: PMC3935923 DOI: 10.1371/journal.pone.0089678] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 01/21/2014] [Indexed: 12/20/2022] Open
Abstract
Recent studies have been successful at utilizing ectopic expression of transcription factors to generate induced cardiomyocytes (iCMs) from fibroblasts, albeit at a low frequency in vitro. This work investigates the influence of small molecules that have been previously reported to improve differentiation to cardiomyocytes as well as reprogramming to iPSCs in conjunction with ectopic expression of the transcription factors Hand2, Nkx2.5, Gata4, Mef2C, and Tbx5 on the conversion to functional iCMs. We utilized a reporter system in which the calcium indicator GCaMP is driven by the cardiac Troponin T promoter to quantify iCM yield. The TGFβ inhibitor, SB431542 (SB), was identified as a small molecule capable of increasing the conversion of both mouse embryonic fibroblasts and adult cardiac fibroblasts to iCMs up to ∼5 fold. Further characterization revealed that inhibition of TGFβ by SB early in the reprogramming process led to the greatest increase in conversion of fibroblasts to iCMs in a dose-responsive manner. Global transcriptional analysis at Day 3 post-induction of the transcription factors revealed an increased expression of genes associated with the development of cardiac muscle in the presence of SB compared to the vehicle control. Incorporation of SB in the reprogramming process increases the efficiency of iCM generation, one of the major goals necessary to enable the use of iCMs for discovery-based applications and for the clinic.
Collapse
Affiliation(s)
- Jamie L. Ifkovits
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (JLI); (JDG)
| | - Russell C. Addis
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jonathan A. Epstein
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - John D. Gearhart
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (JLI); (JDG)
| |
Collapse
|
26
|
Lu B, Atala A. Small molecules and small molecule drugs in regenerative medicine. Drug Discov Today 2013; 19:801-8. [PMID: 24252867 DOI: 10.1016/j.drudis.2013.11.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 10/11/2013] [Accepted: 11/11/2013] [Indexed: 02/07/2023]
Abstract
Regenerative medicine is an emerging, multidisciplinary science that aims to replace or regenerate human cells, tissues or organs, to restore or establish normal function. Research on small molecules and small molecule drugs in regenerative medicine is currently increasing. In this review, we discuss the potential applications of small molecules and small molecule drugs in regenerative medicine. These include enabling novel cell therapy approaches and augmentation of endogenous cells for tissue regeneration, facilitating the generation of target cells for cell therapy, improving the interactions between cells and biomatrices for tissue engineering, and enhancing endogenous stem cell function for tissue regeneration. We also discuss the potential challenges for small molecule drugs in regenerative medicine.
Collapse
Affiliation(s)
- Baisong Lu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA.
| |
Collapse
|
27
|
Teng S, Liu C, Krettek C, Jagodzinski M. The application of induced pluripotent stem cells for bone regeneration: current progress and prospects. TISSUE ENGINEERING PART B-REVIEWS 2013; 20:328-39. [PMID: 24102431 DOI: 10.1089/ten.teb.2013.0301] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Loss of healthy bone tissue and dysosteogenesis are still common and significant problems in clinics. Cell-based therapy using mesenchymal stem cells (MSCs) has been performed in patients for quite some time, but the inherent drawbacks of these cells, such as the reductions in proliferation rate and osteogenic differentiation potential that occur with aging, greatly limit their further application. Moreover, embryonic stem cells (ESCs) have brought new hope to osteoregenerative medicine because of their full pluripotent differentiation potential and excellent performance in bone regeneration. However, the ethical issues involved in destroying human embryos and the immune reactions that occur after transplantation are two major stumbling blocks impeding the clinical application of ESCs. Instead, induced pluripotent stem cells (iPSCs), which are ESC-like pluripotent cells that are reprogrammed from adult somatic cells using defined transcription factors, are considered a more promising source of cells for regenerative medicine because they present no ethical or immunological issues. Here, we summarize the primary technologies for generating iPSCs and the biological properties of these cells, review the current advances in iPSC-based bone regeneration and, finally, discuss the remaining challenges associated with these cells, particularly safety issues and their potential application for osteoregenerative medicine.
Collapse
Affiliation(s)
- Songsong Teng
- 1 Department of Orthopedic Trauma, Hanover Medical School (MHH) , Hanover, Germany
| | | | | | | |
Collapse
|
28
|
Tissue- and cell-type-specific manifestations of heteroplasmic mtDNA 3243A>G mutation in human induced pluripotent stem cell-derived disease model. Proc Natl Acad Sci U S A 2013; 110:E3622-30. [PMID: 24003133 DOI: 10.1073/pnas.1311660110] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Mitochondrial DNA (mtDNA) mutations manifest with vast clinical heterogeneity. The molecular basis of this variability is mostly unknown because the lack of model systems has hampered mechanistic studies. We generated induced pluripotent stem cells from patients carrying the most common human disease mutation in mtDNA, m.3243A>G, underlying mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome. During reprogramming, heteroplasmic mtDNA showed bimodal segregation toward homoplasmy, with concomitant changes in mtDNA organization, mimicking mtDNA bottleneck during epiblast specification. Induced pluripotent stem cell-derived neurons and various tissues derived from teratomas manifested cell-type specific respiratory chain (RC) deficiency patterns. Similar to MELAS patient tissues, complex I defect predominated. Upon neuronal differentiation, complex I specifically was sequestered in perinuclear PTEN-induced putative kinase 1 (PINK1) and Parkin-positive autophagosomes, suggesting active degradation through mitophagy. Other RC enzymes showed normal mitochondrial network distribution. Our data show that cellular context actively modifies RC deficiency manifestation in MELAS and that autophagy is a significant component of neuronal MELAS pathogenesis.
Collapse
|
29
|
Muraro MJ, Kempe H, Verschure PJ. Concise Review: The Dynamics of Induced Pluripotency and Its Behavior Captured in Gene Network Motifs. Stem Cells 2013; 31:838-48. [DOI: 10.1002/stem.1340] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 12/20/2012] [Indexed: 01/01/2023]
|
30
|
Pirozhkova I, Barat A, Dmitriev P, Kim E, Robert T, Guégan J, Bilhou-Nabera C, Busato F, Tost J, Carnac G, Laoudj-Chenivesse D, Lipinski M, Vassetzky Y. Differences in transcription patterns between induced pluripotent stem cells produced from the same germ layer are erased upon differentiation. PLoS One 2013; 8:e53033. [PMID: 23326377 PMCID: PMC3541362 DOI: 10.1371/journal.pone.0053033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 11/22/2012] [Indexed: 11/19/2022] Open
Abstract
Little is known about differences between induced pluripotent stem cells produced from tissues originating from the same germ layer. We have generated human myoblast-derived iPS cells by retroviral transduction of human primary myoblasts with the OCT3/4, SOX2, KLF4 and MYC coding sequences and compared them to iPS produced from human primary fibroblasts. When cultivated in vitro, these iPS cells proved similar to human embryonic stem cells in terms of morphology, expression of embryonic stemness markers and gene promoter methylation patterns. Embryonic bodies were derived that expressed endodermal, mesodermal as well as ectodermal markers. A comparative analysis of transcription patterns revealed significant differences in the gene expression pattern between myoblast- and fibroblast-derived iPS cells. However, these differences were reduced in the mesenchymal stem cells derived from the two iPS cell types were compared.
Collapse
Affiliation(s)
- Iryna Pirozhkova
- CNRS, Université Paris-Sud, UMR-8126, Institut de cancérologie Gustave Roussy, Villejuif, France
| | - Ana Barat
- CNRS, Université Paris-Sud, UMR-8126, Institut de cancérologie Gustave Roussy, Villejuif, France
| | - Petr Dmitriev
- CNRS, Université Paris-Sud, UMR-8126, Institut de cancérologie Gustave Roussy, Villejuif, France
- INSERM U1046, Université Montpellier 1, Université Montpellier 2, Montpellier, France
| | - Elena Kim
- CNRS, Université Paris-Sud, UMR-8126, Institut de cancérologie Gustave Roussy, Villejuif, France
| | - Thomas Robert
- Integrated Biology Platform, Institut de cancérologie Gustave Roussy, Villejuif, France
| | - Justine Guégan
- Integrated Biology Platform, Institut de cancérologie Gustave Roussy, Villejuif, France
| | - Chrystèle Bilhou-Nabera
- CNRS, Université Paris-Sud, UMR-8126, Institut de cancérologie Gustave Roussy, Villejuif, France
| | - Florence Busato
- Laboratory of Epigenetics, Centre national de génotypage CEA-Institut de génomique, Evry, France
| | - Jörg Tost
- Laboratory of Epigenetics, Centre national de génotypage CEA-Institut de génomique, Evry, France
- Laboratory for Functional Genomics, Fondation Jean Dausset – CEPH, Paris, France
| | - Gilles Carnac
- INSERM U1046, Université Montpellier 1, Université Montpellier 2, Montpellier, France
| | | | - Marc Lipinski
- CNRS, Université Paris-Sud, UMR-8126, Institut de cancérologie Gustave Roussy, Villejuif, France
| | - Yegor Vassetzky
- CNRS, Université Paris-Sud, UMR-8126, Institut de cancérologie Gustave Roussy, Villejuif, France
| |
Collapse
|
31
|
Asakura A. Skeletal Muscle-derived Hematopoietic Stem Cells: Muscular Dystrophy Therapy by Bone Marrow Transplantation. ACTA ACUST UNITED AC 2012; Suppl 11. [PMID: 24524008 PMCID: PMC3918728 DOI: 10.4172/2157-7633.s11-005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
For postnatal growth and regeneration of skeletal muscle, satellite cells, a self-renewing pool of muscle stem cells, give rise to daughter myogenic precursor cells that contribute to the formation of new muscle fibers. In addition to this key myogenic cell class, adult skeletal muscle also contains hematopoietic stem cell and progenitor cell populations which can be purified as a side population (SP) fraction or as a hematopoietic marker CD45-positive cell population. These muscle-derived hematopoietic stem/progenitor cell populations are surprisingly capable of differentiation into hematopoietic cells both after transplantation into irradiated mice and during in vitro colony formation assay. Therefore, these muscle-derived hematopoietic stem/progenitor cells appear to have characteristics similar to classical hematopoietic stem/progenitor cells found in bone marrow. This review outlines recent findings regarding hematopoietic stem/progenitor cell populations residing in adult skeletal muscle and discusses their myogenic potential along with their role in the stem cell niche and related cell therapies for approaching treatment of Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Atsushi Asakura
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|