1
|
Della Rocca Y, Diomede F, Konstantinidou F, Gatta V, Stuppia L, Benedetto U, Zimarino M, Lanuti P, Trubiani O, Pizzicannella J. Autologous hGMSC-Derived iPS: A New Proposal for Tissue Regeneration. Int J Mol Sci 2024; 25:9169. [PMID: 39273117 PMCID: PMC11395260 DOI: 10.3390/ijms25179169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
The high mortality in the global population due to chronic diseases highlights the urgency to identify effective alternative therapies. Regenerative medicine provides promising new approaches for this purpose, particularly in the use of induced pluripotent stem cells (iPSCs). The aim of the work is to establish a new pluripotency cell line obtained for the first time by reprogramming human gingival mesenchymal stem cells (hGMSCs) by a non-integrating method. The hGMSC-derived iPS line characterization is performed through morphological analysis with optical and electron scanning microscopy and through the pluripotency markers expression evaluation in cytofluorimetry, immunofluorescence, and RT-PCR. To confirm the pluripotency of new hGMSC-derived iPS, the formation of embryoid bodies (EBs), as an alternative to the teratoma formation test, is studied in morphological analysis and through three germ layers' markers' expression in immunofluorescence and RT-PCR. At the end, a comparative study between parental hGMSCs and derived iPS cells is performed also for the extracellular vesicles (EVs) and their miRNA content. The new hGMSC-derived iPS line demonstrated to be pluripotent in all aspects, thus representing an innovative dynamic platform for personalized tissue regeneration.
Collapse
Affiliation(s)
- Ylenia Della Rocca
- Department of Innovative Technologies in Medicine & Dentistry, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
| | - Francesca Diomede
- Department of Innovative Technologies in Medicine & Dentistry, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
| | - Fanì Konstantinidou
- Department of Psychological Health and Territorial Sciences, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via Luigi Polacchi 11, 66100 Chieti, Italy
| | - Valentina Gatta
- Department of Psychological Health and Territorial Sciences, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via Luigi Polacchi 11, 66100 Chieti, Italy
| | - Liborio Stuppia
- Department of Psychological Health and Territorial Sciences, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via Luigi Polacchi 11, 66100 Chieti, Italy
| | - Umberto Benedetto
- Department of Cardiac Surgery, "S.S. Annunziata" Hospital, ASL 2 Abruzzo, Via dei Vestini, 66100 Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, Via Luigi Polacchi 11, 66100 Chieti, Italy
| | - Marco Zimarino
- Department of Cardiology, "S.S. Annunziata" Hospital, ASL 2 Abruzzo, Via dei Vestini, 66100 Chieti, Italy
| | - Paola Lanuti
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via Luigi Polacchi 11, 66100 Chieti, Italy
- Department of Medicine and Aging Science, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy
| | - Oriana Trubiani
- Department of Innovative Technologies in Medicine & Dentistry, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
| | - Jacopo Pizzicannella
- Department of Engineering and Geology, "G. d'Annunzio" University of Chieti-Pescara, Viale Pindaro, 42, 65127 Pescara, Italy
| |
Collapse
|
2
|
Cell-Free Therapies: The Use of Cell Extracts to Mitigate Irradiation-Injured Salivary Glands. BIOLOGY 2023; 12:biology12020305. [PMID: 36829582 PMCID: PMC9953449 DOI: 10.3390/biology12020305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023]
Abstract
Radiotherapy is a standard treatment for head and neck cancer patients worldwide. However, millions of patients who received radiotherapy consequently suffer from xerostomia because of irreversible damage to salivary glands (SGs) caused by irradiation (IR). Current treatments for IR-induced SG hypofunction only provide temporary symptom alleviation but do not repair the damaged SG, thus resulting in limited treatment efficacy. Therefore, there has recently been a growing interest in regenerative treatments, such as cell-free therapies. This review aims to summarize cell-free therapies for IR-induced SG, with a particular emphasis on utilizing diverse cell extract (CE) administrations. Cell extract is a group of heterogeneous mixtures containing multifunctional inter-cellular molecules. This review discusses the current knowledge of CE's components and efficacy. We propose optimal approaches to improve cell extract treatment from multiple perspectives (e.g., delivery routes, preparation methods, and other details regarding CE administration). In addition, the advantages and limitations of CE treatment are systematically discussed by comparing it to other cell-free (such as conditioned media and exosomes) and cell-based therapies. Although a comprehensive identification of the bioactive factors within CEs and their mechanisms of action have yet to be fully understood, we propose cell extract therapy as an effective, practical, user-friendly, and safe option to conventional therapies in IR-induced SG.
Collapse
|
3
|
Navarro M, Halstead MM, Rincon G, Mutto AA, Ross PJ. bESC from cloned embryos do not retain transcriptomic or epigenetic memory from somatic donor cells. Reproduction 2022; 164:243-257. [PMID: 35951478 DOI: 10.1530/rep-22-0063] [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] [Received: 02/16/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022]
Abstract
In brief Epigenetic reprogramming after mammalian somatic cell nuclear transfer is often incomplete, resulting in low efficiency of cloning. However, gene expression and histone modification analysis indicated high similarities in transcriptome and epigenomes of bovine embryonic stem cells from in vitro fertilized and somatic cell nuclear transfer embryos. Abstract Embryonic stem cells (ESC) indefinitely maintain the pluripotent state of the blastocyst epiblast. Stem cells are invaluable for studying development and lineage commitment, and in livestock, they constitute a useful tool for genomic improvement and in vitro breeding programs. Although these cells have been recently derived from bovine blastocysts, a detailed characterization of their molecular state is lacking. Here, we apply cutting-edge technologies to analyze the transcriptomic and epigenomic landscape of bovine ESC (bESC) obtained from in vitro fertilized (IVF) and somatic cell nuclear transfer (SCNT) embryos. bESC were efficiently derived from SCNT and IVF embryos and expressed pluripotency markers while retaining genome stability. Transcriptome analysis revealed that only 46 genes were differentially expressed between IVF- and SCNT-derived bESC, which did not reflect significant deviation in cellular function. Interrogating histone 3 lysine 4 trimethylation, histone 3 lysine 9 trimethylation, and histone 3 lysine 27 trimethylation with cleavage under targets and tagmentation, we found that the epigenomes of both bESC groups were virtually indistinguishable. Minor epigenetic differences were randomly distributed throughout the genome and were not associated with differentially expressed or developmentally important genes. Finally, the categorization of genomic regions according to their combined histone mark signal demonstrated that all bESC shared the same epigenomic signatures, especially at gene promoters. Overall, we conclude that bESC derived from SCNT and IVF embryos are transcriptomically and epigenetically analogous, allowing for the production of an unlimited source of pluripotent cells from high genetic merit organisms without resorting to transgene-based techniques.
Collapse
Affiliation(s)
- M Navarro
- Instituto de Investigaciones Biotecnológicas 'Dr Rodolfo Ugalde', UNSAM-CONICET, Buenos Aires, Argentina
- Department of Animal Science, University of California, Davis, California, USA
| | - M M Halstead
- Department of Animal Science, University of California, Davis, California, USA
| | | | - A A Mutto
- Instituto de Investigaciones Biotecnológicas 'Dr Rodolfo Ugalde', UNSAM-CONICET, Buenos Aires, Argentina
| | - P J Ross
- Department of Animal Science, University of California, Davis, California, USA
| |
Collapse
|
4
|
Said YM, El-Gamel NEA, Ali SA, Mohamed AF. Evaluation of Human Wharton's Jelly-Derived Mesenchymal Stem Cells Conditioning Medium (hWJ-MSCs-CM) or Scorpion Venom Breast Cancer Cell Line In Vitro. J Gastrointest Cancer 2022; 53:888-901. [PMID: 34988906 DOI: 10.1007/s12029-021-00762-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE The present study aimed to evaluate the anticancer potential of Egyptian scorpion Leiurus quinquestriatus venom (ScV) or human Wharton's jelly-derived mesenchymal stem cells conditioning medium (hWJ-MSCs-CM)/CM against breast cancer (MCF-7) cell line as an alternative effective cancer biotherapy. METHODS Venom (ScV) toxicity was performed recording concentration-dependent viability % and ScV IC50 value was in the order of 100 μg/ml. MCF-7 were treated with hWJ-MSCs-CM used as (25%, 50%, and 75% ml) or the IC50 of ScV. Apoptotic activity was traced via evaluation the apoptotic (Bax, Casp-3, and Casp-9) and anti-apoptotic genes (Bcl2, ALDOA, and PKM2) profile. RESULTS Both Bax and Casp-3 showed a significant upregulation while anti-apoptotic genes were significantly downregulated. In the meantime, Casp-3 and Casp-9 protein were monitored using ELISA, and their level was less than in control. Additionally, MCF-7 apoptosis was monitored using flow cytometry recording a significant DNA accumulation in the G0-G1 and S phases in case of cell treatment with ScV or CM75% ml and 50% ml. Also, there was a significant total necrotic cells % compared with control cells, and total apoptosis under the effect of ScV or CM75% ml was significantly elevated than rest of treatment. CONCLUSION Apoptosis induction was both dose- and time-dependent for hWJ-MSCs-CM and ScV. According to the present study and other studies, there is an ample evidence that hWJ-MSCs-CM and the venom IC50 abolish tumor growth.
Collapse
Affiliation(s)
| | - Nadia E A El-Gamel
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt.
| | - Said A Ali
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| | - Aly Fahmy Mohamed
- The International Center for Advanced Researches (ICTAR), Giza, Egypt
| |
Collapse
|
5
|
Li C, Mills Z, Zheng Z. Novel cell sources for bone regeneration. MedComm (Beijing) 2021; 2:145-174. [PMID: 34766140 PMCID: PMC8491221 DOI: 10.1002/mco2.51] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 01/09/2023] Open
Abstract
A plethora of both acute and chronic conditions, including traumatic, degenerative, malignant, or congenital disorders, commonly induce bone disorders often associated with severe persisting pain and limited mobility. Over 1 million surgical procedures involving bone excision, bone grafting, and fracture repair are performed each year in the U.S. alone, resulting in immense levels of public health challenges and corresponding financial burdens. Unfortunately, the innate self-healing capacity of bone is often inadequate for larger defects over a critical size. Moreover, as direct transplantation of committed osteoblasts is hindered by deficient cell availability, limited cell spreading, and poor survivability, an urgent need for novel cell sources for bone regeneration is concurrent. Thanks to the development in stem cell biology and cell reprogramming technology, many multipotent and pluripotent cells that manifest promising osteogenic potential are considered the regenerative remedy for bone defects. Considering these cells' investigation is still in its relative infancy, each of them offers their own particular challenges that must be conquered before the large-scale clinical application.
Collapse
Affiliation(s)
- Chenshuang Li
- Department of Orthodontics, School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Zane Mills
- College of DentistryUniversity of OklahomaOklahoma CityOklahomaUSA
| | - Zhong Zheng
- Division of Growth and Development, School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
- Department of Surgery, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
6
|
Embryonic Environmental Niche Reprograms Somatic Cells to Express Pluripotency Markers and Participate in Adult Chimaeras. Cells 2021; 10:cells10030490. [PMID: 33668852 PMCID: PMC7996319 DOI: 10.3390/cells10030490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/12/2021] [Accepted: 02/18/2021] [Indexed: 12/31/2022] Open
Abstract
The phenomenon of the reprogramming of terminally differentiated cells can be achieved by various means, like somatic cell nuclear transfer, cell fusion with a pluripotent cell, or the introduction of pluripotency genes. Here, we present the evidence that somatic cells can attain the expression of pluripotency markers after their introduction into early embryos. Mouse embryonic fibroblasts introduced between blastomeres of cleaving embryos, within two days of in vitro culture, express transcription factors specific to blastocyst lineages, including pluripotency factors. Analysis of donor tissue marker DNA has revealed that the progeny of introduced cells are found in somatic tissues of foetuses and adult chimaeras, providing evidence for cell reprogramming. Analysis of ploidy has shown that in the chimaeras, the progeny of introduced cells are either diploid or tetraploid, the latter indicating cell fusion. The presence of donor DNA in diploid cells from chimaeric embryos proved that the non-fused progeny of introduced fibroblasts persisted in chimaeras, which is evidence of reprogramming by embryonic niche. When adult somatic (cumulus) cells were introduced into early cleavage embryos, the extent of integration was limited and only cell fusion-mediated reprogramming was observed. These results show that both cell fusion and cell interactions with the embryonic niche reprogrammed somatic cells towards pluripotency.
Collapse
|
7
|
Luo J, Liu P, Wang L, Huang Y, Wang Y, Geng W, Chen D, Bai Y, Yang Z. Establishment of an immune-related gene pair model to predict colon adenocarcinoma prognosis. BMC Cancer 2020; 20:1071. [PMID: 33167940 PMCID: PMC7654612 DOI: 10.1186/s12885-020-07532-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023] Open
Abstract
Background Colon cancer is the most common type of gastrointestinal cancer and has high morbidity and mortality. Colon adenocarcinoma (COAD) is the main pathological type of colon cancer, and much evidence has supported the correlation between the prognosis of COAD and the immune system. The current study aimed to develop a robust prognostic immune-related gene pair (IRGP) model to estimate the overall survival of patients with COAD. Methods The gene expression profiles and clinical information of patients with colon adenocarcinoma were obtained from the TCGA and GEO databases and were divided into training and validation cohorts. Immune genes were selected that showed a significant association with prognosis. Results Among 1647 immune genes, a model with 17 IRGPs was built that was significantly associated with OS in the training cohort. In the training and validation datasets, the IRGP model divided patients into the high-risk group and low-risk group, and the prognosis of the high-risk group was significantly worse (P<0.001). Univariate and multivariate Cox proportional hazard analyses confirmed the feasibility of this model. Functional analysis confirmed that multiple tumor progression and stem cell growth-related pathways were upregulated in the high-risk groups. Regulatory T cells and macrophages M0 were significantly highly expressed in the high-risk group. Conclusion We successfully constructed an IRGP model that can predict the prognosis of COAD, providing new insights into the treatment strategy of COAD. Supplementary information Supplementary information accompanies this paper at 10.1186/s12885-020-07532-7.
Collapse
Affiliation(s)
- Jihang Luo
- Cancer Hospital, Second Affiliated Hospital of Zunyi Medical University, Zunyi City, 563000, Guizhou Province, China
| | - Puyu Liu
- Department of Pathology, Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Leibo Wang
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Yi Huang
- Cancer Hospital, Second Affiliated Hospital of Zunyi Medical University, Zunyi City, 563000, Guizhou Province, China
| | - Yuanyan Wang
- Cancer Hospital, Second Affiliated Hospital of Zunyi Medical University, Zunyi City, 563000, Guizhou Province, China
| | - Wenjing Geng
- Cancer Hospital, Second Affiliated Hospital of Zunyi Medical University, Zunyi City, 563000, Guizhou Province, China
| | - Duo Chen
- Cancer Hospital, Second Affiliated Hospital of Zunyi Medical University, Zunyi City, 563000, Guizhou Province, China
| | - Yuju Bai
- Cancer Hospital, Second Affiliated Hospital of Zunyi Medical University, Zunyi City, 563000, Guizhou Province, China.
| | - Ze Yang
- Cancer Hospital, Second Affiliated Hospital of Zunyi Medical University, Zunyi City, 563000, Guizhou Province, China.
| |
Collapse
|
8
|
Kunishige R, Kano F, Murata M. The cell resealing technique for manipulating, visualizing, and elucidating molecular functions in living cells. Biochim Biophys Acta Gen Subj 2020; 1864:129329. [DOI: 10.1016/j.bbagen.2019.03.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/05/2019] [Accepted: 03/20/2019] [Indexed: 12/19/2022]
|
9
|
Gao G, Banfield DK. Multiple features within the syntaxin Sed5p mediate its Golgi localization. Traffic 2020; 21:274-296. [DOI: 10.1111/tra.12720] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Guanbin Gao
- The Division of Life ScienceThe Hong Kong University of Science and Technology Hong Kong
| | - David K. Banfield
- The Division of Life ScienceThe Hong Kong University of Science and Technology Hong Kong
| |
Collapse
|
10
|
Zheng Z, Li C, Ha P, Chang GX, Yang P, Zhang X, Kim JK, Jiang W, Pang X, Berthiaume EA, Mills Z, Haveles CS, Chen E, Ting K, Soo C. CDKN2B upregulation prevents teratoma formation in multipotent fibromodulin-reprogrammed cells. J Clin Invest 2019; 129:3236-3251. [PMID: 31305260 PMCID: PMC6668700 DOI: 10.1172/jci125015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 05/21/2019] [Indexed: 12/11/2022] Open
Abstract
Tumorigenicity is a well-documented risk to overcome for pluripotent or multipotent cell applications in regenerative medicine. To address the emerging demand for safe cell sources in tissue regeneration, we established a novel, protein-based reprogramming method that does not require genome integration or oncogene activation to yield multipotent fibromodulin (FMOD)-reprogrammed (FReP) cells from dermal fibroblasts. When compared with induced pluripotent stem cells (iPSCs), FReP cells exhibited a superior capability for bone and skeletal muscle regeneration with markedly less tumorigenic risk. Moreover, we showed that the decreased tumorigenicity of FReP cells was directly related to an upregulation of cyclin-dependent kinase inhibitor 2B (CDKN2B) expression during the FMOD reprogramming process. Indeed, sustained suppression of CDKN2B resulted in tumorigenic, pluripotent FReP cells that formed teratomas in vivo that were indistinguishable from iPSC-derived teratomas. These results highlight the pivotal role of CDKN2B in cell fate determination and tumorigenic regulation and reveal an alternative pluripotent/multipotent cell reprogramming strategy that solely uses FMOD protein.
Collapse
Affiliation(s)
- Zhong Zheng
- Division of Growth and Development, School of Dentistry, and
| | - Chenshuang Li
- Division of Growth and Development, School of Dentistry, and
| | - Pin Ha
- Division of Growth and Development, School of Dentistry, and
| | - Grace X. Chang
- David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Pu Yang
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xinli Zhang
- Division of Growth and Development, School of Dentistry, and
| | - Jong Kil Kim
- Division of Growth and Development, School of Dentistry, and
| | - Wenlu Jiang
- Division of Growth and Development, School of Dentistry, and
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoxiao Pang
- Division of Growth and Development, School of Dentistry, and
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatology Hospital of Chongqing Medical University, Chongqing, China
| | | | - Zane Mills
- Department of Ecology and Evolutionary Biology, and
| | | | - Eric Chen
- Division of Growth and Development, School of Dentistry, and
| | - Kang Ting
- Division of Growth and Development, School of Dentistry, and
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA, Los Angeles, California, USA
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA, Los Angeles, California, USA
| |
Collapse
|
11
|
Xiong XR, Lan DL, Li J, Yin S, Xiong Y, Zi XD. Effects of Cellular Extract on Epigenetic Reprogramming. Cell Reprogram 2019; 21:115-121. [PMID: 31084436 DOI: 10.1089/cell.2018.0074] [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
Functional reprogramming of a differentiated cell toward pluripotent cell may have long-term applications in numerous aspects, especially in regenerative medicine. Evidences accumulating from recent studies suggest that cellular extracts from stem cells or pluripotent cells can induce epigenetic reprogramming and facilitate pluripotency in otherwise highly differentiated cell types. Epigenetic reprogramming using cellular extracts has gained increasing attention and applied to recognize the functional factors, acquire the target cell types, and explain the mechanism of reprogramming. Now, more and more researches have proved that cellular extract treatment is an important strategy of cellular reprogramming. Thus, this review mainly focused on the progresses and potential mechanisms in epigenetic reprogramming using cellular extracts.
Collapse
Affiliation(s)
- Xian-Rong Xiong
- 1 College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Dao-Liang Lan
- 2 Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Chengdu, China
| | - Jian Li
- 1 College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Shi Yin
- 2 Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Chengdu, China
| | - Yan Xiong
- 1 College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Xiang-Dong Zi
- 1 College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| |
Collapse
|
12
|
Stewart MP, Langer R, Jensen KF. Intracellular Delivery by Membrane Disruption: Mechanisms, Strategies, and Concepts. Chem Rev 2018; 118:7409-7531. [PMID: 30052023 PMCID: PMC6763210 DOI: 10.1021/acs.chemrev.7b00678] [Citation(s) in RCA: 456] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intracellular delivery is a key step in biological research and has enabled decades of biomedical discoveries. It is also becoming increasingly important in industrial and medical applications ranging from biomanufacture to cell-based therapies. Here, we review techniques for membrane disruption-based intracellular delivery from 1911 until the present. These methods achieve rapid, direct, and universal delivery of almost any cargo molecule or material that can be dispersed in solution. We start by covering the motivations for intracellular delivery and the challenges associated with the different cargo types-small molecules, proteins/peptides, nucleic acids, synthetic nanomaterials, and large cargo. The review then presents a broad comparison of delivery strategies followed by an analysis of membrane disruption mechanisms and the biology of the cell response. We cover mechanical, electrical, thermal, optical, and chemical strategies of membrane disruption with a particular emphasis on their applications and challenges to implementation. Throughout, we highlight specific mechanisms of membrane disruption and suggest areas in need of further experimentation. We hope the concepts discussed in our review inspire scientists and engineers with further ideas to improve intracellular delivery.
Collapse
Affiliation(s)
- Martin P. Stewart
- Department of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, USA
- The Koch Institute for Integrative Cancer Research,
Massachusetts Institute of Technology, Cambridge, USA
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, USA
- The Koch Institute for Integrative Cancer Research,
Massachusetts Institute of Technology, Cambridge, USA
| | - Klavs F. Jensen
- Department of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, USA
| |
Collapse
|
13
|
Hu J, Zhao Q, Feng Y, Li N, Gu Y, Sun R, Duan L, Wu Y, Shan Z, Lei L. Embryonic germ cell extracts erase imprinted genes and improve the efficiency of induced pluripotent stem cells. Sci Rep 2018; 8:10955. [PMID: 30026469 PMCID: PMC6053380 DOI: 10.1038/s41598-018-29339-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 07/10/2018] [Indexed: 02/06/2023] Open
Abstract
Patient-specific induced pluripotent stem cells (iPSCs) have the potential to be useful in the treatment of human diseases. While prior studies have reported multiple methods to generate iPSCs, DNA methylation continues to limit the totipotency and reprogramming efficiency of iPSCs. Here, we first show the competency of embryonic germ cells (EGCs) as a reprogramming catalyst capable of effectively promoting reprogramming induced by four defined factors, including Oct4, Sox2, Klf4 and c-Myc. Combining EGC extracts with these four factors resulted in formation of more embryonic stem cell-like colonies than did factors alone. Notably, expression of imprinted genes was higher with combined induction than with factors alone. Moreover, iPSCs derived from the combined inductors tended to have more global hypomethylation. Our research not only provides evidence that EGC extracts could activate DNA demethylation and reprogram imprinted genes, but also establishes a new way to enhance reprogramming of iPSCs, which remains a critical safety concern for potential use of iPSCs in regenerative medicine.
Collapse
Affiliation(s)
- Jing Hu
- Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, P. R. China.,Department of Histology and Embryology, Mudanjiang Medical University, Mudanjiang, 157011, P. R. China
| | - Qiaoshi Zhao
- Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, P. R. China
| | - Yukuan Feng
- Key Laboratory of Tumor Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical University, Mudanjiang, 157011, P. R. China
| | - Na Li
- Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, P. R. China
| | - Yanli Gu
- Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, P. R. China
| | - Ruizhen Sun
- Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, P. R. China
| | - Lian Duan
- Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, P. R. China
| | - Yanshuang Wu
- Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, P. R. China
| | - Zhiyan Shan
- Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, P. R. China.
| | - Lei Lei
- Department of Histology and Embryology, Harbin Medical University, Harbin, 150081, P. R. China.
| |
Collapse
|
14
|
Abstract
Puberty involves a series of morphological, physiological and behavioural changes during the last part of the juvenile period that culminates in the attainment of fertility. The activation of the pituitary-gonadal axis by increased hypothalamic secretion of gonadotrophin-releasing hormone (GnRH) is an essential step in the process. The current hypothesis postulates that a loss of transsynaptic inhibition and a rise in excitatory inputs are responsible for the activation of GnRH release. Similarly, a shift in the balance in the expression of puberty activating and puberty inhibitory genes exists during the pubertal transition. In addition, recent evidence suggests that the epigenetic machinery controls this genetic balance, giving rise to the tantalising possibility that epigenetics serves as a relay of environmental signals known for many years to modulate pubertal development. Here, we review the contribution of epigenetics as a regulatory mechanism in the hypothalamic control of female puberty.
Collapse
Affiliation(s)
- C A Toro
- Primate Genetics Section/Division of Neuroscience, Oregon National Primate Research Center/Oregon Health & Science University, Beaverton, OR, USA
| | - C F Aylwin
- Primate Genetics Section/Division of Neuroscience, Oregon National Primate Research Center/Oregon Health & Science University, Beaverton, OR, USA
| | - A Lomniczi
- Primate Genetics Section/Division of Neuroscience, Oregon National Primate Research Center/Oregon Health & Science University, Beaverton, OR, USA
| |
Collapse
|
15
|
Salehi PM, Foroutan T, Javeri A, Taha MF. Extract of mouse embryonic stem cells induces the expression of pluripotency genes in human adipose tissue-derived stem cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2018; 20:1200-1206. [PMID: 29299196 PMCID: PMC5749353 DOI: 10.22038/ijbms.2017.9464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Objective(s): In some previous studies, the extract of embryonic carcinoma cells (ECCs) and embryonic stem cells (ESCs) have been used to reprogram somatic cells to more dedifferentiated state. The aim of this study was to investigate the effect of mouse ESCs extract on the expression of some pluripotency markers in human adipose tissue-derived stem cells (ADSCs). Materials and Methods: Human ADSCs were isolated from subcutaneous abdominal adipose tissue and characterized by flow cytometric analysis for the expression of some mesenchymal stem cell markers and adipogenic and osteogenic differentiation. Frequent freeze-thaw technique was used to prepare cytoplasmic extract of ESCs. Plasma membranes of the ADSCs were reversibly permeabilized by streptolysin-O (SLO). Then the permeabilized ADSCs were incubated with the ESC extract and cultured in resealing medium. After reprogramming, the expression of some pluripotency genes was evaluated by RT-PCR and quantitative real-time PCR (qPCR) analyses. Results: Third-passaged ADSCs showed a fibroblast-like morphology and expressed mesenchymal stem cell markers. They also showed adipogenic and osteogenic differentiation potential. QPCR analysis revealed a significant upregulation in the expression of some pluripotency genes including OCT4, SOX2, NANOG, REX1 and ESG1 in the reprogrammed ADSCs compared to the control group. Conclusion: These findings showed that mouse ESC extract can be used to induce reprogramming of human ADSCs. In fact, this method is applicable for reprogramming of human adult stem cells to a more pluripotent sate and may have a potential in regenerative medicine.
Collapse
Affiliation(s)
- Paria Motamen Salehi
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.,Department of Biology, Faculty of Basic Science, Kharazmi University, Tehran, Iran
| | - Tahereh Foroutan
- Department of Biology, Faculty of Basic Science, Kharazmi University, Tehran, Iran
| | - Arash Javeri
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Masoumeh Fakhr Taha
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
16
|
Pandey P, Daghma DS, Houben A, Kumlehn J, Melzer M, Rutten T. Dynamics of post-translationally modified histones during barley pollen embryogenesis in the presence or absence of the epi-drug trichostatin A. PLANT REPRODUCTION 2017; 30:95-105. [PMID: 28526911 DOI: 10.1007/s00497-017-0302-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/11/2017] [Indexed: 05/11/2023]
Abstract
Improving pollen embryogenesis. Despite the agro-economic importance of pollen embryogenesis, the mechanisms underlying this process are still poorly understood. We describe the dynamics of chromatin modifications (histones H3K4me2, H3K9ac, H3K9me2, and H3K27me3) and chromatin marks (RNA polymerase II CDC phospho-Ser5, and CENH3) during barley pollen embryogenesis. Immunolabeling results show that, in reaction to stress, immature pollen rapidly starts reorganizing several important chromatin modifications indicative of a change in cell fate. This new chromatin modification pattern was accomplished within 24 h from whereon it remained unaltered during subsequent mitotic activity. This indicates that cell fate transition, the central element of pollen embryogenesis, is completed early on during the induction process. Application of the histone deacetylase inhibitor trichostatin A stimulated pollen embryogenesis when used on pollen with a gametophytic style chromatin pattern. However, when this drug was administered to embryogenic pollen, the chromatin markers reversed toward a gametophytic profile, embryogenesis was halted and all pollen invariably died.
Collapse
Affiliation(s)
- Pooja Pandey
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
- Imperial College London, London, UK
| | - Diaa S Daghma
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
- Institute for Experimental Trauma Surgery, Justus-Liebig University of Giessen, Giessen, Germany
| | - Andreas Houben
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
| | - Jochen Kumlehn
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
| | - Michael Melzer
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
| | - Twan Rutten
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany.
| |
Collapse
|
17
|
Nordin F, Ahmad RNR, Farzaneh F. Transactivator protein: An alternative for delivery of recombinant proteins for safer reprogramming of induced Pluripotent Stem Cell. Virus Res 2017; 235:106-114. [DOI: 10.1016/j.virusres.2017.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/08/2017] [Indexed: 10/19/2022]
|
18
|
Transdifferentiation and reprogramming: Overview of the processes, their similarities and differences. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1359-1369. [PMID: 28460880 DOI: 10.1016/j.bbamcr.2017.04.017] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 12/24/2022]
Abstract
Reprogramming, or generation of induced pluripotent stem (iPS) cells (functionally similar to embryonic stem cells or ES cells) by the use of transcription factors (typically: Oct3/4, Sox2, c-Myc, Klf4) called "Yamanaka factors" (OSKM), has revolutionized regenerative medicine. However, factors used to induce stemness are also overexpressed in cancer. Both, ES cells and iPS cells cause teratoma formation when injected to tissues. This raises a safety concern for therapies based on iPS derivates. Transdifferentiation (lineage reprogramming, or -conversion), is a process in which one mature, specialized cell type changes into another without entering a pluripotent state. This process involves an ectopic expression of transcription factors and/or other stimuli. Unlike in the case of reprogramming, tissues obtained by this method do not carry the risk of subsequent teratomagenesis.
Collapse
|
19
|
Induction of male germ cell-like lineage from chicken fetal bone marrow stem cells with chicken testis extract. BIOTECHNOL BIOPROC E 2017. [DOI: 10.1007/s12257-016-0629-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
20
|
Cellular Reprogramming Using Protein and Cell-Penetrating Peptides. Int J Mol Sci 2017; 18:ijms18030552. [PMID: 28273812 PMCID: PMC5372568 DOI: 10.3390/ijms18030552] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 02/27/2017] [Accepted: 02/28/2017] [Indexed: 12/13/2022] Open
Abstract
Recently, stem cells have been suggested as invaluable tools for cell therapy because of their self-renewal and multilineage differentiation potential. Thus, scientists have developed a variety of methods to generate pluripotent stem cells, from nuclear transfer technology to direct reprogramming using defined factors, or induced pluripotent stem cells (iPSCs). Considering the ethical issues and efficiency, iPSCs are thought to be one of the most promising stem cells for cell therapy. Induced pluripotent stem cells can be generated by transduction with a virus, plasmid, RNA, or protein. Herein, we provide an overview of the current technology for iPSC generation and describe protein-based transduction technology in detail.
Collapse
|
21
|
Mirahmadi M, Nakhaei-Rad S, Matin MM, Shahriyari M, Saeinasab M, Mahmoudi Z, Haghighitalab A, Mahdavi-Shahri N, Bahrami AR. Dedifferentiation Effects of Rabbit Regenerating Tissue on Partially Differentiated Cells. Cell Reprogram 2016; 18:333-343. [PMID: 27602600 DOI: 10.1089/cell.2016.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cell Stemness can be achieved by various reprogramming techniques namely, somatic cell nuclear transfer, cell fusion, cell extracts, and introduction of transcription factors from which induced pluripotent stem cells (iPSCs) are obtained. iPSCs are valuable cell sources for drug screening and human disease modeling. Alternatives to virus-based introduction of transcription factors include application of DNA-free methods and introduction of chemically defined culturing conditions. However, the possibility of tumor development is still a hurdle. By taking advantage of NTERA-2 cells, a human embryonal carcinoma cell line, we obtained partially differentiated cells and examined the dedifferentiation capacity of regenerative tissue from rabbit ears. Results indicated that treatment of partially differentiated NTERA-2 cells with the regenerating tissue-conditioned medium (CM) induced expression of key pluripotency markers as examined by real-time polymerase chain reaction, flow cytometry, and immunocytochemistry techniques. In this study, it is reported for the first time that the CM obtained from rabbit regenerating tissue contains dedifferentiation factors, taking cells back to the pluripotency. This system could be a simple and efficient way to reprogram the differentiated cells and generate iPSCs for clinical applications as this system is not accompanied by any viral vector, and reprograms the cells within 10 days of treatment. The results may convince the genomic experts to study the unknown signaling pathways involved in the dedifferentiation by regenerating tissue-CM to authenticate the reprogramming model.
Collapse
Affiliation(s)
- Mahdi Mirahmadi
- 1 Department of Biology, Faculty of Science, Ferdowsi University of Mashhad , Mashhad, Iran
- 2 Stem Cell and Regenerative Research Group, Iranian Academic Center for Education , Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Saeideh Nakhaei-Rad
- 1 Department of Biology, Faculty of Science, Ferdowsi University of Mashhad , Mashhad, Iran
- 3 Institute of Biochemistry and Molecular Biology II, Heinrich-Heine University , Düsseldorf, Germany
| | - Maryam M Matin
- 1 Department of Biology, Faculty of Science, Ferdowsi University of Mashhad , Mashhad, Iran
- 2 Stem Cell and Regenerative Research Group, Iranian Academic Center for Education , Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
- 4 Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad , Mashhad, Iran
| | - Mina Shahriyari
- 1 Department of Biology, Faculty of Science, Ferdowsi University of Mashhad , Mashhad, Iran
| | - Morvarid Saeinasab
- 1 Department of Biology, Faculty of Science, Ferdowsi University of Mashhad , Mashhad, Iran
| | - Zahra Mahmoudi
- 1 Department of Biology, Faculty of Science, Ferdowsi University of Mashhad , Mashhad, Iran
- 4 Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad , Mashhad, Iran
| | - Azadeh Haghighitalab
- 1 Department of Biology, Faculty of Science, Ferdowsi University of Mashhad , Mashhad, Iran
- 4 Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad , Mashhad, Iran
| | - Naser Mahdavi-Shahri
- 1 Department of Biology, Faculty of Science, Ferdowsi University of Mashhad , Mashhad, Iran
| | - Ahmad Reza Bahrami
- 1 Department of Biology, Faculty of Science, Ferdowsi University of Mashhad , Mashhad, Iran
- 4 Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad , Mashhad, Iran
| |
Collapse
|
22
|
Abstract
The derivation of human embryonic stem (hES) cells heralds a new era in stem cell research, generating excitement for their therapeutic potential in regenerative medicine. Pioneering work of embryologists, developmental biologists, and reproductive medicine practitioners in in vitro fertilization clinics has facilitated hES cell research. This review summarizes current research focused on optimizing hES cell culture conditions for good manufacturing practice, directing hES cell differentiation toward trophectoderm and germ cells, and approaches used to reprogram cells for pluripotent cell derivation. The identification of germ stem cells in the testis and the recent controversy over their existence in the ovary raise the possibility of harnessing them for treating young cancer survivors. There is also the potential to harvest fetal stem cells with pluripotent cell-like properties from discarded placental tissues. The recent identification of adult stem/progenitor cell activity in the human endometrium offers a new understanding of common gynecological diseases. Discoveries resulting from research into embryonic, germ, fetal, and adult stem cells are highly relevant to human reproduction.
Collapse
Affiliation(s)
- Caroline E Gargett
- Centre for Women's Health Research, Monash Institute of Medical Research, and Monash University Department of Obstetrics and Gynaecology, Monash Medical Centre, Clayton, Victoria, Australia.
| |
Collapse
|
23
|
Holmqvist S, Lehtonen Š, Chumarina M, Puttonen KA, Azevedo C, Lebedeva O, Ruponen M, Oksanen M, Djelloul M, Collin A, Goldwurm S, Meyer M, Lagarkova M, Kiselev S, Koistinaho J, Roybon L. Creation of a library of induced pluripotent stem cells from Parkinsonian patients. NPJ Parkinsons Dis 2016; 2:16009. [PMID: 28725696 PMCID: PMC5516589 DOI: 10.1038/npjparkd.2016.9] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/01/2016] [Accepted: 04/11/2016] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) are becoming an important source of pre-clinical models for research focusing on neurodegeneration. They offer the possibility for better understanding of common and divergent pathogenic mechanisms of brain diseases. Moreover, iPSCs provide a unique opportunity to develop personalized therapeutic strategies, as well as explore early pathogenic mechanisms, since they rely on the use of patients' own cells that are otherwise accessible only post-mortem, when neuronal death-related cellular pathways and processes are advanced and adaptive. Neurodegenerative diseases are in majority of unknown cause, but mutations in specific genes can lead to familial forms of these diseases. For example, mutations in the superoxide dismutase 1 gene lead to the motor neuron disease amyotrophic lateral sclerosis (ALS), while mutations in the SNCA gene encoding for alpha-synuclein protein lead to familial Parkinson's disease (PD). The generations of libraries of familial human ALS iPSC lines have been described, and the iPSCs rapidly became useful models for studying cell autonomous and non-cell autonomous mechanisms of the disease. Here we report the generation of a comprehensive library of iPSC lines of familial PD and an associated synucleinopathy, multiple system atrophy (MSA). In addition, we provide examples of relevant neural cell types these iPSC can be differentiated into, and which could be used to further explore early disease mechanisms. These human cellular models will be a valuable resource for identifying common and divergent mechanisms leading to neurodegeneration in PD and MSA.
Collapse
Affiliation(s)
- Staffan Holmqvist
- Stem Cell Laboratory for CNS Disease Modeling, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, Lund University, Lund, Sweden
- Strategic Research Area MultiPark, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Šárka Lehtonen
- Stem Cell Laboratory of Molecular Brain Research Group, Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Margarita Chumarina
- Stem Cell Laboratory for CNS Disease Modeling, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, Lund University, Lund, Sweden
- Strategic Research Area MultiPark, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Katja A Puttonen
- Stem Cell Laboratory of Molecular Brain Research Group, Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Carla Azevedo
- Stem Cell Laboratory for CNS Disease Modeling, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, Lund University, Lund, Sweden
- Strategic Research Area MultiPark, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Olga Lebedeva
- Russian Academy of Sciences, Vavilov Institute of General Genetics, Moscow, Russia
| | - Marika Ruponen
- Stem Cell Laboratory of Molecular Brain Research Group, Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Minna Oksanen
- Stem Cell Laboratory of Molecular Brain Research Group, Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Mehdi Djelloul
- Stem Cell Laboratory for CNS Disease Modeling, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, Lund University, Lund, Sweden
- Strategic Research Area MultiPark, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Anna Collin
- Department of Clinical Genetics and Biobanks, Office for Medical Services, Division of Laboratory Medicine, Lund, Sweden
| | - Stefano Goldwurm
- Parkinson Institute, Istituti Clinici di Perfezionamento, Milan, Italy
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Maria Lagarkova
- Russian Academy of Sciences, Vavilov Institute of General Genetics, Moscow, Russia
| | - Sergei Kiselev
- Russian Academy of Sciences, Vavilov Institute of General Genetics, Moscow, Russia
| | - Jari Koistinaho
- Stem Cell Laboratory of Molecular Brain Research Group, Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Laurent Roybon
- Stem Cell Laboratory for CNS Disease Modeling, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, Lund University, Lund, Sweden
- Strategic Research Area MultiPark, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
24
|
Kharizinejad E, Minaee Zanganeh B, Khanlarkhani N, Mortezaee K, Rastegar T, Baazm M, Abolhassani F, Sajjadi SM, Hajian M, Aliakbari F, Barbarestani M. Role of spermatogonial stem cells extract in transdifferentiation of 5-Aza-2'-deoxycytidine-treated bone marrow mesenchymal stem cells into germ-like cells. Microsc Res Tech 2016; 79:365-73. [PMID: 26969916 DOI: 10.1002/jemt.22639] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 01/19/2016] [Accepted: 01/24/2016] [Indexed: 01/22/2023]
Abstract
As one of the induced pluripotent stem cells (iPSCs) methods, spermatogonial stem cells (SSCS ) extract is considered as new approach in stem cell therapy of infertility. 5-aza-2'-deoxycytidine (5-aza-dC) inhibits methyltransferase enzyme, and induces gene reprogramming; herein, the effects of SSCS extract incubation in 5-aza-dC-treated bone marrow mesenchymal stem cells (BMMSCs) has been surveyed. BMMSCs were isolated from femurs of three to four weeks old male NMRI mice, and the cells at passage three were treated with 2 µM 5-aza-dC for 72 hours. SSCs were isolated, cultured, and harvested at passage three to collect SSCS extract; BMMSCs were then incubated with SSCS extract in the three time periods: 72 hours, one week and two weeks. There were five groups: control, sham, extract, 5-aza-dC and extract-5-aza-dC. After one week of incubation, flow cytometry and real-time polymerase chain reaction (PCR) exhibited high levels of expression for β1- and α6-integrins and promyelocytic leukaemia zinc finger (PLZF) in extract and extract-5-aza-dC groups (P < 0.05 vs. control and 5-aza-dC), and cells in these two groups had two forms of morphology, round and fusiform, similar to germ-like cells. 5-aza-dC had no significant effects during the three time periods of evaluation. These data disclose the effectiveness of SSCs extract incubation in transdifferentiation of BMMSCs into germ-like cells; this strategy could introduce a new approach for treatment of male infertility in clinic.
Collapse
Affiliation(s)
- Ebrahim Kharizinejad
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Minaee Zanganeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Khanlarkhani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Rastegar
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Baazm
- Department of Anatomy, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Farid Abolhassani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mehdi Sajjadi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahdieh Hajian
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fereshte Aliakbari
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Barbarestani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Monaghan MG, Holeiter M, Layland SL, Schenke-Layland K. Cardiomyocyte generation from somatic sources - current status and future directions. Curr Opin Biotechnol 2016; 40:49-55. [PMID: 26945640 DOI: 10.1016/j.copbio.2016.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 02/11/2016] [Accepted: 02/15/2016] [Indexed: 12/16/2022]
Abstract
Transdifferentiation of one cell type to another has garnered significant research efforts in recent years. As cardiomyocyte loss following myocardial infarction becomes debilitating for cardiac patients, the option of an autologous source of cardiomyocytes not derived from multi/pluripotent stem cell sources is an attractive option. Such direct programming has been clearly realized with the use of transcription factors, microRNAs and more recently small molecule delivery to enhance epigenetic modifications, all albeit with low efficiencies in vitro. In this review, we aim to present a brief overview of the current in vitro and in vivo transdifferentiation strategies in the generation of cardiomyocytes from somatic sources. The interdisciplinary fields of tissue, cell, material and regenerative engineering offer many opportunities to synergistically achieve directly programmed cardiac tissue in vitro and enhance transdifferentiation in vivo. This review aims to present a concise outlook on this topic with these fields in mind.
Collapse
Affiliation(s)
- Michael G Monaghan
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University, Tübingen, Germany
| | - Monika Holeiter
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University, Tübingen, Germany
| | - Shannon L Layland
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University, Tübingen, Germany; Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology (IGB), Stuttgart, Germany
| | - Katja Schenke-Layland
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University, Tübingen, Germany; Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology (IGB), Stuttgart, Germany; Department of Medicine/Cardiology, Cardiovascular Research Laboratories, University of California, Los Angeles, CA, USA.
| |
Collapse
|
26
|
Li CS, Yang P, Ting K, Aghaloo T, Lee S, Zhang Y, Khalilinejad K, Murphy MC, Pan HC, Zhang X, Wu B, Zhou YH, Zhao Z, Zheng Z, Soo C. Fibromodulin reprogrammed cells: A novel cell source for bone regeneration. Biomaterials 2016; 83:194-206. [PMID: 26774565 PMCID: PMC4754141 DOI: 10.1016/j.biomaterials.2016.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/27/2015] [Accepted: 01/01/2016] [Indexed: 02/05/2023]
Abstract
Pluripotent or multipotent cell-based therapeutics are vital for skeletal reconstruction in non-healing critical-sized defects since the local endogenous progenitor cells are not often adequate to restore tissue continuity or function. However, currently available cell-based regenerative strategies are hindered by numerous obstacles including inadequate cell availability, painful and invasive cell-harvesting procedures, and tumorigenesis. Previously, we established a novel platform technology for inducing a quiescent stem cell-like stage using only a single extracellular proteoglycan, fibromodulin (FMOD), circumventing gene transduction. In this study, we further purified and significantly increased the reprogramming rate of the yield multipotent FMOD reprogrammed (FReP) cells. We also exposed the 'molecular blueprint' of FReP cell osteogenic differentiation by gene profiling. Radiographic analysis showed that implantation of FReP cells into a critical-sized SCID mouse calvarial defect, contributed to the robust osteogenic capability of FReP cells in a challenging clinically relevant traumatic scenario in vivo. The persistence, engraftment, and osteogenesis of transplanted FReP cells without tumorigenesis in vivo were confirmed by histological and immunohistochemical staining. Taken together, we have provided an extended potency, safety, and molecular profile of FReP cell-based bone regeneration. Therefore, FReP cells present a high potential for cellular and gene therapy products for bone regeneration.
Collapse
Affiliation(s)
- Chen-Shuang Li
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA; Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Pu Yang
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Kang Ting
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Tara Aghaloo
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Soonchul Lee
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA; Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University, Gyeonggi-do, 463-712, South Korea
| | - Yulong Zhang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kambiz Khalilinejad
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Maxwell C Murphy
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hsin Chuan Pan
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xinli Zhang
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Benjamin Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yan-Heng Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Zhong Zheng
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Chia Soo
- UCLA Division of Plastic Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
27
|
Components of chicken egg white extract smaller than 3 kDa in size promote 293T cell proliferation. Cytotechnology 2015; 68:1115-22. [PMID: 26541834 DOI: 10.1007/s10616-015-9868-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 03/23/2015] [Indexed: 10/22/2022] Open
Abstract
We previously found that chicken egg white extract could promote cell survival and proliferation. In the present study, we further separated this extract into its components to identify those primarily responsible for promoting cell proliferation. Components of differing molecular weight were separated from chicken egg white extract by ultrafiltration and 293T cell cultures were supplemented with various concentrations. The effects on cell proliferation were subsequently determined by a CellTiter 96 Aqueous One Solution Cell Proliferation Assay kit (Promega). We demonstrate that components from chicken egg white smaller than 3 kDa in size are able to function as active ingredients promoting cellular proliferation. This discovery may identify a new and convenient additive for cell culture media to promote cell growth and proliferation.
Collapse
|
28
|
Zhu XQ, Pan XH, Yao L, Li W, Cui J, Wang G, Mrsny RJ, Hoffman AR, Hu JF. Converting Skin Fibroblasts into Hepatic-like Cells by Transient Programming. J Cell Biochem 2015; 117:589-98. [PMID: 26312781 DOI: 10.1002/jcb.25355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 08/25/2015] [Indexed: 12/30/2022]
Affiliation(s)
- Xiang-Qing Zhu
- Research Center of Stem Cell, Tissue and Organ Engineering; Kunming Army General Hospital; Kunming Yunnan P. R. China
| | - Xing-Hua Pan
- Research Center of Stem Cell, Tissue and Organ Engineering; Kunming Army General Hospital; Kunming Yunnan P. R. China
| | - Ling Yao
- Stanford University Medical School; Palo Alto California
| | - Wei Li
- Stem Cell and Cancer Center; The First Affiliated Hospital; Jilin University; Changchun P. R. China
| | - Jiuwei Cui
- Stem Cell and Cancer Center; The First Affiliated Hospital; Jilin University; Changchun P. R. China
| | - Guanjun Wang
- Stem Cell and Cancer Center; The First Affiliated Hospital; Jilin University; Changchun P. R. China
| | - Randall J. Mrsny
- GMR Epigenetics; Palo Alto California
- Department of Pharmacy & Pharmacology; University of Bath; Bath England
| | | | - Ji-Fan Hu
- Stanford University Medical School; Palo Alto California
- Stem Cell and Cancer Center; The First Affiliated Hospital; Jilin University; Changchun P. R. China
| |
Collapse
|
29
|
Bose B, Shenoy P S. Pluripotent Conversion of Muscle Stem Cells Without Reprogramming Factors or Small Molecules. Stem Cell Rev Rep 2015; 12:73-89. [PMID: 26358783 DOI: 10.1007/s12015-015-9620-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Muscle derived stem cells (MDSCs) are multipotent stem cells that can differentiate into several lineages including skeletal muscle precursor cells. Here, we show that MDSCs from myostatin null mice (Mstn (-/-) ) can be readily induced into pluripotent stem cells without using reprogramming factors. Microarray studies revealed a strong upregulation of markers like Leukemia Inhibitory factor (LIF) and Leukemia Inhibitory factor receptor (LIFR) in Mstn (-/-) MDSCs as compared to wild type MDSCs (WT-MDSCs). Furthermore when cultured in mouse embryonic stem cell media with LIF for 95 days, Mstn (-/-) MDSCs formed embryonic stem cell (ES) like colonies. We termed such ES like cells as the culture-induced pluripotent stem cells (CiPSC). CiPSCs from Mstn (-/-) MDSCs were phenotypically similar to ESCs, expressed high levels of Oct4, Nanog, Sox2 and SSEA-1, maintained a normal karyotype. Furthermore, CiPSCs formed embryoid bodies and teratomas when injected into immunocompromised mice. In addition, CiPSCs differentiated into somatic cells of all three lineages. We further show that culturing in ES cell media, resulted in hypermethylation and downregulation of BMP2 in Mstn(-/-) MDSCs. Western blot further confirmed a down regulation of BMP2 signaling in Mstn (-/-) MDSCs in supportive of pluripotent reprogramming. Given that down regulation of BMP2 has been shown to induce pluripotency in cells, we propose that lack of myostatin epigenetically reprograms the MDSCs to become pluripotent stem cells. Thus, here we report the successful establishment of ES-like cells from adult stem cells of the non-germline origin under culture-induced conditions without introducing reprogramming genes.
Collapse
Affiliation(s)
- Bipasha Bose
- School of Biological Sciences, Nanyang Technological University, 60, Nanyang Drive, Singapore, 637551, Singapore.
- Stem Cell and Tissue Engineering Division, Yenepoya Research Center, Yenepoya University, University Road, Derlakatte, Mangalore, Karnataka, 575018, India.
| | - Sudheer Shenoy P
- School of Biological Sciences, Nanyang Technological University, 60, Nanyang Drive, Singapore, 637551, Singapore.
- Stem Cell and Tissue Engineering Division, Yenepoya Research Center, Yenepoya University, University Road, Derlakatte, Mangalore, Karnataka, 575018, India.
| |
Collapse
|
30
|
Sun Y, Guo F, Bagnoli M, Xue FX, Sun BC, Shmulevich I, Mezzanzanica D, Chen KX, Sood AK, Yang D, Zhang W. Key nodes of a microRNA network associated with the integrated mesenchymal subtype of high-grade serous ovarian cancer. CHINESE JOURNAL OF CANCER 2015; 34:28-40. [PMID: 25556616 PMCID: PMC4302087 DOI: 10.5732/cjc.014.10284] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metastasis is the main cause of cancer mortality. One of the initiating events of cancer metastasis of epithelial tumors is epithelial-to-mesenchymal transition (EMT), during which cells dedifferentiate from a relatively rigid cell structure/morphology to a flexible and changeable structure/morphology often associated with mesenchymal cells. The presence of EMT in human epithelial tumors is reflected by the increased expression of genes and levels of proteins that are preferentially present in mesenchymal cells. The combined presence of these genes forms the basis of mesenchymal gene signatures, which are the foundation for classifying a mesenchymal subtype of tumors. Indeed, tumor classification schemes that use clustering analysis of large genomic characterizations, like The Cancer Genome Atlas (TCGA), have defined mesenchymal subtype in a number of cancer types, such as high-grade serous ovarian cancer and glioblastoma. However, recent analyses have shown that gene expression-based classifications of mesenchymal subtypes often do not associate with poor survival. This “paradox” can be ameliorated using integrated analysis that combines multiple data types. We recently found that integrating mRNA and microRNA (miRNA) data revealed an integrated mesenchymal subtype that is consistently associated with poor survival in multiple cohorts of patients with serous ovarian cancer. This network consists of 8 major miRNAs and 214 mRNAs. Among the 8 miRNAs, 4 are known to be regulators of EMT. This review provides a summary of these 8 miRNAs, which were associated with the integrated mesenchymal subtype of serous ovarian cancer.
Collapse
Affiliation(s)
- Yan Sun
- Departments of Pathology, The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P. R. China. ,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Liu Y, Ostrup O, Li R, Li J, Vajta G, Kragh PM, Schmidt M, Purup S, Hyttel P, Klærke D, Callesen H. Long-term effect on in vitro cloning efficiency after treatment of somatic cells with Xenopus egg extract in the pig. Reprod Fertil Dev 2015; 26:1017-31. [PMID: 25145414 DOI: 10.1071/rd13147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 07/02/2013] [Indexed: 11/23/2022] Open
Abstract
In somatic cell nuclear transfer (SCNT), donor cell reprogramming is considered as a biologically important and vulnerable event. Various donor cell pre-treatments with Xenopus egg extracts can promote reprogramming. Here we investigated if the reprogramming effect of one treatment with Xenopus egg extract on donor cells was maintained for several cell passages. The extract treatment resulted in increased cell-colony formation from early passages in treated porcine fibroblasts (ExTES), and increased development of cloned embryos. Partial dedifferentiation was observed in ExTES cells, shown as a tendency towards upregulation of NANOG, c-MYC and KLF-4 and downregulation of DESMIM compared with ExTES at Passage 2. Compared with our routine SCNT, continuously increased development of cloned embryos was observed in the ExTES group, and ExTES cloned blastocysts displayed hypermethylated DNA patterns and hypermethylation of H3K4me3 and H3K27me3 in ICM compared with TE. All seven recipients became pregnant after transferral of ExTES cloned embryos and gave birth to 7-22 piglets per litter (average 12). In conclusion, our results demonstrate that one treatment of porcine fibroblasts with Xenopus egg extract can result in long-term increased ability of the cells to promote their in vitro function in subsequent SCNT. Finally these cells can also result in successful development of cloned embryos to term.
Collapse
Affiliation(s)
- Ying Liu
- Department of Animal Science, Aarhus University, DK-8830 Tjele, Denmark
| | - Olga Ostrup
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, DK-1870 Frederiksberg C, Denmark
| | - Rong Li
- Department of Animal Science, Aarhus University, DK-8830 Tjele, Denmark
| | - Juan Li
- Department of Animal Science, Aarhus University, DK-8830 Tjele, Denmark
| | - Gábor Vajta
- Department of Animal Science, Aarhus University, DK-8830 Tjele, Denmark
| | - Peter M Kragh
- Department of Animal Science, Aarhus University, DK-8830 Tjele, Denmark
| | - Mette Schmidt
- Department of Veterinary Reproduction and Obstetrics, University of Copenhagen, DK-1870 Frederiksberg C, Denmark
| | - Stig Purup
- Department of Animal Science, Aarhus University, DK-8830 Tjele, Denmark
| | - Poul Hyttel
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, DK-1870 Frederiksberg C, Denmark
| | - Dan Klærke
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, DK-1870 Frederiksberg C, Denmark
| | - Henrik Callesen
- Department of Animal Science, Aarhus University, DK-8830 Tjele, Denmark
| |
Collapse
|
32
|
Floyd ZE, Floyd EZ, Staszkiewicz J, Power RA, Kilroy G, Kirk-Ballard H, Barnes CW, Strickler KL, Rim JS, Harkins LL, Gao R, Kim J, Eilertsen KJ. Prolonged proteasome inhibition cyclically upregulates Oct3/4 and Nanog gene expression, but reduces induced pluripotent stem cell colony formation. Cell Reprogram 2015; 17:95-105. [PMID: 25826722 DOI: 10.1089/cell.2014.0030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
There is ample evidence that the ubiquitin-proteasome system is an important regulator of transcription and its activity is necessary for maintaining pluripotency and promoting cellular reprogramming. Moreover, proteasome activity contributes to maintaining the open chromatin structure found in pluripotent stem cells, acting as a transcriptional inhibitor at specific gene loci generally associated with differentiation. The current study was designed to understand further the role of proteasome inhibition in reprogramming and its ability to modulate endogenous expression of pluripotency-related genes and induced pluripotent stem cells (iPSCs) colony formation. Herein, we demonstrate that acute combinatorial treatment with the proteasome inhibitors MG101 or MG132 and the histone deacetylase (HDAC) inhibitor valproic acid (VPA) increases gene expression of the pluripotency marker Oct3/4, and that MG101 alone is as effective as VPA in the induction of Oct3/4 mRNA expression in fibroblasts. Prolonged proteasome inhibition cyclically upregulates gene expression of Oct3/4 and Nanog, but reduces colony formation in the presence of the iPSC induction cocktail. In conclusion, our results demonstrate that the 26S proteasome is an essential modulator in the reprogramming process. Its inhibition enhances expression of pluripotency-related genes; however, efficient colony formation requires proteasome activity. Therefore, discovery of small molecules that increase proteasome activity might lead to more efficient cell reprogramming and generation of pluripotent cells.
Collapse
Affiliation(s)
| | - Elizabeth Z Floyd
- 1 Ubiquitin Lab, Pennington Biomedical Research Center, Louisiana State University System , Baton Rouge, LA, 70803
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
No JG, Choi MK, Kwon DJ, Yoo JG, Yang BC, Park JK, Kim DH. Cell-free extract from porcine induced pluripotent stem cells can affect porcine somatic cell nuclear reprogramming. J Reprod Dev 2015; 61:90-8. [PMID: 25736622 PMCID: PMC4410095 DOI: 10.1262/jrd.2014-078] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Pretreatment of somatic cells with undifferentiated cell extracts, such as embryonic stem cells and mammalian oocytes, is an attractive alternative method for reprogramming control. The properties of induced pluripotent stem cells (iPSCs) are similar to those of embryonic stem cells; however, no studies have reported somatic cell nuclear reprogramming using iPSC extracts. Therefore, this study aimed to evaluate the effects of porcine iPSC extracts treatment on porcine ear fibroblasts and early development of porcine cloned embryos produced from porcine ear skin fibroblasts pretreated with the porcine iPSC extracts. The Chariot(TM) reagent system was used to deliver the iPSC extracts into cultured porcine ear skin fibroblasts. The iPSC extracts-treated cells (iPSC-treated cells) were cultured for 3 days and used for analyzing histone modification and somatic cell nuclear transfer. Compared to the results for nontreated cells, the trimethylation status of histone H3 lysine residue 9 (H3K9) in the iPSC-treated cells significantly decreased. The expression of Jmjd2b, the H3K9 trimethylation-specific demethylase gene, significantly increased in the iPSC-treated cells; conversely, the expression of the proapoptotic genes, Bax and p53, significantly decreased. When the iPSC-treated cells were transferred into enucleated porcine oocytes, no differences were observed in blastocyst development and total cell number in blastocysts compared with the results for control cells. However, H3K9 trimethylation of pronuclear-stage-cloned embryos significantly decreased in the iPSC-treated cells. Additionally, Bax and p53 gene expression in the blastocysts was significantly lower in iPSC-treated cells than in control cells. To our knowledge, this study is the first to show that an extracts of porcine iPSCs can affect histone modification and gene expression in porcine ear skin fibroblasts and cloned embryos.
Collapse
Affiliation(s)
- Jin-Gu No
- Animal Biotechnology Division; Department of Biological Science, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
34
|
López-Iglesias P, Alcaina Y, Tapia N, Sabour D, Arauzo-Bravo MJ, Sainz de la Maza D, Berra E, O'Mara AN, Nistal M, Ortega S, Donovan PJ, Schöler HR, De Miguel MP. Hypoxia induces pluripotency in primordial germ cells by HIF1α stabilization and Oct4 deregulation. Antioxid Redox Signal 2015; 22:205-23. [PMID: 25226357 DOI: 10.1089/ars.2014.5871] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS To study the mechanisms of pluripotency induction, we compared gene expression in pluripotent embryonic germ cells (EGCs) and unipotent primordial germ cells (PGCs). RESULTS We found 11 genes ≥1.5-fold overexpressed in EGCs. None of the genes identified was the Yamanaka genes but instead related to glycolytic metabolism. The prospect of pluripotency induction by cell metabolism manipulation was investigated by hypoxic culturing. Hypoxia induced a glycolytic program in PGCs in detriment of mitochondrial oxidative phosphorylation. We demonstrate that hypoxia alone induces reprogramming in PGCs, giving rise to hypoxia-induced EGC-like cells (hiEGLs), which differentiate into cells of the three germ layers in vitro and contribute to the internal cell mass of the blastocyst in vivo, demonstrating pluripotency. The mechanism of hypoxia induction involves HIF1α stabilization and Oct4 deregulation. However, hiEGL cannot be passaged long term. Self-renewal capacity is not achieved by hypoxia likely due to the lack of upregulation of c-Myc and Klf4. Gene expression analysis of hypoxia signaling suggests that hiEGLs have not reached the stabilization phase of cell reprogramming. INNOVATION AND CONCLUSION Our data suggest that the two main properties of stemness, pluripotency and self-renewal, are differentially regulated in PGC reprogramming induced by hypoxia.
Collapse
Affiliation(s)
- Pilar López-Iglesias
- 1 Cell Engineering Laboratory, IdiPaz, La Paz Hospital Research Institute , Madrid Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Doh MS, Han DMR, Oh DH, Kim SH, Choi MR, Chai YG. Profiling of Proteins Regulated by Venlafaxine during Neural Differentiation of Human Cells. Psychiatry Investig 2015; 12:81-91. [PMID: 25670950 PMCID: PMC4310925 DOI: 10.4306/pi.2015.12.1.81] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 03/07/2014] [Accepted: 03/25/2014] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Antidepressants are known to positively influence several factors in patients with depressive disorders, resulting in increased neurogenesis and subsequent relief of depressive disorders. To study the effects of venlafaxine during neural differentiation at the cellular level, we looked at its effect on protein expression and regulation mechanisms during neural differentiation. METHODS After exposing NCCIT cell-derived EBs to venlafaxine during differentiation (1 day and 7 days), changes in protein expression were analyzed by 2-DE and MALDI-TOF MS analysis. Gene levels of proteins regulated by venlafaxine were analyzed by real-time RT-PCR. RESULTS Treatment with venlafaxine decreased expression of prolyl 4-hydroxylase (P4HB), ubiquitin-conjugating enzyme E2K (HIP2) and plastin 3 (T-plastin), and up-regulated expression of growth factor beta-3 (TGF-β3), dihydropyrimidinase-like 3 (DPYSL3), and pyruvate kinase (PKM) after differentiation for 1 and 7 days. In cells exposed to venlafaxine, the mRNA expression patterns of HIP2 and PKM, which function as negative and positive regulators of differentiation and neuronal survival, respectively, were consistent with the observed changes in protein expression. CONCLUSION Our findings may contribute to improve understanding of molecular mechanism of venlafaxine.
Collapse
Affiliation(s)
- Mi Sook Doh
- Department of Molecular and Life Sciences, Hanyang University, Ansan, Republic of Korea
| | - Dal Mu Ri Han
- Department of Molecular and Life Sciences, Hanyang University, Ansan, Republic of Korea
| | - Dong Hoon Oh
- Department of Neuropsychiatry, College of Medicine and Institute of Mental Health, Hanyang University, Seoul, Republic of Korea
| | - Seok Hyeon Kim
- Department of Neuropsychiatry, College of Medicine and Institute of Mental Health, Hanyang University, Seoul, Republic of Korea
| | - Mi Ran Choi
- Department of Molecular and Life Sciences, Hanyang University, Ansan, Republic of Korea
| | - Young Gyu Chai
- Department of Molecular and Life Sciences, Hanyang University, Ansan, Republic of Korea
| |
Collapse
|
36
|
Slater JA, Zhou S, Puscheck EE, Rappolee DA. Stress-induced enzyme activation primes murine embryonic stem cells to differentiate toward the first extraembryonic lineage. Stem Cells Dev 2014; 23:3049-64. [PMID: 25144240 PMCID: PMC4267551 DOI: 10.1089/scd.2014.0157] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 08/21/2014] [Indexed: 12/11/2022] Open
Abstract
Extracellular stresses influence transcription factor (TF) expression and therefore lineage identity in the peri-implantation mouse embryo and its stem cells. This potentially affects pregnancy outcome. To understand the effects of stress signaling during this critical period of pregnancy, we exposed cultured murine embryonic stem cells (mESCs) to hyperosmotic stress. We then measured stress-enzyme-dependent regulation of key pluripotency and lineage TFs. Hyperosmotic stress slowed mESC accumulation due to slowing of the cell cycle over 72 h, after a small apoptotic response within 12 h. Phosphoinositide 3-kinase (PI3K) enzymatic signaling was responsible for stem cell survival under stressed conditions. Stress initially triggered mESC differentiation after 4 h through MEK1, c-Jun N-terminal kinase (JNK), and PI3K enzymatic signaling, which led to proteasomal degradation of Oct4, Nanog, Sox2, and Rex1 TF proteins. Concurrent with this post-transcriptional effect was the decreased accumulation of potency TF mRNA transcripts. After 12-24 h of stress, cells adapted, cell cycle resumed, and Oct4 and Nanog mRNA and protein expression returned to approximately normal levels. The TF protein recovery was mediated by p38MAPK and PI3K signaling, as well as by MEK2 and/or MEK1. However, due to JNK signaling, Rex1 expression did not recover. Probing for downstream lineages revealed that although mESCs did not differentiate morphologically during 24 h of stress, they were primed to differentiate by upregulating markers of the first lineage differentiating from mESCs, extraembryonic endoderm. Thus, although two to three TFs that mark pluripotency recover expression by 24 h of stress, there is nonetheless sustained Rex1 suppression and a priming of mESCs for differentiation to the earliest lineage.
Collapse
Affiliation(s)
- Jill A. Slater
- Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility, CS Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan
- Program for Reproductive Sciences, Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Sichang Zhou
- Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility, CS Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan
- Program for Reproductive Sciences, Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Elizabeth Ella Puscheck
- Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility, CS Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan
| | - Daniel A. Rappolee
- Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility, CS Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan
- Program for Reproductive Sciences, Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan
- Institutes for Environmental Health Science, Wayne State University School of Medicine, Detroit, Michigan
- Department of Biology, University of Windsor, Windsor, Ontario, Canada
| |
Collapse
|
37
|
Pluripotent state induction in mouse embryonic fibroblast using mRNAs of reprogramming factors. Int J Mol Sci 2014; 15:21840-64. [PMID: 25437916 PMCID: PMC4284681 DOI: 10.3390/ijms151221840] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 10/31/2014] [Accepted: 11/05/2014] [Indexed: 12/12/2022] Open
Abstract
Reprogramming of somatic cells has great potential to provide therapeutic treatments for a number of diseases as well as provide insight into mechanisms underlying early embryonic development. Improvement of induced Pluripotent Stem Cells (iPSCs) generation through mRNA-based methods is currently an area of intense research. This approach provides a number of advantages over previously used methods such as DNA integration and insertional mutagenesis. Using transfection of specifically synthesized mRNAs of various pluripotency factors, we generated iPSCs from mouse embryonic fibroblast (MEF) cells. The genetic, epigenetic and functional properties of the iPSCs were evaluated at different times during the reprogramming process. We successfully introduced synthesized mRNAs, which localized correctly inside the cells and exhibited efficient and stable translation into proteins. Our work demonstrated a robust up-regulation and a gradual promoter de-methylation of the pluripotency markers, including non-transfected factors such as Nanog, SSEA-1 (stage-specific embryonic antigen 1) and Rex-1 (ZFP-42, zinc finger protein 42). Using embryonic stem cells (ESCs) conditions to culture the iPS cells resulted in formation of ES-like colonies after approximately 12 days with only five daily repeated transfections. The colonies were positive for alkaline phosphatase and pluripotency-specific markers associated with ESCs. This study revealed the ability of pluripotency induction and generation of mouse mRNA induced pluripotent stem cells (mRNA iPSCs) using transfection of specifically synthesized mRNAs of various pluripotency factors into mouse embryonic fibroblast (MEF) cells. These generated iPSCs exhibited molecular and functional properties similar to ESCs, which indicate that this method is an efficient and viable alternative to ESCs and can be used for further biological, developmental and therapeutic investigations.
Collapse
|
38
|
Skowron K, Tomsia M, Czekaj P. An experimental approach to the generation of human embryonic stem cells equivalents. Mol Biotechnol 2014; 56:12-37. [PMID: 24146427 DOI: 10.1007/s12033-013-9702-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recently, particular attention has been paid to the human embryonic stem cells (hESC) in the context of their potential application in regenerative medicine; however, ethical concerns prevent their clinical application. Induction of pluripotency in somatic cells seems to be a good alternative for hESC recruitment regarding its potential use in tissue regeneration, disease modeling, and drug screening. Since Yamanaka's team in 2006 restored pluripotent state of somatic cells for the first time, a significant progress has been made in the area of induced pluripotent stem cells (iPSC) generation. Here, we review the current state of knowledge in the issue of techniques applied to establish iPSC. Somatic cell nuclear transfer, cell fusion, cell extracts reprogramming, and techniques of direct reprogramming are described. Retroviral and lentiviral transduction are depicted as ways of cell reprogramming with the use of integrating vectors. Contrary to them, adenoviruses, plasmids, single multiprotein expression vectors, and PiggyBac transposition systems are examples of non-integrative vectors used in iPSC generation protocols. Furthermore, reprogramming with the delivery of specific proteins, miRNA or small chemical compounds are presented. Finally, the changes occurring during the reprogramming process are described. It is concluded that subject to some limitations iPSC could become equivalents for hESC in regenerative medicine.
Collapse
Affiliation(s)
- Katarzyna Skowron
- Students Scientific Society, Medical University of Silesia, Katowice, Poland
| | | | | |
Collapse
|
39
|
Kim SY, Kim TS, Park SH, Lee MR, Eun HJ, Baek SK, Ko YG, Kim SW, Seong HH, Campbell KHS, Lee JH. Siberian Sturgeon Oocyte Extract Induces Epigenetic Modifications of Porcine Somatic Cells and Improves Developmental Competence of SCNT Embryos. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2014; 27:266-77. [PMID: 25049951 PMCID: PMC4093206 DOI: 10.5713/ajas.2013.13699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 11/29/2013] [Accepted: 11/25/2013] [Indexed: 11/27/2022]
Abstract
Somatic cell nuclear transfer (SCNT) has generally demonstrated that a differentiated cell can convert into a undifferentiated or pluripotent state. In the SCNT experiment, nuclear reprogramming is induced by exposure of introduced donor nuclei to the recipient cytoplasm of matured oocytes. However, because the efficiency of SCNT still remains low, a combination of SCNT technique with the ex-ovo method may improve the normal development of SCNT embryos. Here we hypothesized that treatment of somatic cells with extracts prepared from the germinal vesicle (GV) stage Siberian sturgeon oocytes prior to their use as nuclear donor for SCNT would improve in vitro development. A reversible permeability protocol with 4 μg/mL of digitonin for 2 min at 4°C in order to deliver Siberian sturgeon oocyte extract (SOE) to porcine fetal fibroblasts (PFFs) was carried out. As results, the intensity of H3K9ac staining in PFFs following treatment of SOE for 7 h at 18°C was significantly increased but the intensity of H3K9me3 staining in PFFs was significantly decreased as compared with the control (p<0.05). Additionally, the level of histone acetylation in SCNT embryos at the zygote stage was significantly increased when reconstructed using SOE-treated cells (p<0.05), similar to that of IVF embryos at the zygote stage. The number of apoptotic cells was significantly decreased and pluripotency markers (Nanog, Oct4 and Sox2) were highly expressed in the blastocyst stage of SCNT embryos reconstructed using SOE-treated cells as nuclear donor (p<0.05). And there was observed a better development to the blastocyst stage in the SOE-treated group (p<0.05). Our results suggested that pre-treatment of cells with SOE could improve epigenetic reprogramming and the quality of porcine SCNT embryos.
Collapse
Affiliation(s)
- So-Young Kim
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 660-701, Korea
| | - Tae-Suk Kim
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 660-701, Korea
| | - Sang-Hoon Park
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 660-701, Korea
| | - Mi-Ran Lee
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 660-701, Korea
| | - Hye-Ju Eun
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 660-701, Korea
| | - Sang-Ki Baek
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 660-701, Korea
| | - Yeoung-Gyu Ko
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 660-701, Korea
| | - Sung-Woo Kim
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 660-701, Korea
| | - Hwan-Hoo Seong
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 660-701, Korea
| | - Keith H S Campbell
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 660-701, Korea
| | - Joon-Hee Lee
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 660-701, Korea
| |
Collapse
|
40
|
Ramesh T, Lee SH, Lee CS, Kwon YW, Cho HJ. Somatic cell dedifferentiation/reprogramming for regenerative medicine. Int J Stem Cells 2014; 2:18-27. [PMID: 24855516 DOI: 10.15283/ijsc.2009.2.1.18] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2009] [Indexed: 12/14/2022] Open
Abstract
The concept of dedifferentiation or reprogramming of a somatic cell into a pluripotent embryonic stem cell-like cell (ES-like cell), which give rise to three germ layers and differentiate various cell types, opens a new era in stem cell biology and provides potential therapeutic modality in regenerative medicine. Here, we outline current dedifferentiation/reprogramming methods and their technical hurdles, and the safety and therapeutic applications of reprogrammed pluripotent stem cells in regenerative medicine. This review summarizes the concept and data of somatic cell nuclear transfer, fusion of somatic cells with ES cells, viral or non-viral transduction of pluripotency-related genes into somatic cells, introduction of extract (or proteins) of pluripotent cells into somatic cells. Dedifferentiated/reprogrammed ES-like cells could be a perfect genetic match (autologous or tailored pluripotent stem cells) for future applications. Further studies regarding technical refinements as well as mechanistic analysis of dedifferentiation induction and re-differentiation into specific cell types will provide us with the substantial application of pluripotent stem cells to therapeutic purposes.
Collapse
Affiliation(s)
- Thiyagarajan Ramesh
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea ; National Research Laboratory for Cardiovascular Stem Cells, Seoul National University College of Medicine, Seoul, Korea
| | - Sun-Hee Lee
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea ; National Research Laboratory for Cardiovascular Stem Cells, Seoul National University College of Medicine, Seoul, Korea
| | - Choon-Soo Lee
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea ; National Research Laboratory for Cardiovascular Stem Cells, Seoul National University College of Medicine, Seoul, Korea
| | - Yoo-Wook Kwon
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea ; National Research Laboratory for Cardiovascular Stem Cells, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun-Jai Cho
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea ; National Research Laboratory for Cardiovascular Stem Cells, Seoul National University College of Medicine, Seoul, Korea ; Cardiovascular Center, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
41
|
|
42
|
Hu K. All roads lead to induced pluripotent stem cells: the technologies of iPSC generation. Stem Cells Dev 2014; 23:1285-300. [PMID: 24524728 PMCID: PMC4046204 DOI: 10.1089/scd.2013.0620] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 02/12/2014] [Indexed: 12/26/2022] Open
Abstract
Generation of induced pluripotent stem cells (iPSCs) via the ectopic expression of reprogramming factors is a simple, advanced, yet often perplexing technology due to low efficiency, slow kinetics, and the use of numerous distinct systems for factor delivery. Scientists have used almost all available approaches for the delivery of reprogramming factors. Even the well-established retroviral vectors confuse some scientists due to different tropisms in use. The canonical virus-based reprogramming poses many problems, including insertional mutagenesis, residual expression and re-activation of reprogramming factors, uncontrolled silencing of transgenes, apoptosis, cell senescence, and strong immunogenicity. To eliminate or alleviate these problems, scientists have tried various other approaches for factor delivery and transgene removal. These include transient transfection, nonintegrating viral vectors, Cre-loxP excision of transgenes, excisable transposon, protein transduction, RNA transfection, microRNA transfection, RNA virion, RNA replicon, nonintegrating replicating episomal plasmids, minicircles, polycistron, and preintegration of inducible reprogramming factors. These alternative approaches have their own limitations. Even iPSCs generated with RNA approaches should be screened for possible transgene insertions mediated by active endogenous retroviruses in the human genome. Even experienced researchers may encounter difficulty in selecting and using these different technologies. This survey presents overviews of iPSC technologies with the intention to provide a quick yet comprehensive reference for both new and experienced reprogrammers.
Collapse
Affiliation(s)
- Kejin Hu
- Department of Biochemistry and Molecular Genetics, UAB Stem Cell Insitute, School of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
43
|
Zhang W, Yang W, Liu X, Zhang L, Huang W, Zhang Y. Rapidly constructed scaffold-free embryonic stem cell sheets for ocular surface reconstruction. SCANNING 2014; 36:286-92. [PMID: 23861021 DOI: 10.1002/sca.21103] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Revised: 04/25/2013] [Accepted: 05/09/2013] [Indexed: 05/13/2023]
Abstract
This study is an extension of our previously published work demonstrating the generation of corneal epithelial sheets using optimal centrifugation procedure. In this study, we investigated the therapeutic potential of embryonic stem cells (ESCs) for ocular surface reconstruction in rabbits with limbal stem cell deficiency (LSCD) using the centrifugation method. Rabbits with LSCD were assigned to two groups: 30 LSCD rabbits were treated with scaffold-free embryonic stem cells sheets (SESCS) and six LSCD rabbits received no treatment (control group). The two groups were followed up for 15 days using slit lamp observation. Cytokeratin K3, mucin 5AC, and OCT-4 were used to evaluate the re-epithelialization of the cornea. Fluorescent DiIC18(3)-DS dye was used to trace the transplanted cells. Atomic force microscopy (AFM) was used to investigate the cultured epithelial cells of the SESCS-treated group. The SESCS transplant facilitated the reconstruction of the ocular surface in 75% of the treated animals. Conjunctivalization and neovascularization were observed in the control group. The SESCS group was K3 positive and MUC5AC negative, and OCT-4 was observed on the re-epithelialized corneal epithelium. Labeled SESCS cells were detected in vivo at 15 days post-transplant. Using AFM, three different types of cultured cells were identified in the rabbit corneal epithelium of the SESCS treatment group. The SESCS were demonstrated to reconstruct the ocular surface in rabbits with LSCD. ES cells differentiated into corneal epithelial cells when in direct contact with the stroma and thus can serve as a cell source in corneal tissue engineering.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Ophthalmology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, P.R. China
| | | | | | | | | | | |
Collapse
|
44
|
Xiong XR, Lan DL, Li J, Zi XD, Ma L, Wang Y. Cellular extract facilitates nuclear reprogramming by altering DNA methylation and pluripotency gene expression. Cell Reprogram 2014; 16:215-22. [PMID: 24738992 DOI: 10.1089/cell.2013.0078] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The functional reprogramming of a differentiated cell to a pluripotent state presents potential beneficial applications in disease mechanisms and regenerative medicine. Epigenetic modifications enable differentiated cells to perpetuate molecular memory to retain their identity. Therefore, the aim of this study was to investigate the reprogramming modification of yak fibroblast cells that were permeabilized and incubated in the extracts of mesenchymal stem cells derived from mice adipose tissue [adipose-derived stem cells (ADSCs)]. According to the results, the treatment of ADSC extracts promoted colony formation. Moreover, pluripotent gene expression was associated with the loss of repressive histone modifications and increased global demethylation. The genes Col1a1 and Col1a2, which are typically found in differentiated cells only, demonstrated decreased expression and increased methylation in the 5'-flanking regulatory regions. Moreover, yak fibroblast cells that were exposed to ADSC extracts resulted in significantly different eight-cell and blastocyst formation rates of cloned embryos compared with their untreated counterparts. This investigation provides the first evidence that nuclear reprogramming of yak fibroblast cells is modified after the ADSC extract treatment. This research also presents a methodology for studying the dedifferentiation of somatic cells that can potentially lead to an efficient way of reprogramming somatic cells toward a pluripotent state without genetic alteration.
Collapse
Affiliation(s)
- Xian-Rong Xiong
- 1 College of Life Science and Technology, Southwest University for Nationalities , Chengdu, Sichuan, 610041, China
| | | | | | | | | | | |
Collapse
|
45
|
Mahapatra PS, Bag S. Reprogramming of buffalo (Bubalus bubalis) foetal fibroblasts with avian egg extract for generation of pluripotent stem cells. Res Vet Sci 2014; 96:292-8. [DOI: 10.1016/j.rvsc.2014.02.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 01/06/2014] [Accepted: 02/09/2014] [Indexed: 01/12/2023]
|
46
|
Talaei-Khozani T, Heidari F, Esmaeilpour T, Vojdani Z, Mostafavi-Pour Z, Rohani L. Cardiomyocyte marker expression in mouse embryonic fibroblasts by cell-free cardiomyocyte extract and epigenetic manipulation. IRANIAN JOURNAL OF MEDICAL SCIENCES 2014; 39:203-12. [PMID: 24753644 PMCID: PMC3993041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 01/22/2013] [Accepted: 04/07/2013] [Indexed: 10/28/2022]
Abstract
BACKGROUND The regenerative capacity of the mammalian heart is quite limited. Recent reports have focused on reprogramming mesenchymal stem cells into cardiomyocytes. We investigated whether fibroblasts could transdifferentiate into myocardium. METHODS Mouse embryonic fibroblasts were treated with Trichostatin A (TSA) and 5-Aza-2-Deoxycytidine (5-aza-dC). The treated cells were permeabilized with streptolysin O and exposed to the mouse cardiomyocyte extract and cultured for 1, 10, and 21 days. Cardiomyocyte markers were detected by immunohistochemistry. Alkaline phosphatase activity and OCT4 were also detected in cells treated by chromatin-modifying agents. RESULTS The cells exposed to a combination of 5-aza-dC and TSA and permeabilized in the presence of the cardiomyocyte extract showed morphological changes. The cells were unable to express cardiomyocyte markers after 24 h. Immunocytochemical assays showed a notable degree of myosin heavy chain and α-actinin expressions after 10 days. The expression of the natriuretic factor and troponin T occurred after 21 days in these cells. The cells exposed to chromatin-modifying agents also expressed cardiomyocyte markers; however, the proportion of reprogrammed cells was clearly smaller than that in the cultures exposed to 5-aza-dC , TSA, and extract. CONCLUSION It seems that the fibroblasts were able to eliminate the previous epigenetic markers and form new ones according to the factors existing in the extract. Since no beating was observed, at least up to 21 days, the cells may need an appropriate extracellular matrix for their function.
Collapse
Affiliation(s)
- Tahereh Talaei-Khozani
- Laboratory for Stem Cell Research, Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran;
,Department of Tissue Engineering, School of Advance Sciences and Technology, Shiraz University of Medical Sciences, Shiraz, Iran;
| | - Fatemeh Heidari
- Laboratory for Stem Cell Research, Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran;
| | - Tahereh Esmaeilpour
- Laboratory for Stem Cell Research, Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran;
| | - Zahra Vojdani
- Laboratory for Stem Cell Research, Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran;
| | - Zohrah Mostafavi-Pour
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Leili Rohani
- Laboratory for Stem Cell Research, Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran;
| |
Collapse
|
47
|
Kole D, Ambady S, Page RL, Dominko T. Maintenance of multipotency in human dermal fibroblasts treated with Xenopus laevis egg extract requires exogenous fibroblast growth factor-2. Cell Reprogram 2014; 16:18-28. [PMID: 24405062 PMCID: PMC3920749 DOI: 10.1089/cell.2013.0066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Direct reprogramming of a differentiated somatic cell into a developmentally more plastic cell would offer an alternative to applications in regenerative medicine that currently depend on either embryonic stem cells (ESCs), adult stem cells, or induced pluripotent stem cells (iPSCs). Here we report the potential of select Xenopus laevis egg extract fractions, in combination with exogenous fibroblast growth factor-2 (FGF2), to affect life span, morphology, gene expression, protein translation, and cellular localization of OCT4 and NANOG transcription factors, and the developmental potential of human dermal fibroblasts in vitro. A gradual change in morphology is accompanied by translation of embryonic transcription factors and their nuclear localization and a life span exceeding 60 population doublings. Cells acquire the ability to follow adipogenic, neuronal, and osteogenic differentiation under appropriate induction conditions in vitro. Analysis of active extract fractions reveals that Xenopus egg protein and RNAs as well as exogenously supplemented FGF2 are required and sufficient for induction and maintenance of this phenotypic change. Factors so far identified in the active fractions include FGF2 itself, transforming growth factor-β, maskin, and nucleoplasmin. Identification of critical factors needed for reprogramming may allow for nonviral, chemically defined derivation of human-induced multipotent cells that can be maintained by exogenous FGF2.
Collapse
Affiliation(s)
- Denis Kole
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA, 01609
| | - Sakthikumar Ambady
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609
| | - Raymond L. Page
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609
- Bioengineering Institute, Worcester Polytechnic Institute, Worcester, MA, 01609
- CellThera, Inc., Worcester, MA, 01605
| | - Tanja Dominko
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA, 01609
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609
- Bioengineering Institute, Worcester Polytechnic Institute, Worcester, MA, 01609
- CellThera, Inc., Worcester, MA, 01605
| |
Collapse
|
48
|
Kwon YW, Chung YJ, Kim J, Lee HJ, Park J, Roh TY, Cho HJ, Yoon CH, Koo BK, Kim HS. Comparative study of efficacy of dopaminergic neuron differentiation between embryonic stem cell and protein-based induced pluripotent stem cell. PLoS One 2014; 9:e85736. [PMID: 24465672 PMCID: PMC3899054 DOI: 10.1371/journal.pone.0085736] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 12/01/2013] [Indexed: 11/29/2022] Open
Abstract
In patients with Parkinson's disease (PD), stem cells can serve as therapeutic agents to restore or regenerate injured nervous system. Here, we differentiated two types of stem cells; mouse embryonic stem cells (mESCs) and protein-based iPS cells (P-iPSCs) generated by non-viral methods, into midbrain dopaminergic (mDA) neurons, and then compared the efficiency of DA neuron differentiation from these two cell types. In the undifferentiated stage, P-iPSCs expressed pluripotency markers as ES cells did, indicating that protein-based reprogramming was stable and authentic. While both stem cell types were differentiated to the terminally-matured mDA neurons, P-iPSCs showed higher DA neuron-specific markers' expression than ES cells. To investigate the mechanism of the superior induction capacity of DA neurons observed in P-iPSCs compared to ES cells, we analyzed histone modifications by genome-wide ChIP sequencing analysis and their corresponding microarray results between two cell types. We found that Wnt signaling was up-regulated, while SFRP1, a counter-acting molecule of Wnt, was more suppressed in P-iPSCs than in mESCs. In PD rat model, transplantation of neural precursor cells derived from both cell types showed improved function. The present study demonstrates that P-iPSCs could be a suitable cell source to provide patient-specific therapy for PD without ethical problems or rejection issues.
Collapse
Affiliation(s)
- Yoo-Wook Kwon
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Yeon-Ju Chung
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Joonoh Kim
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Ho-Jae Lee
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Jihwan Park
- Division of Molecular and Life Sciences,Pohang University of Science and Technology, Pohang, Korea
| | - Tae-Young Roh
- Division of Molecular and Life Sciences,Pohang University of Science and Technology, Pohang, Korea
| | - Hyun-Jai Cho
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University, Seoul, Korea
| | - Chang-Hwan Yoon
- Cardiovascular center, Seoul National University Bundang Hospital, Seoul National University, Seoul, Korea
| | - Bon-Kwon Koo
- Department of Internal Medicine, Seoul National University, Seoul, Korea
| | - Hyo-Soo Kim
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University, Seoul, Korea
- Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul, Korea
- * E-mail:
| |
Collapse
|
49
|
Stem cell reprogramming: generation of patient-specific stem cells by somatic cell nuclear reprogramming. DRUG DISCOVERY TODAY. TECHNOLOGIES 2013; 5:e105-48. [PMID: 24125543 DOI: 10.1016/j.ddtec.2008.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
50
|
Lønne GK, Gammelsaeter R, Haglerød C. Composition characterization and clinical efficacy study of a salmon egg extract. Int J Cosmet Sci 2013; 35:515-22. [PMID: 23738607 DOI: 10.1111/ics.12071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 06/01/2013] [Indexed: 04/30/2025]
Abstract
OBJECTIVE There is an increasing demand for scientifically documented over-the-counter products on the cosmetic market. Salmon eggs are rich in proteins, vitamins and minerals with anti-oxidative and anti-inflammatory properties, as well as free amino acids and lipids documented to be beneficial for skin. Of the fatty acids, several are commonly used as skin penetration enhancers. The unique combination of active substances led us to study whether an extract from salmon eggs could serve as an ingredient for skin care. METHODS We conducted a double-blinded, randomized clinical trial with 66 healthy female volunteers. Efficacy of the salmon egg extract was evaluated at concentrations of 1% and 5% in vehicle formulation, and responses after 7, 14, 28 and 56 days of treatment were compared with baseline. Composition of the extract was analysed to improve the understanding of the effects of the extract on skin. The salmon egg extract was safety-tested by repeat insult patch test. RESULTS Treatment of facial skin with the salmon egg extract significantly improved all parameters investigated, wrinkles, pigmentation, redness, brightness and hydration and led to global improvement of the facial skin. Efficacy of the extract was dose dependent and time dependent. There were no adverse reactions noted during the course of the repeat insult patch test, demonstrating that the extract causes neither skin irritation nor sensitization. Furthermore, chemical analyses of the extract revealed the composition of a vast number of active substances, including unsaturated fatty acids, vitamins, proteins, minerals, DNA and RNA. CONCLUSION The salmon egg extract serves as a skin care ingredient that significantly improves characteristics important for perception of skin ageing and health. The efficacy of the treatment is conceivably accounted for by the unique combination of numerous active substances present in the salmon egg extract.
Collapse
Affiliation(s)
- G K Lønne
- Regenics AS, PO Box 4536, Nydalen, 0404, Oslo, Norway
| | | | | |
Collapse
|