1
|
Fujihara K, Yoneda T, Sugidono A, Okada Y, Hiyama S, Kajikawa S, Fukunaga Y, Koch M, Izu Y. Collagen XII deficiency promotes ligament-specific heterotopic ossification via fibrochondrocyte differentiation. Biochem Biophys Res Commun 2025; 757:151621. [PMID: 40088675 DOI: 10.1016/j.bbrc.2025.151621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
Heterotopic ossification of tendons and ligaments causes pain and dysfunction, significantly reducing quality of life. However, its underlying mechanisms remain elusive. In addition to injury, tissue organization and stiffness have been implicated in heterotopic ossification. Collagen XII, a member of the fibril-associated collagens with interrupted triple helices (FACIT) family, plays a crucial role in maintaining the structural integrity and function of tendons and ligaments. Its deficiency alters tissue stiffness and predisposes ligaments to rupture. In this study, we investigated whether collagen XII contributes to the development of heterotopic ossification. Three-dimensional microcomputed tomography (3D-μCT) and X-ray analyses revealed heterotopic bone formation in the knee and ankle ligaments, but not in tendons, of Col12a1-deficient mice, with a 100 % incidence in mice older than 19 weeks. Histological analysis showed the presence of Alcian blue- and Toluidine blue-positive fibrochondrocyte-like cells in Col12a1-deficient ligaments, which were subsequently replaced by bone tissue, as indicated by Alizarin red staining. Real-time qPCR analysis of knee ligaments demonstrated a slight increase in chondrogenic markers and a significant upregulation of osteogenic markers in Col12a1-deficient mice compared with wild-type controls. In vitro chondrogenesis and osteogenesis assays using primary tenocytes from wild-type and Col12a1-deficient mice revealed that collagen XII deficiency enhanced osteogenic potential, whereas chondrogenic potential remained comparable. Our findings indicate that collagen XII deficiency specifically induces heterotopic bone formation in knee and ankle ligaments, occurring via fibrochondrocytes rather than through endochondral or intramembranous ossification.
Collapse
Affiliation(s)
- Kei Fujihara
- Graduate School of Veterinary Medicine, Nippon Veterinary and Life Science University, 1-7-1 Kyounan, Musashino, Tokyo, 180-8602, Japan; Department of Comparative Cell Biology, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, 1-7-1 Kyounan, Musashino, Tokyo, 180-8602, Japan; Department of Laboratory Animal Science, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoinooka, Imabari, Ehime, 794-8555, Japan
| | - Taiju Yoneda
- Department of Laboratory Animal Science, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoinooka, Imabari, Ehime, 794-8555, Japan
| | - Akira Sugidono
- Department of Laboratory Animal Science, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoinooka, Imabari, Ehime, 794-8555, Japan
| | - Yukina Okada
- Department of Animal Risk Management, Faculty of Risk and Crisis Management, Chiba Institute of Science, 15-8 Shiomi, Choshi, Chiba, 288-0025, Japan
| | - Sakura Hiyama
- Department of Animal Risk Management, Faculty of Risk and Crisis Management, Chiba Institute of Science, 15-8 Shiomi, Choshi, Chiba, 288-0025, Japan
| | - Shuhei Kajikawa
- Department of Laboratory Animal Science, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoinooka, Imabari, Ehime, 794-8555, Japan
| | - Yuko Fukunaga
- Department of Animal Risk Management, Faculty of Risk and Crisis Management, Chiba Institute of Science, 15-8 Shiomi, Choshi, Chiba, 288-0025, Japan
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931, Cologne, Germany
| | - Yayoi Izu
- Graduate School of Veterinary Medicine, Nippon Veterinary and Life Science University, 1-7-1 Kyounan, Musashino, Tokyo, 180-8602, Japan; Department of Comparative Cell Biology, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, 1-7-1 Kyounan, Musashino, Tokyo, 180-8602, Japan; Department of Laboratory Animal Science, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoinooka, Imabari, Ehime, 794-8555, Japan.
| |
Collapse
|
2
|
Kidwai FK, Canalis E, Robey PG. Induced pluripotent stem cell technology in bone biology. Bone 2023; 172:116760. [PMID: 37028583 PMCID: PMC10228209 DOI: 10.1016/j.bone.2023.116760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/09/2023]
Abstract
Technologies on the development and differentiation of human induced pluripotent stem cells (hiPSCs) are rapidly improving, and have been applied to create cell types relevant to the bone field. Differentiation protocols to form bona fide bone-forming cells from iPSCs are available, and can be used to probe details of differentiation and function in depth. When applied to iPSCs bearing disease-causing mutations, the pathogenetic mechanisms of diseases of the skeleton can be elucidated, along with the development of novel therapeutics. These cells can also be used for development of cell therapies for cell and tissue replacement.
Collapse
Affiliation(s)
- Fahad K Kidwai
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, United States of America
| | - Ernesto Canalis
- Center for Skeletal Research, Orthopedic Surgery and Medicine, UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030-4037, United States of America
| | - Pamela G Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, United States of America.
| |
Collapse
|
3
|
Bradykinin, as a Reprogramming Factor, Induces Transdifferentiation of Brain Astrocytes into Neuron-like Cells. Biomedicines 2021; 9:biomedicines9080923. [PMID: 34440126 PMCID: PMC8389672 DOI: 10.3390/biomedicines9080923] [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: 06/03/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Kinins are endogenous, biologically active peptides released into the plasma and tissues via the kallikrein-kinin system in several pathophysiological events. Among kinins, bradykinin (BK) is widely distributed in the periphery and brain. Several studies on the neuro-modulatory actions of BK by the B2BK receptor (B2BKR) indicate that this neuropeptide also functions during neural fate determination. Previously, BK has been shown to induce differentiation of nerve-related stem cells into neuron cells, but the response in mature brain astrocytes is unknown. Herein, we used rat brain astrocyte (RBA) to investigate the effect of BK on cell transdifferentiation into a neuron-like cell morphology. Moreover, the signaling mechanisms were explored by zymographic, RT-PCR, Western blot, and immunofluorescence staining analyses. We first observed that BK induced RBA transdifferentiation into neuron-like cells. Subsequently, we demonstrated that BK-induced RBA transdifferentiation is mediated through B2BKR, PKC-δ, ERK1/2, and MMP-9. Finally, we found that BK downregulated the astrocytic marker glial fibrillary acidic protein (GFAP) and upregulated the neuronal marker neuron-specific enolase (NSE) via the B2BKR/PKC-δ/ERK pathway in the event. Therefore, BK may be a reprogramming factor promoting brain astrocytic transdifferentiation into a neuron-like cell, including downregulation of GFAP and upregulation of NSE and MMP-9 via the B2BKR/PKC-δ/ERK cascade. Here, we also confirmed the transdifferentiative event by observing the upregulated neuronal nuclear protein (NeuN). However, the electrophysiological properties of the cells after BK treatment should be investigated in the future to confirm their phenotype.
Collapse
|
4
|
Yin L, Hu P, Shi X, Qian W, Zhau HE, Pandol SJ, Lewis MS, Chung LWK, Wang R. Cancer cell's neuroendocrine feature can be acquired through cell-cell fusion during cancer-neural stem cell interaction. Sci Rep 2020; 10:1216. [PMID: 31988304 PMCID: PMC6985266 DOI: 10.1038/s41598-020-58118-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/10/2020] [Indexed: 02/04/2023] Open
Abstract
Advanced and therapy-resistant prostate tumors often display neural or neuroendocrine behavior. We assessed the consequences of prostate cancer cell interaction with neural cells, which are rich in the human prostate and resident of the prostate tumor. In 3-dimensional co-culture with neurospheres, red fluorescent human LNCaP cells formed agglomerates on the neurosphere surface. Upon induced neural differentiation, some red fluorescent cells showed morphology of fully differentiated neural cells, indicating fusion between the cancer and neural stem cells. These fusion hybrids survived for extended times in a quiescent state. A few eventually restarted cell division and propagated to form derivative hybrid progenies. Clones of the hybrid progenies were highly heterogeneous; most had lost prostatic and epithelial markers while some had acquired neural marker expression. These results indicate that cancer cells can fuse with bystander neural cells in the tumor microenvironment; and cancer cell fusion is a direct route to tumor cell heterogeneity.
Collapse
Affiliation(s)
- Liyuan Yin
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Peizhen Hu
- Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xianping Shi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Weiping Qian
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Haiyen E Zhau
- Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephen J Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael S Lewis
- Department of Pathology, Greater Los Angeles Veterans Affairs Health System, Los Angeles, CA, USA
| | - Leland W K Chung
- Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ruoxiang Wang
- Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Pathology, Greater Los Angeles Veterans Affairs Health System, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Radhakrishnan S, Trentz OA, Reddy MS, Rela M, Kandasamy M, Sellathamby S. In vitro transdifferentiation of human adipose tissue-derived stem cells to neural lineage cells - a stage-specific incidence. Adipocyte 2019; 8:164-177. [PMID: 31033391 PMCID: PMC6768268 DOI: 10.1080/21623945.2019.1607424] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/26/2019] [Accepted: 03/31/2019] [Indexed: 01/31/2023] Open
Abstract
The present Study investigated the intrinsic ability of adipose tissue-derived stem cells (ADSCs) and their neural transdifferentiation in a stage-specific manner. Woodbury's Chemical induction was implemented with modifications to achieve neural transdifferentiation. In Group I, ADSCs were preinduced with β-mercaptoethanol (β-ME) and later, with neural induction medium (NIM). In Group II, ADSCs were directly treated with NIM. In Group III, a DNA methyltransferase (DNMT) inhibitor 5-azacytidine was applied to understand whether transdifferentiation is controlled by epigenetic marks. Irrespective of the presence of (β-ME), the differentiation protocol resulted in glial-lineage cells. Group III produced poorly -differentiated neural cells with neuron-specific enolase positivity. A neuroprogenitor stage (NPC) was identified at d 11 after induction only in Group I. In other groups, this stage was not morphologically distinct. We explored the stage-specific incidence NPC, by alternatively treating them with basic fibroblast growth factor (bFGF), and antioxidants to validate if different signalling could cause varied outcomes (Group IV). They differentiated into neurons, as defined by cell polarity and expression of specific proteins. Meanwhile, neuroprogenitors exposed to NIM (Group I) produced glial-lineage cells. Further refinement and study of the occurrence and terminal differentiation of neuroprogenitors would identify a promising source for neural tissue replacement.
Collapse
Affiliation(s)
- Subathra Radhakrishnan
- National Foundation for Liver Research (NFLR), Gleneagles Global Health City, Chennai, India
- Department of Biomedical Science, Bharathidasan University, Tiruchirappalli, India
| | - Omana Anna Trentz
- MIOT Institute of Research (MIR), MIOT International, Chennai, India
| | - Mettu Srinivas Reddy
- National Foundation for Liver Research (NFLR), Gleneagles Global Health City, Chennai, India
- Institute of Liver Disease and Transplantation, Gleneagles Global Health City, Chennai, India
| | - Mohamed Rela
- National Foundation for Liver Research (NFLR), Gleneagles Global Health City, Chennai, India
- Institute of Liver Disease and Transplantation, Gleneagles Global Health City, Chennai, India
| | - Mahesh Kandasamy
- Department of Animal Science, Bharathidasan University, Tiruchirappalli, India
| | | |
Collapse
|
6
|
Bianchi MV, Awaja F, Altankov G. Dynamic adhesive environment alters the differentiation potential of young and ageing mesenchymal stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 78:467-474. [DOI: 10.1016/j.msec.2017.04.110] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 11/27/2022]
|
7
|
Huda F, Fan Y, Suzuki M, Konno A, Matsuzaki Y, Takahashi N, Chan JKY, Hirai H. Fusion of Human Fetal Mesenchymal Stem Cells with "Degenerating" Cerebellar Neurons in Spinocerebellar Ataxia Type 1 Model Mice. PLoS One 2016; 11:e0164202. [PMID: 27802273 PMCID: PMC5089746 DOI: 10.1371/journal.pone.0164202] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 09/21/2016] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) migrate to damaged tissues, where they participate in tissue repair. Human fetal MSCs (hfMSCs), compared with adult MSCs, have higher proliferation rates, a greater differentiation capacity and longer telomeres with reduced senescence. Therefore, transplantation of quality controlled hfMSCs is a promising therapeutic intervention. Previous studies have shown that intravenous or intracortical injections of MSCs result in the emergence of binucleated cerebellar Purkinje cells (PCs) containing an MSC-derived marker protein in mice, thus suggesting a fusion event. However, transdifferentiation of MSCs into PCs or transfer of a marker protein from an MSC to a PC cannot be ruled out. In this study, we unequivocally demonstrated the fusion of hfMSCs with murine PCs through a tetracycline-regulated (Tet-off) system with or without a Cre-dependent genetic inversion switch (flip-excision; FLEx). In the FLEx-Tet system, we performed intra-cerebellar injection of viral vectors expressing tetracycline transactivator (tTA) and Cre recombinase into either non-symptomatic (4-week-old) or clearly symptomatic (6–8-month-old) spinocerebellar ataxia type 1 (SCA1) mice. Then, the mice received an injection of 50,000 genetically engineered hfMSCs that expressed GFP only in the presence of Cre recombinase and tTA. We observed a significant emergence of GFP-expressing PCs and interneurons in symptomatic, but not non-symptomatic, SCA1 mice 2 weeks after the MSC injection. These results, together with the results obtained using age-matched wild-type mice, led us to conclude that hfMSCs have the potential to preferentially fuse with degenerating PCs and interneurons but not with healthy neurons.
Collapse
Affiliation(s)
- Fathul Huda
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma 371–8511, Japan
- Physiology Division, Department of Anatomy Physiology and Cell Biology, Faculty of Medicine Universitas Padjadjaran, Bandung, 40161, Indonesia
| | - Yiping Fan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, 229899, Singapore
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
| | - Mamiko Suzuki
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma 371–8511, Japan
| | - Ayumu Konno
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma 371–8511, Japan
| | - Yasunori Matsuzaki
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma 371–8511, Japan
| | - Nobutaka Takahashi
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma 371–8511, Japan
| | - Jerry K. Y. Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, 229899, Singapore
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, 169857, Singapore
| | - Hirokazu Hirai
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma 371–8511, Japan
- * E-mail:
| |
Collapse
|
8
|
Andersen DC, Jensen CH, Skovrind I, Johnsen RH, Traustadottir GA, Aagaard KS, Ganesalingam S, Sheikh SP. Neonatal epicardial-derived progenitors aquire myogenic traits in skeletal muscle, but not cardiac muscle. Int J Cardiol 2016; 222:448-456. [PMID: 27505332 DOI: 10.1016/j.ijcard.2016.07.165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 07/27/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND/OBJECTIVES Epicardium-derived progenitor cells (EPDCs) differentiate into all heart cell types in the embryonic heart, yet their differentiation into cardiomyocytes in the adult heart is limited and poorly described. This may be due to EPDCs lacking myogenic potential or the inert adult heart missing regenerative signals essential for directed differentiation of EPDCs. Herein, we aimed to evaluate the myogenic potential of neonatal EPDCs in adult and neonatal mouse myocardium, as well as in skeletal muscle. The two latter tissues have an intrinsic capability to develop and regenerate, in contrast to the adult heart. METHODS Highly purified mouse EPDCs were transplanted into damaged neonatal and adult myocardium as well as regenerating skeletal muscle. Co-cultures with skeletal myoblasts were used to distinguish fusion independent myogenic conversion. RESULTS No donor EPDC-derived cardiomyocytes were observed in hearts. In contrast, a remarkable contribution of EPDCs to skeletal muscle myofiber formation was evident in vivo. Furthermore, co-cultures of EPDCs with myoblasts showed that EPDCs became part of multinucleated fibers and appeared to acquire myogenic traits independent of a fusion event. Fluorescence activated cell sorting of EPDCs co-cultured with and without myoblasts and subsequent qRT-PCR of 64 transcripts established that the myogenic phenotype conversion was accomplished through induction of a transcriptional myogenic program. CONCLUSION These results suggest that EPDCs may be more myogenic than previously anticipated. But, the heart may lack factors for induction of myogenesis of EPDCs, a scenario that should be taken into consideration when aiming for repair of damaged myocardium by stem cell transplantation.
Collapse
Affiliation(s)
- Ditte C Andersen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Clinical Institute, University of Southern Denmark, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark.
| | - Charlotte H Jensen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| | - Ida Skovrind
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Clinical Institute, University of Southern Denmark, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| | - Rikke Helin Johnsen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Clinical Institute, University of Southern Denmark, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| | - Gunnhildur Asta Traustadottir
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Cardiovascular and Renal Research, University of Southern Denmark, Winsloewparken 21(3rd), 5000 Odense, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| | - Katrine S Aagaard
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Cardiovascular and Renal Research, University of Southern Denmark, Winsloewparken 21(3rd), 5000 Odense, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| | - Suganya Ganesalingam
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Cardiovascular and Renal Research, University of Southern Denmark, Winsloewparken 21(3rd), 5000 Odense, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| | - Søren P Sheikh
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Cardiovascular and Renal Research, University of Southern Denmark, Winsloewparken 21(3rd), 5000 Odense, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| |
Collapse
|
9
|
Greene CA, Green CR, Sherwin T. Transdifferentiation of chondrocytes into neuron-like cells induced by neuronal lineage specifying growth factors. Cell Biol Int 2014; 39:185-91. [PMID: 25183647 DOI: 10.1002/cbin.10358] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 07/06/2014] [Indexed: 12/25/2022]
Abstract
We previously reported that neural-crest-derived stromal cells from adult human and rat corneas can differentiate into neuron-like cells when treated with neuronal lineage specifying growth factors. However, it remains unclear whether this level of cell plasticity is unique to the corneal stromal cell population present in the eye. In this study, non-neural-crest-derived chondrocytes from the xiphosternum of adult rats were subjected to the same differentiation protocol. Cells of the adult rat xiphosternum can also differentiate into neuron-like cells when treated with neurogenic differentiation specifying growth factors. After 1 week in neurogenic differentiation culture conditions, the chondrocytes changed from a round to a stellate morphology and started to express neuron-specific protein neurofilament-200 (NF-200), microtubule associated protein-2 (Map-2), and β-III tubulin. Lineage-specifying growth factors can affect changes in morphology and protein expression of adult cells in culture, findings that challenge the notion of a restricted differentiation potential of adult cell populations and questions the stability of the differentiated state of cells.
Collapse
Affiliation(s)
- Carol Ann Greene
- Department of Ophthalmology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | | | | |
Collapse
|
10
|
Bone and lymph node metastases from neuroblastoma detected by (18)F-DOPA-PET/CT and confirmed by posttherapy (131)I-MIBG but negative on diagnostic (123)I-MIBG scan. Clin Nucl Med 2014; 39:673. [PMID: 24893128 DOI: 10.1097/rlu.0000000000000475] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
11
|
Greene CA, Chang CY, Fraser CJ, Nelidova DE, Chen JA, Lim A, Brebner A, McGhee J, Sherwin T, Green CR. Cells from the adult corneal stroma can be reprogrammed to a neuron-like cell using exogenous growth factors. Exp Cell Res 2014; 322:122-32. [DOI: 10.1016/j.yexcr.2013.12.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 12/05/2013] [Accepted: 12/11/2013] [Indexed: 12/27/2022]
|
12
|
Huang JG, Shen CB, Wu WB, Ren JW, Xu L, Liu S, Yang Q. Primary cilia mediate sonic hedgehog signaling to regulate neuronal-like differentiation of bone mesenchymal stem cells for resveratrol induction in vitro. J Neurosci Res 2014; 92:587-96. [PMID: 24464877 DOI: 10.1002/jnr.23343] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 08/10/2013] [Accepted: 11/10/2013] [Indexed: 01/12/2023]
Affiliation(s)
- Jia-Gui Huang
- Department of Neurology; The First Affiliated Hospital of Chongqing Medical University; Chongqing China
| | - Chang-Bo Shen
- Department of Neurology; The First Affiliated Hospital of Chongqing Medical University; Chongqing China
| | - Wen-Bin Wu
- Department of Neurology; Sichuan Provincial Academy of Medical Sciences and Sichuan Provincial People's Hospital; Chengdu Sichuan China
| | - Jun-Wei Ren
- Department of Neurology; The First Affiliated Hospital of Chongqing Medical University; Chongqing China
- Department of Neurology; Chongqing Fuling Central Hospital, Fuling District; Chongqing China
| | - Lan Xu
- Department of Neurology; The First Affiliated Hospital of Chongqing Medical University; Chongqing China
| | - Shu Liu
- Department of Neurology; The First Affiliated Hospital of Chongqing Medical University; Chongqing China
| | - Qin Yang
- Department of Neurology; The First Affiliated Hospital of Chongqing Medical University; Chongqing China
| |
Collapse
|
13
|
Ullah M, Sittinger M, Ringe J. Transdifferentiation of adipogenically differentiated cells into osteogenically or chondrogenically differentiated cells: phenotype switching via dedifferentiation. Int J Biochem Cell Biol 2014; 46:124-137. [PMID: 24269783 DOI: 10.1016/j.biocel.2013.11.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 10/16/2013] [Accepted: 11/05/2013] [Indexed: 11/25/2022]
Abstract
Reprogramming is a new wave in cellular therapies to achieve the vital goals of regenerative medicine. Transdifferentiation, whereas the differentiated state of cells could be reprogrammed into other cell types, meaning cells are no more locked in their differentiated circle. Hence, cells of choice from abundant and easily available sources such as fibroblast and adipose tissue could be converted into cells of demand, to restore the diseased tissues. Before diverting this new approach into effective clinical use, transdifferentiation could not be simply overlooked, as it challenges the normal paradigms of biological laws, where mature cells transdifferentiate not only within same germ layers, but even across the lineage boundaries. How unipotent differentiated cells reprogram into another, and whether transdifferentiation proceeds via a direct cell-to-cell conversion or needs dedifferentiation. To address such questions, MSC were adipogenically differentiated followed by direct transdifferentiation, and subsequently examined by histology, immunohistochemistry, qPCR and single cell analysis. Direct cellular conversion of adipogenic lineage cells into osteogenic or chondrogenic resulted in mixed culture of both lineage cells (adipogenic and new acquiring osteogenic/chondrogenic phenotypes). On molecular level, such conversion was confirmed by significantly upregulated expression of PPARG, FABP4, SPP1 and RUNX2. Chondrogenic transdifferentiation was verified by significantly upregulated expression of PPARG, FABP4, SOX9 and COL2A1. Single cell analysis did not support the direct cell-to-cell conversion, rather described the involvement of dedifferentiation. Moreover, some differentiated single cells did not change their phenotype and were resistant to transdifferentiation, suggesting that differentiated cells behave differently during cellular conversion. An obvious characterization of differentiated cells could be helpful to understand the process of transdifferentiation.
Collapse
Affiliation(s)
- Mujib Ullah
- Tissue Engineering Laboratory & Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| | - Michael Sittinger
- Tissue Engineering Laboratory & Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| | - Jochen Ringe
- Tissue Engineering Laboratory & Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
14
|
MeCP2 dependent heterochromatin reorganization during neural differentiation of a novel Mecp2-deficient embryonic stem cell reporter line. PLoS One 2012; 7:e47848. [PMID: 23112857 PMCID: PMC3480415 DOI: 10.1371/journal.pone.0047848] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/21/2012] [Indexed: 01/17/2023] Open
Abstract
The X-linked Mecp2 is a known interpreter of epigenetic information and mutated in Rett syndrome, a complex neurological disease. MeCP2 recruits HDAC complexes to chromatin thereby modulating gene expression and, importantly regulates higher order heterochromatin structure. To address the effects of MeCP2 deficiency on heterochromatin organization during neural differentiation, we developed a versatile model for stem cell in vitro differentiation. Therefore, we modified murine Mecp2 deficient (Mecp2−/y) embryonic stem cells to generate cells exhibiting green fluorescent protein expression upon neural differentiation. Subsequently, we quantitatively analyzed heterochromatin organization during neural differentiation in wild type and in Mecp2 deficient cells. We found that MeCP2 protein levels increase significantly during neural differentiation and accumulate at constitutive heterochromatin. Statistical analysis of Mecp2 wild type neurons revealed a significant clustering of heterochromatin per nuclei with progressing differentiation. In contrast we found Mecp2 deficient neurons and astroglia cells to be significantly impaired in heterochromatin reorganization. Our results (i) introduce a new and manageable cellular model to study the molecular effects of Mecp2 deficiency, and (ii) support the view of MeCP2 as a central protein in heterochromatin architecture in maturating cells, possibly involved in stabilizing their differentiated state.
Collapse
|
15
|
Transdifferentiation: a cell and molecular reprogramming process. Cell Tissue Res 2012; 348:379-96. [PMID: 22526624 DOI: 10.1007/s00441-012-1403-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 03/01/2012] [Indexed: 12/13/2022]
Abstract
Evidence has emerged recently indicating that differentiation is not entirely a one-way process, and that it is possible to convert one cell type to another, both in vitro and in vivo. This phenomenon is called transdifferentiation, and is generally defined as the stable switch of one cell type to another. Transdifferentiation plays critical roles during development and in regeneration pathways in nature. Although this phenomenon occurs rarely in nature, recent studies have been focused on transdifferentiation and the reprogramming ability of cells to produce specific cells with new phenotypes for use in cell therapy and regenerative medicine. Thus, understanding the principles and the mechanism of this process is important for producing desired cell types. Here some well-documented examples of transdifferentiation, and their significance in development and regeneration are reviewed. In addition, transdifferentiation pathways are considered and their potential molecular mechanisms, especially the role of master switch genes, are considered. Finally, the significance of transdifferentiation in regenerative medicine is discussed.
Collapse
|
16
|
Parameswaran S, Balasubramanian S, Rao MS, Ahmad I. Concise review: non-cell autonomous reprogramming: a nucleic acid-free approach to induction of pluripotency. Stem Cells 2011; 29:1013-20. [PMID: 21544901 DOI: 10.1002/stem.655] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The reprogramming of somatic cells to a pluripotent state by the expression of a defined set of exogenous transcription factors represents a significant breakthrough for the use of stem cells in regenerative medicine. It has the potential to make autologous stem cell therapy practical and promote better understanding of the disease processes by generating patient specific stem cells. Several strategies have been used to generate induced pluripotent stem cells (iPSCs) that include nucleic acid and non-nucleic acid-based approaches, with and without epigenetic modifications. The purpose of these different approaches for generating iPSCs, besides understanding the underlying mechanism, is to develop a facile method for reprogramming without genetic alteration, suitable for clinical use. Here, we discuss different strategies for generating iPSCs, with an emphasis on a recent non-cell autonomous approach to reprogram somatic progenitors that regenerate cornea to a pluripotent state through the recruitment of endogenous transcription factors.
Collapse
Affiliation(s)
- Sowmya Parameswaran
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198-5840, USA
| | | | | | | |
Collapse
|
17
|
Li W, Liu H, Cheng ZJ, Su YH, Han HN, Zhang Y, Zhang XS. DNA methylation and histone modifications regulate de novo shoot regeneration in Arabidopsis by modulating WUSCHEL expression and auxin signaling. PLoS Genet 2011; 7:e1002243. [PMID: 21876682 PMCID: PMC3158056 DOI: 10.1371/journal.pgen.1002243] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Accepted: 07/06/2011] [Indexed: 12/11/2022] Open
Abstract
Plants have a profound capacity to regenerate organs from differentiated somatic tissues, based on which propagating plants in vitro was made possible. Beside its use in biotechnology, in vitro shoot regeneration is also an important system to study de novo organogenesis. Phytohormones and transcription factor WUSCHEL (WUS) play critical roles in this process but whether and how epigenetic modifications are involved is unknown. Here, we report that epigenetic marks of DNA methylation and histone modifications regulate de novo shoot regeneration of Arabidopsis through modulating WUS expression and auxin signaling. First, functional loss of key epigenetic genes—including METHYLTRANSFERASE1 (MET1) encoding for DNA methyltransferase, KRYPTONITE (KYP) for the histone 3 lysine 9 (H3K9) methyltransferase, JMJ14 for the histone 3 lysine 4 (H3K4) demethylase, and HAC1 for the histone acetyltransferase—resulted in altered WUS expression and developmental rates of regenerated shoots in vitro. Second, we showed that regulatory regions of WUS were developmentally regulated by both DNA methylation and histone modifications through bisulfite sequencing and chromatin immunoprecipitation. Third, DNA methylation in the regulatory regions of WUS was lost in the met1 mutant, thus leading to increased WUS expression and its localization. Fourth, we did a genome-wide transcriptional analysis and found out that some of differentially expressed genes between wild type and met1 were involved in signal transduction of the phytohormone auxin. We verified that the increased expression of AUXIN RESPONSE FACTOR3 (ARF3) in met1 indeed was due to DNA demethylation, suggesting DNA methylation regulates de novo shoot regeneration by modulating auxin signaling. We propose that DNA methylation and histone modifications regulate de novo shoot regeneration by modulating WUS expression and auxin signaling. The study demonstrates that, although molecular components involved in organogenesis are divergently evolved in plants and animals, epigenetic modifications play an evolutionarily convergent role in this process. Plants have a strong ability to generate organs from differentiated somatic tissues. Due to this feature, shoot regeneration in vitro has been used as an important way for producing whole plants in agriculture and biotechnology. Phytohormones and the transcription factor WUSCHEL (WUS) are essential for reprogramming during de novo shoot regeneration. Epigenetic modifications are also critical for mammalian cell differentiation and organogenesis. Here, we show that epigenetic modifications mediate the de novo shoot regeneration in Arabidopsis. Mutations of key epigenetic genes resulted in altered WUS expression and developmental rates of regenerated shoots in vitro. Bisulfite sequencing and chromatin immunoprecipitation revealed that the regulatory regions of WUS were developmentally regulated by both DNA methylation and histone modifications. By transcriptome analysis, we identified that some differentially expressed genes between wild type and met1 are involved in signal transduction of the phytohormone auxin. Our results suggest that DNA methylation and histone modifications regulate de novo shoot regeneration by modulating WUS expression and auxin signaling. The study demonstrates that, although molecular components involved in organogenesis are divergently evolved in plants and animals, epigenetic modifications play an evolutionarily convergent role during de novo organogenesis.
Collapse
Affiliation(s)
- Wei Li
- State Key Laboratory of Crop Biology, Shandong Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, China
| | - Hui Liu
- State Key Laboratory of Crop Biology, Shandong Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, China
| | - Zhi Juan Cheng
- State Key Laboratory of Crop Biology, Shandong Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, China
| | - Ying Hua Su
- State Key Laboratory of Crop Biology, Shandong Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, China
| | - Hua Nan Han
- State Key Laboratory of Crop Biology, Shandong Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, China
| | - Yan Zhang
- State Key Laboratory of Crop Biology, Shandong Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, China
| | - Xian Sheng Zhang
- State Key Laboratory of Crop Biology, Shandong Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, China
- * E-mail:
| |
Collapse
|
18
|
Kurpisz M. Preclinical and clinical studies on application of human myoblasts in regeneration of the postinfarction heart. Transplant Proc 2010; 42:3323-7. [PMID: 20970682 DOI: 10.1016/j.transproceed.2010.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Principal definitions of stem cell subdivisions as well as of the physiology of the renewal of their descendants have been elucidated in recent years. Regeneration mechanisms have been outlined using as an example the intestinal villus niche. Sources of stem cells for preclinical studies and the main conclusions from clinical trials have been developed in the vast majority in the 21st century. Meta-analyses and summaries have been focused so far on bone marrow stem cells and muscle-derived stem cells, which have been most often tried to date. Polish clinical trials on postinfarcted hearts have been consistent with the world literature regarding the major conclusions for myocardial regeneration. The controversies include possible side effects of stem cell applications. The necessity for genetic modification of the stem cells, which are mainly myoblasts, has been justified by the results of recently performed trials, initial examples including transfections of proangiogenic factors into human primary myoblast suspensions.
Collapse
Affiliation(s)
- M Kurpisz
- Department of Reproductive Biology and Stem Cells, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland.
| |
Collapse
|
19
|
Zhang Y, Shen W, Sun B, Lv C, Dou Z. Plasticity of marrow mesenchymal stem cells from human first-trimester fetus: from single-cell clone to neuronal differentiation. Cell Reprogram 2010; 13:57-64. [PMID: 20954967 DOI: 10.1089/cell.2010.0044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Recent results have shown that bone marrow mesenchymal stem cells (BMSCs) from human first-trimester abortus (hfBMSCs) are closer to embryonic stem cells and perform greater telomerase activity and faster propagation than mid- and late-prophase fetal and adult BMSCs. However, no research has been done on the plasticity of hfBMSCs into neuronal cells using single-cell cloned strains without cell contamination. In this study, we isolated five single cells from hfBMSCs and obtained five single-cell cloned strains, and investigated their biological property and neuronal differentiation potential. We found that four of the five strains showed similar expression profile of surface antigen markers to hfBMSCs, and most of them differentiated into neuron-like cells expressing Nestin, Pax6, Sox1, β-III Tubulin, NF-L, and NSE under induction. One strain showed different expression profile of surface antigen markers from the four strains and hfBMSCs, and did not differentiate toward neuronal cells. We demonstrated for the first time that some of single-cell cloned strains from hfBMSCs can differentiate into nerve tissue-like cell clusters under induction in vitro, and that the plasticity of each single-cell cloned strain into neuronal cells is different.
Collapse
Affiliation(s)
- Yihua Zhang
- Shaanxi Branch of National Stem Cell Engineering and Technology Centre, College of Veterinary Medicine, Northwest A&F University , Shaanxi, People's Republic of China
| | | | | | | | | |
Collapse
|
20
|
Abstract
Stem cell therapy is a potential treatment for spinal cord injury (SCI), and a variety of different stem cell types have been evaluated in animal models and humans with SCI. No consensus exists regarding the type of stem cell, if any, that will prove to be effective therapeutically. Most data suggest that no single therapy will be sufficient to overcome all the biological complications caused by SCI. Rationales for therapeutic use of stem cells for SCI include replacement of damaged neurons and glial cells, secretion of trophic factors, regulation of gliosis and scar formation, prevention of cyst formation, and enhancement of axon elongation. Most therapeutic approaches that use stem cells involve implantation of these cells into the spinal cord. The attendant risks of stem cell therapy for SCI--including tumor formation, or abnormal circuit formation leading to dysfunction--must be weighed against the potential benefits of this approach. This Review will examine the biological effects of SCI, the opportunities for stem cell treatment, and the types of stem cells that might be used therapeutically. The limited information available on the possible benefits of stem cell therapy to humans will also be discussed.
Collapse
Affiliation(s)
- Vibhu Sahni
- MGH-HMS Center for Nervous System Repair, Harvard Medical School, 50 Blossom Street, Boston, MA 02114, USA
| | | |
Collapse
|
21
|
Liao D, Gong P, Li X, Tan Z, Yuan Q. Co-culture with Schwann cells is an effective way for adipose-derived stem cells neural transdifferentiation. Arch Med Sci 2010; 6:145-51. [PMID: 22371738 PMCID: PMC3281332 DOI: 10.5114/aoms.2010.13885] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2008] [Revised: 10/22/2008] [Accepted: 11/22/2008] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Adipose-derived stem cells (ADSCs) could accomplish neural transdifferentiation with the presence of certain growth factors in vitro. It has been proved that bone marrow stromal cells (BMSCs) can realize neural transdifferentiation only by being co-cultured with Schwann cells (SCs), and in our former studies we have confirmed that ADSCs could do so too. This paper aims to investigate whether the neural induction efficiency of co-culture is as high as that of other strategies using chemicals or chemicals combined with some growth factors. MATERIAL AND METHODS We isolated and multiplied ADSCs from adult Sprague-Dawley rats, and SCs from sciatic nerves of 1-to-2-day-old Sprague-Dawley rat pups, then induced ADSCs neural transdifferentiation through 2% dimethyl sulphoxide (DMSO) and DMSO combined with growth factors. Meanwhile we co-cultured ADSCs and SCs in Transwell culture dishes without intercellular contacts. Immunostaining and RT-PCR were adopted to investigate the neural transdifferentiation of ADSCs. Then we compared the expression differences for genes S100, nestin and GFAP of the above three protocols by real-time PCR. RESULTS Both immunostaining and RT-PCR proved that ADSCs could accomplish neural transdifferentiation through each of the above three protocols. And real-time PCR further shows that the gene expression relative quantities for the above three genes are not statistically different between co-culture and induction through DMSO combined with growth factors (p > 0.05), but both of them are statistically different from induction only by DMSO (p < 0.05). CONCLUSIONS Co-culturing ADSCs and SCs may be a simple, effective and practical way for ADSCs neural transdifferentiation.
Collapse
Affiliation(s)
- Dapeng Liao
- State Key Laboratory of Oral Diseases (Sichuan University), Chengdu, China
| | - Ping Gong
- Dental Implant Centre, West China College of Stomatology, Sichuan University, Chengdu, China
| | - Xiaojie Li
- State Key Laboratory of Oral Diseases (Sichuan University), Chengdu, China
| | - Zhen Tan
- Dental Implant Centre, West China College of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases (Sichuan University), Chengdu, China
| |
Collapse
|
22
|
Ni WF, Yin LH, Lu J, Xu HZ, Chi YL, Wu JB, Zhang N. In vitro neural differentiation of bone marrow stromal cells induced by cocultured olfactory ensheathing cells. Neurosci Lett 2010; 475:99-103. [PMID: 20347932 DOI: 10.1016/j.neulet.2010.03.056] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Revised: 03/10/2010] [Accepted: 03/22/2010] [Indexed: 12/26/2022]
Abstract
Bone marrow stromal cells (BMSCs) could be induced to differentiate into neural cells under certain conditions, nevertheless, optimal protocols that could be reproducible and reliable in generating transplantable BMSCs in vitro are still not available. We studied for the first time the neural differentiation of BMSCs induced by coculturing with olfactory ensheathing cells (OECs). BMSCs and OECs were isolated from bone marrow and nasal olfactory lamina propria of adult SD rats respectively, then brought to coculture with transwell culture dishes. At various time points (0h, 6h, 12h, 24h, 72h, 1 week and 2 weeks post-coculture), BMSCs were morphologically observed and processed for immunofluorescence and reverse transcription-polymerase chain reaction (RT-PCR). The number of cells assuming neural morphology dramatically increased at 1- and 2-week-post-coculture, so as the number of immunoreactive cells labeled by neural markers NSE, beta-III-tubulin, MAP2, GFAP and p75(NTR). Our findings demonstrate that BMSCs can efficiently differentiate into neural cells when coculturing with OECs, and the present protocol provides an alternative neurogenesis pathway for generating sufficient numbers of neural cells from BMSCs.
Collapse
Affiliation(s)
- Wen-Fei Ni
- Department of Spine Surgery, The Second Affiliated Hospital of Wenzhou Medical College, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Soloviev A, Prudnikov I, Tsyvkin V, Tishkin S, Kyrychenko S, Zelensky S, Ivanova I. Electrophysiological and contractile evidence of the ability of human mesenchymal stromal cells to correct vascular malfunction in rats after ionizing irradiation. J Physiol Sci 2010; 60:161-72. [PMID: 20052569 PMCID: PMC10717491 DOI: 10.1007/s12576-009-0080-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Accepted: 12/08/2009] [Indexed: 10/20/2022]
Abstract
The effect of intravenous administration of human mesenchymal stromal stem cells (hMSC) has been evaluated by means of large-conductance calcium-dependent potassium channel (BK(Ca)) activity measurements in thoracic aorta smooth muscle cells (SMC) obtained from non-fatal whole-body irradiated rats, using the patch clamp technique in whole-cell modification, and the standard acetylcholine (ACh) test to evaluate functional endothelium integrity using SM contractile recordings. Myofilament calcium sensitivity was estimated using simultaneous contractile recordings versus [Ca(2+)](i). Arterial blood was measured in intact and irradiated rats before and after hMSC administration. Stimulation of isolated SMC from the control group of animals with depolarizing voltage steps showed that outward K(+) currents sensitive to the BK(Ca) inhibitor paxilline were expressed. Outward currents in SMC obtained from irradiated animals were significantly reduced on the 30th day of post-irradiation. Irradiation led to a significant elevation in arterial blood pressure and reduced ACh-induced relaxation responses in irradiated rats as compared with the control group. Simultaneous measurements of contractile force and [Ca(2+)](i) showed that myofilament Ca(2+) sensitivity had increased following irradiation. Intravenously injected hMSC effectively restored BK(Ca) current and the amplitude of ACh-induced endothelium-dependent vasodilatation in vascular tissues obtained from post-irradiated rats. SMC obtained from irradiated rats treated with hMSC demonstrated a significantly increased paxilline-sensitive component of outward potassium currents, indicating that BK(Ca) activity had been restored. hMSC administration normalized increased blood pressure and myofilament Ca(2+) sensitivity in irradiated animals. When administered to healthy rats, hMSC were without effects on either of these. This study does not provide any immunohistochemical proof of hMSC engraftment in the host rats. PCR analysis showed that hMSCs were negative for hematopoietic cell markers and positive for hMSC markers. There were no clinical signs of graft-versus-host disease throughout the experimental period of 30 days. The data obtained suggest that hMSC demonstrate a clearly expressed ability to normalize vascular function damaged following irradiation, i.e. to reduce an elevated arterial blood pressure and myofilament Ca(2+) sensitivity, and to repair BK(Ca) function and endothelium-dependent relaxation in vascular tissues obtained from irradiated animals. Thus, hMSC seem to be worthwhile therapeutic approach in cases of ionizing irradiation accident or radiation beam therapy.
Collapse
Affiliation(s)
- Anatoly Soloviev
- Department for Experimental Therapeutics, Institute of Pharmacology and Toxicology, Academy of Medical Science, 14 Eugene Pottier Str., 03057, Kiev, Ukraine.
| | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
The application of stem cells and their use in tissue-engineering approaches is emerging in clinical therapeutic intervention strategies. The use of adult stem cells, either autologous or allogenic, does not raise ethical concerns, in contrast to embryonic stem cells. Mesenchymal stromal cells (MSCs) can be easily obtained from bone marrow or from adipose tissue and further expanded in vitro. Due to their differentiation capacity, MSCs are very attractive for tissue engineering purposes. Furthermore, MSCs secrete a variety of mediators that have beneficial effects on the regenerating tissue. In this review we give an insight into stem cell hierarchy, define the properties of MSCs and summarize recent reports of their administration in urological diseases.
Collapse
Affiliation(s)
- Katrin Montzka
- Department of Urology, RWTH University Aachen, Aachen, Germany.
| | | |
Collapse
|
25
|
Abstract
Over the past few years a great deal of interest has been generated in using stem cells/progenitors to treat degenerative diseases that afflict different tissues, including retina. This interest is due to the defining properties of stem cells/progenitors, the ability of these cells to self-renew and generate all the basic cell types of the particular tissue to which they belong. In addition, the recent reports of plasticity of the adult tissue-specific stem cells/progenitors and directed differentiation of the embryonic cells (ES) has fueled the hope for cell and gene therapy using stem cells from heterologous sources. Will this approach work for treating retinal degeneration? Here, we review the current state of knowledge about obtaining retinal cells from heterologous sources, including ES cells.
Collapse
Affiliation(s)
- Ani M Das
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, 68198, USA
| | | | | |
Collapse
|
26
|
Mathews LA, Crea F, Farrar WL. Epigenetic gene regulation in stem cells and correlation to cancer. Differentiation 2009; 78:1-17. [PMID: 19443100 PMCID: PMC2706282 DOI: 10.1016/j.diff.2009.04.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Revised: 04/03/2009] [Accepted: 04/06/2009] [Indexed: 01/08/2023]
Abstract
Through the classic study of genetics, much has been learned about the regulation and progression of human disease. Specifically, cancer has been defined as a disease driven by genetic alterations, including mutations in tumor-suppressor genes and oncogenes, as well as chromosomal abnormalities. However, the study of normal human development has identified that in addition to classical genetics, regulation of gene expression is also modified by 'epigenetic' alterations including chromatin remodeling and histone variants, DNA methylation, the regulation of polycomb group proteins, and the epigenetic function of non-coding RNA. These changes are modifications inherited during both meiosis and mitosis, yet they do not result in alterations of the actual DNA sequence. A number of biological questions are directly influenced by epigenetics, such as how does a cell know when to divide, differentiate or remain quiescent, and more importantly, what happens when these pathways become altered? Do these alterations lead to the development and/or progression of cancer? This review will focus on summarizing the limited current literature involving epigenetic alterations in the context of human cancer stems cells (CSCs). The extent to which epigenetic changes define cell fate, identity, and phenotype are still under intense investigation, and many questions remain largely unanswered. Before discussing epigenetic gene silencing in CSCs, the different classifications of stem cells and their properties will be introduced. This will be followed by an introduction to the different epigenetic mechanisms. Finally, there will be a discussion of the current knowledge of epigenetic modifications in stem cells, specifically what is known from rodent systems and established cancer cell lines, and how they are leading us to understand human stem cells.
Collapse
Affiliation(s)
- Lesley A. Mathews
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | | | - W. L. Farrar
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
27
|
Montzka K, Lassonczyk N, Tschöke B, Neuss S, Führmann T, Franzen R, Smeets R, Brook GA, Wöltje M. Neural differentiation potential of human bone marrow-derived mesenchymal stromal cells: misleading marker gene expression. BMC Neurosci 2009; 10:16. [PMID: 19257891 PMCID: PMC2655300 DOI: 10.1186/1471-2202-10-16] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Accepted: 03/03/2009] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND In contrast to pluripotent embryonic stem cells, adult stem cells have been considered to be multipotent, being somewhat more restricted in their differentiation capacity and only giving rise to cell types related to their tissue of origin. Several studies, however, have reported that bone marrow-derived mesenchymal stromal cells (MSCs) are capable of transdifferentiating to neural cell types, effectively crossing normal lineage restriction boundaries. Such reports have been based on the detection of neural-related proteins by the differentiated MSCs. In order to assess the potential of human adult MSCs to undergo true differentiation to a neural lineage and to determine the degree of homogeneity between donor samples, we have used RT-PCR and immunocytochemistry to investigate the basal expression of a range of neural related mRNAs and proteins in populations of non-differentiated MSCs obtained from 4 donors. RESULTS The expression analysis revealed that several of the commonly used marker genes from other studies like nestin, Enolase2 and microtubule associated protein 1b (MAP1b) are already expressed by undifferentiated human MSCs. Furthermore, mRNA for some of the neural-related transcription factors, e.g. Engrailed-1 and Nurr1 were also strongly expressed. However, several other neural-related mRNAs (e.g. DRD2, enolase2, NFL and MBP) could be identified, but not in all donor samples. Similarly, synaptic vesicle-related mRNA, STX1A could only be detected in 2 of the 4 undifferentiated donor hMSC samples. More significantly, each donor sample revealed a unique expression pattern, demonstrating a significant variation of marker expression. CONCLUSION The present study highlights the existence of an inter-donor variability of expression of neural-related markers in human MSC samples that has not previously been described. This donor-related heterogeneity might influence the reproducibility of transdifferentiation protocols as well as contributing to the ongoing controversy about differentiation capacities of MSCs. Therefore, further studies need to consider the differences between donor samples prior to any treatment as well as the possibility of harvesting donor cells that may be inappropriate for transplantation strategies.
Collapse
Affiliation(s)
- Katrin Montzka
- Department of Neurology, RWTH Aachen University, Aachen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Acosta S, Lavarino C, Paris R, Garcia I, de Torres C, Rodríguez E, Beleta H, Mora J. Comprehensive characterization of neuroblastoma cell line subtypes reveals bilineage potential similar to neural crest stem cells. BMC DEVELOPMENTAL BIOLOGY 2009; 9:12. [PMID: 19216736 PMCID: PMC2647534 DOI: 10.1186/1471-213x-9-12] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2008] [Accepted: 02/12/2009] [Indexed: 12/30/2022]
Abstract
Background Neuroblastic tumors (NBT) derive from neural crest stem cells (NCSC). Histologically, NBT are composed by neuroblasts and Schwannian cells. In culture, neuroblastic (N-), substrate-adherent (S-) and intermediate phenotype (I-) cell subtypes arise spontaneously. Methods Here, neuroblastoma (NB) cell line subtypes were characterized according to embryonic peripheral nervous system development markers (GAP43, Phox2b, Sox10, c-kit, GD2, NF68, vimentin, S100β, calcyclin and ABCG2), morphological features, gene expression and differentiation potential. I-type cells were investigated as a bipotential (neuronal and glial) differentiation stage. Results Positive immunostaining of NCSC (GAP43, c-kit, NF68, vimentin and Phox2b) and undifferentiated cell (ABCG2) markers was observed in all NB subtypes. N- and I-type cells displayed cytoplasmic membrane GD2 staining, while nuclear calcyclin was restricted to S-type. N- and I-type cells showed similar phenotype and immunoreactivity pattern. Differential gene expression was associated with each cell subtype. N- and I-type cells displayed similar differentiation capacity towards neuronal and glial lineage fates. S-type cells, upon induction, did not show a neuronal-like phenotype, despite gene expression changes. Conclusion Results suggest that N- and I-type NB cell subtypes represent an immature bilineage stage, able to progress towards neuronal and glial fates upon induction of differentiation. S-type cells appear irreversibly committed to a glial lineage fate.
Collapse
Affiliation(s)
- Sandra Acosta
- Developmental tumor biology laboratory, Hospital Sant Joan de Déu, Barcelona, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Abstract
The working definition of a stem cell includes self-renewal and the ability to differentiate into several cell types. There are also aspects of clonality and potency. Stem cells can be derived from early embryos after the formation of the blastocyst or from fetal, postnatal, or adult sources. Neural stem cells (NSCs) arise from embryonic ectoderm that forms neuroepithelial cells. The neuroepithelial cells generate radial glia that produce fetal and adult NSCs within the central nervous system (CNS). Adult NSC and restricted progenitors are found in the several regions of the CNS throughout life. Human embryonic stem cells, with their ability for self-renewal, clonal capacity, normal karyotype, and potential to form NSCs, easily may be the best source of NSCs and progenitors for treating disease. However, the complexity of NSCs, neural patterning, and the formation of multiple populations of neurons, astrocytes, and oligodendrocytes warrant the need for intense studies to characterize these cells and to define the microenvironment that will be needed to support them in the diseased CNS. Ways to produce well-defined populations, avoid oncogenicity, and ensure survival need to be clarified before clinical application can begin.
Collapse
Affiliation(s)
- Leslie P Weiner
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
31
|
Abstract
Stem cells, although difficult to define, hold great promise as tools for understanding development and as therapeutic agents. However, as with any new field, uncritical enthusiasm can outstrip reality. In this review, we have listed nine common myths that we believe affect our approach to evaluating stem cells for therapy. We suggest that careful consideration needs to be given to each of these issues when evaluating a particular cell for its use in therapy. Data need to be collected and reported for failed as well as successful experiments and a rigorous scientific approach taken to evaluate the undeniable promise of stem cell biology.
Collapse
Affiliation(s)
- Tim Magnus
- Stem Cell Section, Laboratory of Neurosciences, National Institute on Aging, NIH333 Cassell Drive, Room 406A, Baltimore, MD 21224, USA
| | - Ying Liu
- Stem Cell Section, Laboratory of Neurosciences, National Institute on Aging, NIH333 Cassell Drive, Room 406A, Baltimore, MD 21224, USA
| | - Graham C Parker
- Children's Research Center of Michigan, The Carman and Ann Adams Department of Pediatrics, Wayne State University School of Medicine, Children's Hospital of MichiganDetroit, MI 48201, USA
| | - Mahendra S Rao
- Stem Cell Section, Laboratory of Neurosciences, National Institute on Aging, NIH333 Cassell Drive, Room 406A, Baltimore, MD 21224, USA
- Corporate Research Laboratories, Invitrogen Corporation1620 Faraday Avenue, Carlsbad, CA 92008, USA
- Author for correspondence ()
| |
Collapse
|
32
|
Neurotrophic Schwann-cell factors induce neural differentiation of bone marrow stromal cells. Neuroreport 2008; 18:1713-7. [PMID: 17921874 DOI: 10.1097/wnr.0b013e3282f0d3b0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Neural transdifferentiation of bone marrow stromal cells has been questioned, because cell fusion could explain the development of new cell types, misinterpreted as transdifferentiated cells. We performed here cocultures of bone marrow stromal cells and Schwann cells, without possibility that both cell types can establish contact. In these conditions, bone marrow stromal cells expressed nestin 4 h after beginning cocultures, and strong expression of neuronal markers was disclosed at 72 h, increasing at 1 and 2 weeks. Our results support that neural transdifferentiation of bone marrow stromal cells is induced by soluble factors provided by glial cells, and suggest that cell fusion should not be significant when local bone marrow stromal cells administration for neural repair is considered.
Collapse
|
33
|
Neural transdifferentiation of bone marrow stromal cells obtained by chemical agents is a short-time reversible phenomenon. Neurosci Res 2007; 60:275-80. [PMID: 18164086 DOI: 10.1016/j.neures.2007.11.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Revised: 10/17/2007] [Accepted: 11/13/2007] [Indexed: 11/21/2022]
Abstract
Bone marrow stromal cells (BMSC) can acquire morphological and immunohistochemical features of neural cells when they are treated with diverse chemical agents, a finding interpreted as result of cell transdifferentiation. With the purpose of a better knowledge of the possible utility of BMSC for strategies of Nervous System (NS) repair, we have studied the morphological and immunohistochemical changes induced in BMSC by chemical agents, in comparison with those that happen when BMSC are co-cultured with Schwann cells. While chemical BMSC transdifferentiation is a short-time reversible phenomenon, BMSC transdifferentiation obtained by Schwann cell-derived neurotrophic factors remains stable after it has been reached. These findings question the possible clinical utility of BMSC trandifferentiation using chemical agents, and support that neural transdifferentiation of BMSC is a biological phenomenon that can be obtained in vivo because of the presence of environmental factors.
Collapse
|
34
|
Williams MD, Hanna EY, El-Naggar AK. Anaplastic ameloblastic fibrosarcoma arising from recurrent ameloblastic fibroma: restricted molecular abnormalities of certain genes to the malignant transformation. ACTA ACUST UNITED AC 2007; 104:72-5. [PMID: 17577546 DOI: 10.1016/j.tripleo.2006.07.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Revised: 06/30/2006] [Accepted: 07/17/2006] [Indexed: 12/17/2022]
Abstract
A rare case of anaplastic ameloblastic fibrosarcoma (AS) arising in an ameloblastic fibroma (AF) of the maxilla of a 48-year-old patient 10 years after the primary excision is presented. The recurrent tumor retained focal areas of AF but manifested heterogeneous malignant features ranging from low-grade spindle to highly pleomorphic sarcomas. Biomarker analysis showed alterations of the p53 and c-KIT genes restricted to the sarcomatous component. The biological implications of these findings in the future management of these tumors are discussed.
Collapse
Affiliation(s)
- Michelle D Williams
- Department of Pathology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | |
Collapse
|
35
|
|
36
|
Roh J, Cho EA, Seong I, Limb JK, Lee S, Han SJ, Kim J. Down-regulation of Sox10 with specific small interfering RNA promotes transdifferentiation of Schwannoma cells into myofibroblasts. Differentiation 2006; 74:542-51. [PMID: 17177851 DOI: 10.1111/j.1432-0436.2006.00084.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neural crest-derived cells are noted for the long lasting plasticity during lineage commitment process and the potential for transdifferentiation into other neural crest derivatives. Schwann cells in particular have been reported to transdifferentiate into melanocytes and myofibroblasts. Detailed studies of transdifferentiation at the molecular level have been hampered by difficulty in isolating sufficient quantity of primary cells or cellular materials. Here, we describe a robust in vitro system in which Schwannoma cells undergo an apparent transdifferentiation into myofibroblasts. Importantly, we induce the transdifferentiation by down-regulating a single transcription factor, Sox10, thereby identifying a key molecular event in this process. Myofibroblasts thus generated showed carbachol-stimulated contraction and calcium transients and express several established myofibroblast-specific genes. These results suggest that generating desired cell types based on "knock-down" of critical genes may be a viable strategy.
Collapse
Affiliation(s)
- Jiwon Roh
- Division of Molecular Life Sciences and Center for Cell Signaling Research, Ewha Womans University, Sudaemun-gu, Seoul 120-750, Korea
| | | | | | | | | | | | | |
Collapse
|
37
|
Batts SA, Raphael Y. Transdifferentiation and its applicability for inner ear therapy. Hear Res 2006; 227:41-7. [PMID: 17070000 DOI: 10.1016/j.heares.2006.08.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2006] [Revised: 08/16/2006] [Accepted: 08/31/2006] [Indexed: 11/20/2022]
Abstract
During normal development, cells divide, then differentiate to adopt their individual form and function in an organism. Under most circumstances, mature cells cannot transdifferentiate, changing their fate to adopt a different form and function. Because differentiated cells cannot usually divide, the repair of injuries as well as regeneration largely depends on the activation of stem cell reserves. The mature cochlea is an exception among epithelial cell layers in that it lacks stem cells. Consequently, the sensory hair cells that receive sound information cannot be replaced, and their loss results in permanent hearing impairment. The lack of a spontaneous cell replacement mechanism in the organ of Corti, the mammalian auditory sensory epithelium, has led researchers to investigate circumstances in which transdifferentiation does occur. The hope is that this information can be used to design therapies to replace lost hair cells and restore impaired hearing in humans.
Collapse
Affiliation(s)
- Shelley A Batts
- Department of Otolaryngology, Kresge Hearing Research Institute, MSRB-3, Room 9301, Ann Arbor, MI 48109-0648, USA
| | | |
Collapse
|
38
|
Buzańska L, Jurga M, Stachowiak EK, Stachowiak MK, Domańska-Janik K. Neural stem-like cell line derived from a nonhematopoietic population of human umbilical cord blood. Stem Cells Dev 2006; 15:391-406. [PMID: 16846376 DOI: 10.1089/scd.2006.15.391] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The ability of stem and progenitor cells to proliferate and differentiate into other lineages is widely viewed as a characteristic of stem cells. Previously, we have reported that cells from a CD34(-) (nonhematopoietic) adherent subpopulation of human cord blood can acquire a feature of multipotential neural progenitors in vitro. In the present study, using these cord blood-derived stem cells, we have established a clonal cell line termed HUCB-NSCs (human umbilical cord blood-neural stem cells) that expresses several neural antigens and has been grown in culture for more than 60 passages. During this time, HUCB-NSCs retained their growth rate, the ability to differentiate into neuronal-, astrocyte-, and oligodendrocyte-like cells and displayed a stable karyotype. DNA microarray analysis of HUCB-NSCs revealed enhanced expression of selected genes encoding putative stem and progenitor cell markers when compared to other mononuclear cells. dBcAMP-induced HUCBNSCs were further differentiated into more advanced neuronal cells. This is the first report of the establishment and characterization of a nontransformed HUCB-NSC line that can be grown continuously in a monolayer culture and induced to terminal differentiation. These cells should further our understanding of the regulatory mechanisms involved in NSC self-renewal and differentiation.
Collapse
|
39
|
Swanger SA, Neuhuber B, Himes BT, Bakshi A, Fischer I. Analysis of allogeneic and syngeneic bone marrow stromal cell graft survival in the spinal cord. Cell Transplant 2006; 14:775-86. [PMID: 16454352 DOI: 10.3727/000000005783982594] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bone marrow stromal cells (MSC) are attractive candidates for developing cell therapies for central nervous system (CNS) disorders. They can be easily obtained, expanded in culture, and promote modest functional recovery following transplantation into animal models of injured or degenerative CNS. While syngeneic MSC grafts can be used efficiently, achieving long-term survival of allogeneic MSC grafts has been a challenge. We hypothesize that improved graft survival will enhance the functional recovery promoted by MSC. To improve MSC graft survival, we tested two dosages of the immune suppressant cyclosporin A (CsA) in an allogeneic model. Syngeneic transplantation of MSC where cells survive well without immune suppression was used as a control. Sprague-Dawley rats treated with standard dose (n = 12) or high-dose (n = 12) CsA served as allogeneic hosts; Fisher 344 rats (n = 12) served as syngeneic hosts. MSC were derived from transgenic Fisher 344 rats expressing human placental alkaline phosphatase and were grafted into cervical spinal cord. Animals treated with standard dose CsA showed significant decreases in allograft size 4 weeks posttransplantation; high CsA doses yielded significantly better graft survival 4 and 8 weeks posttransplantation compared to standard CsA. As expected, syngeneic MSC transplants showed good graft survival after 4 and 8 weeks. To investigate MSC graft elimination, we analyzed immune cell infiltration and cell death. Macrophage infiltration was high after 1 week in all groups. After 4 weeks, high-dose CsA and syngeneic animals showed significant reductions in macrophages at the graft site. Few T lymphocytes were detected in any group at each time point. Cell death occurred throughout the study; however, little apoptotic activity was detected. Histochemical analysis revealed no evidence of neural differentiation. These results indicate that allogeneic transplantation with appropriate immune suppression permits long-term survival of MSC; thus, both allogeneic and syngeneic strategies could be utilized in devising novel therapies for CNS injury.
Collapse
Affiliation(s)
- Sharon A Swanger
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129, USA
| | | | | | | | | |
Collapse
|
40
|
Abstract
Stem cell research has known an enormous development, and cellular transplantation holds great promise for regenerative medicine. However, some aspects, such as the mechanisms underlying stem cell plasticity (cell fusion vs true transdifferentiation) and the functional improvement after stem cell transplantation, are highly debated. Furthermore, the great variability in methodology used by several groups, sometimes leads to confusing, contradicting results. In this chapter, we review a number of studies in this area with an eye on possible technical and other difficulties in interpretation of the obtained results.
Collapse
Affiliation(s)
- Karen Ann Pauwelyn
- University of Leuven, Stem Cell Institute Leuven (SCIL)/Laboratory of Hepatology, UZ Gasthuisberg, Herestraat 49, 3000 Leuven, Belgium
| | | |
Collapse
|
41
|
Abstract
ESC derivation, use and SCNT have raised many moral and ethical issues. In this opinion piece I have focused on the argument that morally less ambiguous alternatives to ESC derived from the ICM of blastocysts exist. These possibilities range from using multiple adult stem cell populations each of which is uniquely suited for a particular disease target or identifying adult ESC-like populations, using transdifferentiated ESC-like cells or alternate methods of deriving ESC. I suggest that while it is important to support such efforts, current results do not provide sufficient compelling data to allow one to stop the use of ESC and perhaps adult cells will never be a reliable alternative. All options need to be fully explored and decisions need to be made with scientific rigor and respect for each individual's moral compass.
Collapse
Affiliation(s)
- Mahendra S Rao
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
42
|
Tsai MS, Hwang SM, Tsai YL, Cheng FC, Lee JL, Chang YJ. Clonal amniotic fluid-derived stem cells express characteristics of both mesenchymal and neural stem cells. Biol Reprod 2005; 74:545-51. [PMID: 16306422 DOI: 10.1095/biolreprod.105.046029] [Citation(s) in RCA: 191] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Recent evidence has shown that amniotic fluid may be a novel source of fetal stem cells for therapeutic transplantation. We previously developed a two-stage culture protocol to isolate a population of amniotic fluid-derived mesenchymal stem cells (AFMSCs) from second-trimester amniocentesis. AFMSCs maintain the capacity to differentiate into multiple mesenchymal lineages and neuron-like cells. It is unclear whether amniotic fluid contains heterogeneous populations of stem cells or a subpopulation of primitive stem cells that are similar to marrow stromal cells showing the behavior of neural progenitors. In this study, we showed a subpopulation of amniotic fluid-derived stem cells (AF-SCs) at the single-cell level by limiting dilution. We found that NANOG- and POU5F1 (also known as OCT4)-expressing cells still existed in the expanded single cell-derived AF-SCs. Aside from the common mesenchymal characteristics, these clonal AF-SCs also exhibit multiple phenotypes of neural-derived cells such as NES, TUBB3, NEFH, NEUNA60, GALC, and GFAP expressions both before and after neural induction. Most importantly, HPLC analysis showed the evidence of dopamine release in the extract of dopaminergic-induced clonal AF-SCs. The results of this study suggest that besides being an easily accessible and expandable source of fetal stem cells, amniotic fluid will provide a promising source of neural progenitor cells that may be used in future cellular therapies for neurodegenerative diseases and nervous system injuries.
Collapse
Affiliation(s)
- Ming-Song Tsai
- Prenatal Diagnosis Center, Cathay General Hospital, Taipei 106, Taiwan.
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
This review discusses current understanding of the role that endogenous and exogenous progenitor cells may have in the treatment of the diseased heart. In the last several years, a major effort has been made in an attempt to identify immature cells capable of differentiating into cell lineages different from the organ of origin to be employed for the regeneration of the damaged heart. Embryonic stem cells (ESCs) and bone marrow-derived cells (BMCs) have been extensively studied and characterized, and dramatic advances have been made in the clinical application of BMCs in heart failure of ischemic and nonischemic origin. However, a controversy exists concerning the ability of BMCs to acquire cardiac cell lineages and reconstitute the myocardium lost after infarction. The recognition that the adult heart possesses a stem cell compartment that can regenerate myocytes and coronary vessels has raised the unique possibility to rebuild dead myocardium after infarction, to repopulate the hypertrophic decompensated heart with new better functioning myocytes and vascular structures, and, perhaps, to reverse ventricular dilation and wall thinning. Cardiac stem cells may become the most important cell for cardiac repair.
Collapse
Affiliation(s)
- Annarosa Leri
- Cardiovascular Research Institute, Department of Medicine, New York Medical College, Valhalla, NY10595, USA
| | | | | |
Collapse
|
44
|
Abstract
Autoimmune inflammatory diseases of the central nervous system (CNS) are highly complex in their interaction of different cell populations. The main therapy focus in the last years has been the inhibition of the immune system. Recent progress has shown that endogenous as well as transplanted neural stem cells might positively influence the outcome of such diseases. In this review, we discuss the current concept of the underlying pathogenesis with a specific focus on local CNS cells and potential treatment options.
Collapse
Affiliation(s)
- T Magnus
- Laboratory of Neurosciences, National Institute on Aging, Baltimore, Maryland, 21224, USA.
| | | |
Collapse
|
45
|
Ranganath RM. Asymmetric cell divisions in flowering plants - one mother, "two-many" daughters. PLANT BIOLOGY (STUTTGART, GERMANY) 2005; 7:425-48. [PMID: 16163608 DOI: 10.1055/s-2005-865899] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Plant development shows a fascinating range of asymmetric cell divisions. Over the years, however, cellular differentiation has been interpreted mostly in terms of a mother cell dividing mitotically to produce two daughter cells of different fates. This popular view has masked the significance of an entirely different cell fate specification pathway, where the mother cell first becomes a coenocyte and then cellularizes to simultaneously produce more than two specialized daughter cells. The "one mother - two different daughters" pathways rely on spindle-assisted mechanisms, such as translocation of the nucleus/spindle to a specific cellular site and orientation of the spindle, which are coordinated with cell-specific allocation of cell fate determinants and cytokinesis. By contrast, during "coenocyte-cellularization" pathways, the spindle-assisted mechanisms are irrelevant since cell fate specification emerges only after the nuclear divisions are complete, and the number of specialized daughter cells produced depends on the developmental context. The key events, such as the formation of a coenocyte and migration of the nuclei to specific cellular locations, are coordinated with cellularization by unique types of cell wall formation. Both one mother - two different daughters and the coenocyte-cellularization pathways are used by higher plants in precise spatial and time windows during development. In both the pathways, epigenetic regulation of gene expression is crucial not only for cell fate specification but also for its maintenance through cell lineage. In this review, the focus is on the coenocyte-cellularization pathways in the context of our current understanding of the asymmetric cell divisions. Instances where cell differentiation does not involve an asymmetric division are also discussed to provide a comprehensive account of cell differentiation.
Collapse
Affiliation(s)
- R M Ranganath
- Cytogenetics and Developmental Biology Laboratory, Department of Botany, Bangalore University, India.
| |
Collapse
|
46
|
Lu P, Tuszynski MH. Can bone marrow-derived stem cells differentiate into functional neurons? Exp Neurol 2005; 193:273-8. [PMID: 15869931 DOI: 10.1016/j.expneurol.2005.01.031] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/18/2005] [Accepted: 01/31/2005] [Indexed: 01/09/2023]
Affiliation(s)
- P Lu
- Department of Neurosciences, University of California at San Diego, La Jolla, CA 92093-0626, USA.
| | | |
Collapse
|
47
|
Abstract
Stem cell transdifferentiation in the adult organism is the most common and questioned mechanism of growth and repair. Recent data suggest that adult stem cells are capable of generating mature cells beyond their own tissue boundaries, a process called developmental plasticity. To date, the most versatile cell discovered is the bone marrow progenitor cell.
Collapse
Affiliation(s)
- Annarosa Leri
- Cardiovascular Research Institute, Department of Medicine, New York Medical College, Valhalla, New York.
| | | | | |
Collapse
|
48
|
Neuhuber B, Timothy Himes B, Shumsky JS, Gallo G, Fischer I. Axon growth and recovery of function supported by human bone marrow stromal cells in the injured spinal cord exhibit donor variations. Brain Res 2005; 1035:73-85. [PMID: 15713279 DOI: 10.1016/j.brainres.2004.11.055] [Citation(s) in RCA: 230] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2004] [Indexed: 12/28/2022]
Abstract
Bone marrow stromal cells (MSC) are non-hematopoietic support cells that can be easily derived from bone marrow aspirates. Human MSC are clinically attractive because they can be expanded to large numbers in culture and reintroduced into patients as autografts or allografts. We grafted human MSC derived from aspirates of four different donors into a subtotal cervical hemisection in adult female rats and found that cells integrated well into the injury site, with little migration away from the graft. Immunocytochemical analysis demonstrated robust axonal growth through the grafts of animals treated with MSC, suggesting that MSC support axonal growth after spinal cord injury (SCI). However, the amount of axon growth through the graft site varied considerably between groups of animals treated with different MSC lots, suggesting that efficacy may be donor-dependent. Similarly, a battery of behavioral tests showed partial recovery in some treatment groups but not others. Using ELISA, we found variations in secretion patterns of selected growth factors and cytokines between different MSC lots. In a dorsal root ganglion explant culture system, we tested efficacy of conditioned medium from three donors and found that average axon lengths increased for all groups compared to control. These results suggest that human MSC produce factors important for mediating axon outgrowth and recovery after SCI but that MSC lots from different donors vary considerably. To qualify MSC lots for future clinical application, such notable differences in donor or lot-lot efficacy highlight the need for establishing adequate characterization, including the development of relevant efficacy assays.
Collapse
Affiliation(s)
- Birgit Neuhuber
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | | | | | | | | |
Collapse
|
49
|
Chiu B, Wan JZM, Abley D, Akabutu J. Induction of vascular endothelial phenotype and cellular proliferation from human cord blood stem cells cultured in simulated microgravity. ACTA ASTRONAUTICA 2005; 56:918-922. [PMID: 15835045 DOI: 10.1016/j.actaastro.2005.01.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Recent studies have demonstrated that stem cells derived from adult hematopoietic tissues are capable of trans-differentiation into non-hematopoietic cells, and that the culture in microgravity (microg) may modulate the proliferation and differentiation. We investigated the application of microg to human umbilical cord blood stem cells (CBSC) in the induction of vascular endothelial phenotype expression and cellular proliferation. CD34+ mononuclear cells were isolated from waste human umbilical cord blood samples and cultured in simulated microg for 14 days. The cells were seeded in rotary wall vessels (RWV) with or without microcarrier beads (MCB) and vascular endothelial growth factor was added during culture. Controls consisted of culture in 1 G. The cell cultures in RWV were examined by inverted microscopy. Cell counts, endothelial cell and leukocyte markers performed by flow cytometry and FACS scan were assayed at days 1, 4, 7 and at the termination of the experiments. Culture in RWV revealed significantly increased cellular proliferation with three-dimensional (3D) tissue-like aggregates. At day 4, CD34+ cells cultured in RWV bioreactor without MCB developed vascular tubular assemblies and exhibited endothelial phenotypic markers. These data suggest that CD34+ human umbilical cord blood progenitors are capable of trans-differentiation into vascular endothelial cell phenotype and assemble into 3D tissue structures. Culture of CBSC in simulated microg may be potentially beneficial in the fields of stem cell biology and somatic cell therapy.
Collapse
Affiliation(s)
- Brian Chiu
- Department of Laboratory Medicine and Pathology, University of Alberta Hospitals, Edmonton, Alberta, Canada.
| | | | | | | |
Collapse
|
50
|
Liu Y, Han SSW, Wu Y, Tuohy TMF, Xue H, Cai J, Back SA, Sherman LS, Fischer I, Rao MS. CD44 expression identifies astrocyte-restricted precursor cells. Dev Biol 2004; 276:31-46. [PMID: 15531362 DOI: 10.1016/j.ydbio.2004.08.018] [Citation(s) in RCA: 164] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2004] [Revised: 08/05/2004] [Accepted: 08/13/2004] [Indexed: 11/25/2022]
Abstract
The precise lineage between neural stem cells and mature astrocytes remains poorly defined. To examine astrocyte development, we have characterized glial precursors from neural tissue derived from early embryonic ages. We show that CD44 identifies an astrocyte-restricted precursor cell (ARP) that is committed to generating astrocytes in vitro and in vivo in both rodent and human tissue. CD44+ cells arise later in development than neuronal-restricted precursors (NRPs) or tripotential glial-restricted precursors (GRPs). ARPs are distinguished from GRP and NRP cells by their antigenic profile and differentiation ability. ARPs can be generated from GRP cells in mass or clonal cultures and in vivo after transplantation, suggesting a sequential differentiation of neuroepithelial stem cells (NEPs) to GRPs to ARPs and then to astrocytes. The properties of ARPs are different from other astrocyte precursors described previously in their expression of CD44 and S-100beta and absence of other lineage markers. Using a CD44 misexpression transgenic mouse model (CNP-CD44 mouse), we show that CD44 overexpression in vivo and in vitro decreases the number of mature glia and increases the number of O4+/GFAP+ cells tenfold. Misexpression of CD44 in culture inhibits oligodendrocytes and arrests cells at the precursor state. In summary, our data provide strong evidence for the existence of a CD44+ ARP in the developing nervous system.
Collapse
Affiliation(s)
- Ying Liu
- Laboratory of Neurosciences, Gerontology Research Center, National Institute on Aging, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|