1
|
Alanazi MM, Alsanea S, Kumar A, Alehaideb Z, Matou-Nasri S, AlGhamdi KM. Modulatory effects of oxytocin on normal human cultured melanocyte proliferation, migration, and melanogenesis. Tissue Cell 2024; 91:102579. [PMID: 39388927 DOI: 10.1016/j.tice.2024.102579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/04/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Melanocytes are specialized melanin-producing neural crest-derived cells. Melanocyte proliferation and melanin production (i.e., melanogenesis) are crucial for determining skin color. Disruption of these processes can cause pigmentary skin disorders, including hypo-pigmentary disorders such as vitiligo and hyper-pigmentary disorders such as melasma. Understanding these processes is important for discovering new targets to tackle these skin disorders. Therefore, this study aimed to investigate the effects of oxytocin (OXT) on melanocyte functions. Normal Human Cultured Melanocytes (NHCM) were treated with different OXT doses to investigate OXT effects and mechanisms on NHCM proliferation, migration, and on melanogenesis. OXT significantly increased NHCM proliferation and migration in a dose-dependent manner after 72 h of treatment. In addition, OXT dose-dependently upregulated melanogenesis-related microphtalmia-associated transcription factor, tyrosinase, tyrosinase-related protein (TYRP)-1, and TYRP-2 expression accompanied by an increased trend in melanosome number and maturation stage. Furthermore, OXT at concentrations (62.5-125 nM) increased melanin production. These findings suggest the involvement of OXT receptor (OXTR). In addition, this study demonstrates that OXT stimulates melanocyte proliferation, migration, with a tendency toward melanosome maturation, while it modulates melanin production in a dose-dependent manner. Thus, OXT system including its receptor OXTR may be a potential therapeutic target for skin pigmentary disorders.
Collapse
Affiliation(s)
- Mohammed M Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University (KSU), Riyadh, Saudi Arabia.
| | - Sary Alsanea
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University (KSU), Riyadh, Saudi Arabia
| | - Ashok Kumar
- Vitiligo Research Chair, Department of Dermatology (DOD), College of Medicine (COM), KSU, Riyadh, Saudi Arabia
| | - Zeyad Alehaideb
- Department of Core Medical Research Facility and Platform, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs (MNGHA), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | - Sabine Matou-Nasri
- Blood and Cancer Research Department, KAIMRC, KSAU-HS, MNGHA, Riyadh, Saudi Arabia
| | - Khalid M AlGhamdi
- Vitiligo Research Chair, Department of Dermatology (DOD), College of Medicine (COM), KSU, Riyadh, Saudi Arabia; Department of Dermatology (DOD), College of Medicine (COM), King Saud University (KSU), Riyadh, Saudi Arabia.
| |
Collapse
|
2
|
Keshavarzi F, Salehi MS, Pandamooz S, Zare R, Zamani M, Mostafavi-Pour Z, Pooneh Mokarram P. Valproic acid and/or rapamycin preconditioning protects hair follicle stem cells from oxygen glucose serum deprivation-induced oxidative injury via activating Nrf2 pathway. MOLECULAR BIOLOGY RESEARCH COMMUNICATIONS 2024; 13:103-116. [PMID: 38915453 PMCID: PMC11194030 DOI: 10.22099/mbrc.2024.49302.1922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Among leading causes of the ischemic stroke pathogenesis, oxidative stress strongly declines rate of stem cell engraftment at the injury site, and disables stem cell-based therapy as a key treatment for ischemia stroke. To overcome this therapeutic limitation, preconditioning has been represented a possible approach to augment the adaptation and viability of stem cells to oxidative stress. Here, we illustrated protective impacts of valproic acid (VPA) and/or rapamycin (RAPA) preconditioning unto oxygen glucose and serum deprivation (OGSD)-stimulated cell damage in hair follicle-derived stem cells (HFSCs) and surveyed the plausible inducement mechanisms. OGSD, as an in vitro cell injury model, was established and HFSCs viability was observed using MTT assay after VPA, RAPA, and VPA-RAPA preconditioning under OGSD. ROS and MDA production was assessed to reflect oxidative stress. Real-time PCR and western blotting were employed to investigate Nrf2 expression. The activity of Nrf2-related antioxidant enzymes including NQO1, GPx and GSH level were examined. VEGF and BDNF mRNA expression levels were analyzed. Our results showed that VPA and/or RAPA preconditioning ameliorated OGSD-induced decline in HFSCs viability. In addition, they considerably prohibited ROS and MDA generation in the OGSD-treated HFSCs. Furthermore, VPA and/or RAPA preconditioning stimulated Nrf2 nuclear repositioning and NQO1 and GPx activity and GSH amount, as well as expression of paracrine factors VEGF and BDNF in OGSD-treated HFSCs. Thus, the protective effects afforded by VPA and/or RAPA preconditioning, which involved Nrf2-modulated oxidant stress and regulation of VEGF and BDNF expression, display a simple strategy to augment cell-transplantation efficiency for ischemic stroke.
Collapse
Affiliation(s)
- Fatemeh Keshavarzi
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razieh Zare
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mozhdeh Zamani
- Autophagy Research Center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zohreh Mostafavi-Pour
- Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooneh Pooneh Mokarram
- Autophagy Research Center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
3
|
Karimi-Haghighi S, Pandamooz S, Jurek B, Fattahi S, Safari A, Azarpira N, Dianatpour M, Hooshmandi E, Bayat M, Owjfard M, Zafarmand SS, Mostaghel M, Mousavi SM, Jashire Nezhad N, Eraghi V, Fadakar N, Rahimi Jaberi A, Garcia-Esperon C, Spratt N, Levi C, Salehi MS, Borhani-Haghighi A. From Hair to the Brain: The Short-Term Therapeutic Potential of Human Hair Follicle-Derived Stem Cells and Their Conditioned Medium in a Rat Model of Stroke. Mol Neurobiol 2023; 60:2587-2601. [PMID: 36694047 DOI: 10.1007/s12035-023-03223-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 01/04/2023] [Indexed: 01/26/2023]
Abstract
The short-term therapeutic impacts of stem cells and their derivatives were frequently reported in preclinical investigations of ischemic stroke (IS); however, several drawbacks including accessibility, abundancy, and ethical concerns limited their clinical application. We describe here for the first time the therapeutic potential of human hair follicle-derived stem cells (hHFSCs) and their conditioned medium (CM) in a rat model of IS. Furthermore, we hypothesized that a combination of cell therapy with repeated CM administration might enhance the restorative efficiency of this approach compared to each treatment alone. Middle cerebral artery occlusion was performed for 30 min to induce IS. Immediately after reperfusion, hHFSCs were transplanted through the intra-arterial route and/or hHFSC-CM administered intranasally. The neurological outcomes, short-term spatial working memory, and infarct size were evaluated. Furthermore, relative expression of seven target genes in three categories of neuronal markers, synaptic markers, and angiogenic markers was assessed. The hHFSCs and hHFSC-CM treatments improved neurological impairments and reduced infarct size in the IS rats. Moreover, molecular data elucidated that IS was accompanied by attenuation in the expression of neuronal and synaptic markers in the evaluated brain regions and the interventions rescued these expression changes. Although there was no considerable difference between hHFSCs and hHFSC-CM treatments in the improvement of neurological function and decrement of infarct size, combination therapy was more effective to reduce infarction and elevation of target gene expression especially in the hippocampus. These findings highlight the curative potential of hHFSCs and their CM in a rat model of IS.
Collapse
Affiliation(s)
| | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benjamin Jurek
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
| | | | - Anahid Safari
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Etrat Hooshmandi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahnaz Bayat
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mandana Mostaghel
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Nahid Jashire Nezhad
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vida Eraghi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nima Fadakar
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Rahimi Jaberi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Carlos Garcia-Esperon
- Department of Neurology, John Hunter Hospital, Newcastle, Australia
- Hunter Medical Research Institute and the University of Newcastle, Newcastle, Australia
| | - Neil Spratt
- Department of Neurology, John Hunter Hospital, Newcastle, Australia
- Hunter Medical Research Institute and the University of Newcastle, Newcastle, Australia
| | - Christopher Levi
- Department of Neurology, John Hunter Hospital, Newcastle, Australia
- Hunter Medical Research Institute and the University of Newcastle, Newcastle, Australia
| | - Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Afshin Borhani-Haghighi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Neurology, John Hunter Hospital, Newcastle, Australia.
- Hunter Medical Research Institute and the University of Newcastle, Newcastle, Australia.
| |
Collapse
|
4
|
Mousavi SM, Akbarpour B, Karimi-Haghighi S, Pandamooz S, Belém-Filho IJA, Masís-Calvo M, Salimi H, Lashanizadegan R, Pouramini A, Owjfard M, Hooshmandi E, Bayat M, Zafarmand SS, Dianatpour M, Salehi MS, Borhani-Haghighi A. Therapeutic potential of hair follicle-derived stem cell intranasal transplantation in a rat model of ischemic stroke. BMC Neurosci 2022; 23:47. [PMID: 35879657 PMCID: PMC9316709 DOI: 10.1186/s12868-022-00732-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 07/15/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Stem cell-based therapy has received considerable attention as a potential candidate in the treatment of ischemic stroke; however, employing an appropriate type of stem cells and an effective delivery route are still challenging. In the present study, we investigated the therapeutic effect of safe, noninvasive, and brain-targeted intranasal administration of hair follicle-derived stem cells (HFSCs) in a rat model of ischemic stroke. METHODS Stem cells were obtained from the adult rat hair follicles. In experiment 1, stroke was induced by 30 min middle cerebral artery occlusion (MCAO) and stem cells were intranasally transplanted immediately after ischemia. In experiment 2, stroke was induced by 120 min MCAO and stem cells were administered 24 h after cerebral ischemia. In all experimental groups, neurological performance, short-term spatial working memory and infarct volume were assessed. Moreover, relative expression of major trophic factors in the striatum and cortex was evaluated by the quantitative PCR technique. The end point of experiment 1 was day 3 and the end point of experiment 2 was day 15. RESULTS In both experiments, intranasal administration of HFSCs improved functional performance and decreased infarct volume compared to the MCAO rats. Furthermore, NeuN and VEGF expression were higher in the transplanted group and stem cell therapy partially prevented BDNF and neurotrophin-3 over-expression induced by cerebral ischemia. CONCLUSIONS These findings highlight the curative potential of HFSCs following intranasal transplantation in a rat model of ischemic stroke.
Collapse
Affiliation(s)
- Seyedeh Maryam Mousavi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bijan Akbarpour
- Department of Basic Sciences, Faculty of Veterinary Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran.
| | | | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Haniye Salimi
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ramin Lashanizadegan
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Pouramini
- Department of Basic Sciences, Faculty of Veterinary Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Etrat Hooshmandi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahnaz Bayat
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mehdi Dianatpour
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | | |
Collapse
|
5
|
Mokabber H, Vatankhah MA, Najafzadeh N. The regulatory role of microRNAs in the development, cyclic changes, and cell differentiation of the hair follicle. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.01.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
6
|
Solis-Castro OO, Rivolta MN, Boissonade FM. Neural Crest-Derived Stem Cells (NCSCs) Obtained from Dental-Related Stem Cells (DRSCs): A Literature Review on Current Knowledge and Directions toward Translational Applications. Int J Mol Sci 2022; 23:ijms23052714. [PMID: 35269856 PMCID: PMC8911272 DOI: 10.3390/ijms23052714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/21/2022] [Accepted: 02/27/2022] [Indexed: 11/21/2022] Open
Abstract
Evidence from dental-related stem cells (DRSCs) suggests an enhanced potential for ectodermal lineage differentiation due to their neural crest origin. Growing evidence that DRSC cultures can produce cells with a neural crest-derived stem cell (NCSC)-like phenotype supports their potential for future therapeutic approaches for neurodegenerative diseases and nerve injuries. However, most of the evidence is limited to the characterization of DRSCs as NCSCs by detecting the expression of neural crest markers. Only a few studies have provided proof of concept of an improved neuro-glial differentiation or direct applicability in relevant models. In addition, a current problem is that several of the existing protocols do not meet manufacturing standards for transferability to a clinical scenario. This review describes the current protocols to obtain NCSCs from DRSCs and their characterization. Also, it provides important considerations from previous work where DRSCs were established and characterized as mesenchymal stromal cells but studied for their neuro-glial differentiation potential. The therapeutic advancement of DRSCs would depend on establishing protocols that can yield a neural crest-like phenotype efficiently, using appropriate manufacturing standards and testing them in relevant models of disease or injury. Achieving these conditions could then facilitate and validate the therapeutic potential of DRSC-NCSCs in regenerative therapies.
Collapse
Affiliation(s)
- Oscar O. Solis-Castro
- School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK;
- The Neuroscience Institute, University of Sheffield, Sheffield S10 2TN, UK;
| | - Marcelo N. Rivolta
- The Neuroscience Institute, University of Sheffield, Sheffield S10 2TN, UK;
- Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Fiona M. Boissonade
- School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK;
- The Neuroscience Institute, University of Sheffield, Sheffield S10 2TN, UK;
- Correspondence:
| |
Collapse
|
7
|
Abreu CM, Marques AP. Recreation of a hair follicle regenerative microenvironment: Successes and pitfalls. Bioeng Transl Med 2022; 7:e10235. [PMID: 35079623 PMCID: PMC8780054 DOI: 10.1002/btm2.10235] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/15/2021] [Accepted: 05/18/2021] [Indexed: 12/19/2022] Open
Abstract
The hair follicle (HF) is an exquisite skin appendage endowed with cyclical regenerative capacity; however, de novo follicle formation does not naturally occur. Consequently, patients suffering from extensive skin damage or hair loss are deprived of the HF critical physiological and/or aesthetic functions, severally compromising skin function and the individual's psychosocial well-being. Translation of regenerative strategies has been prevented by the loss of trichogenic capacity that relevant cell populations undergo in culture and by the lack of suitable human-based in vitro testing platforms. Here, we provide a comprehensive overview of the major difficulties associated with HF regeneration and the approaches used to overcome these drawbacks. We describe key cellular requirements and discuss the importance of the HF extracellular matrix and associated signaling for HF regeneration. Finally, we summarize the strategies proposed so far to bioengineer human HF or hair-bearing skin models and disclose future trends for the field.
Collapse
Affiliation(s)
- Carla M. Abreu
- 3B's Research Group, I3Bs ‐ Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAvePark–Parque de Ciência e Tecnologia, University of MinhoGuimarãesPortugal
- ICVS/3B's–PT Government Associate LaboratoryGuimarãesPortugal
| | - Alexandra P. Marques
- 3B's Research Group, I3Bs ‐ Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAvePark–Parque de Ciência e Tecnologia, University of MinhoGuimarãesPortugal
- ICVS/3B's–PT Government Associate LaboratoryGuimarãesPortugal
| |
Collapse
|
8
|
Ahmed MM, Cushman CH, DeCaprio JA. Merkel Cell Polyomavirus: Oncogenesis in a Stable Genome. Viruses 2021; 14:v14010058. [PMID: 35062263 PMCID: PMC8781562 DOI: 10.3390/v14010058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 12/14/2022] Open
Abstract
Merkel cell polyomavirus (MCV) is the causative agent for the majority of Merkel cell carcinoma (MCC) cases. Polyomavirus-associated MCC (MCCP) is characterized by the integration of MCV DNA into the tumor genome and a low tumor mutational burden. In contrast, nonviral MCC (MCCN) is characterized by a high tumor mutational burden induced by UV damage. Since the discovery of MCV, much work in the field has focused on understanding the molecular mechanisms of oncogenesis driven by the MCV tumor (T) antigens. Here, we review our current understanding of how the activities of large T (LT) and small T (ST) promote MCC oncogenesis in the absence of genomic instability. We highlight how both LT and ST inhibit tumor suppressors to evade growth suppression, an important cancer hallmark. We discuss ST interactions with cellular proteins, with an emphasis on those that contribute to sustaining proliferative signaling. Finally, we examine active areas of research into open questions in the field, including the origin of MCC and mechanisms of viral integration.
Collapse
Affiliation(s)
- Mona M. Ahmed
- Program in Virology, Graduate School of Arts and Sciences, Harvard University, Cambridge, MA 02138, USA; (M.M.A.); (C.H.C.)
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Camille H. Cushman
- Program in Virology, Graduate School of Arts and Sciences, Harvard University, Cambridge, MA 02138, USA; (M.M.A.); (C.H.C.)
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - James A. DeCaprio
- Program in Virology, Graduate School of Arts and Sciences, Harvard University, Cambridge, MA 02138, USA; (M.M.A.); (C.H.C.)
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Correspondence:
| |
Collapse
|
9
|
Windmöller BA, Höving AL, Knabbe C, Greiner JFW. Inter- and Intrapopulational Heterogeneity of Characteristic Markers in Adult Human Neural Crest-derived Stem Cells. Stem Cell Rev Rep 2021; 18:1510-1520. [PMID: 34748196 PMCID: PMC9033708 DOI: 10.1007/s12015-021-10277-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 11/24/2022]
Abstract
Adult human neural crest-derived stem cells (NCSCs) are found in a variety of adult tissues and show an extraordinary broad developmental potential. Despite their great differentiation capacity, increasing evidence suggest a remaining niche-dependent variability between different NCSC-populations regarding their differentiation behavior and expression signatures. In the present study, we extended the view on heterogeneity of NCSCs by identifying heterogeneous expression levels and protein amounts of characteristic markers even between NCSCs from the same niche of origin. In particular, populations of neural crest-derived inferior turbinate stem cells (ITSCs) isolated from different individuals showed significant variations in characteristic NCSC marker proteins Nestin, S100 and Slug in a donor-dependent manner. Notably, increased nuclear protein amounts of Slug were accompanied by a significantly elevated level of nuclear NF-κB-p65 protein, suggesting an NF-κB-dependent regulation of NCSC-makers. In addition to this interpopulational genetic heterogeneity of ITSC-populations from different donors, single ITSCs also revealed a strong heterogeneity regarding the protein amounts of Nestin, S100, Slug and NF-κB-p65 even within the same clonal culture. Our present findings therefor strongly suggest ITSC-heterogeneity to be at least partly based on an interpopulational genetic heterogeneity dependent on the donor accompanied by a stochastic intrapopulational heterogeneity between single cells. We propose this stochastic intrapopulational heterogeneity to occur in addition to the already described genetic variability between clonal NCSC-cultures and the niche-dependent plasticity of NCSCs. Our observations offer a novel perspective on NCSC-heterogeneity, which may build the basis to understand heterogeneous NCSC-behavior.
Collapse
Affiliation(s)
- Beatrice A Windmöller
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany.,Forschungsverbund BioMedizin Bielefeld FBMB e.V, Bielefeld, Germany.,Department of Cellular Neurophysiology, Faculty of Medicine, University of Bielefeld, Bielefeld, Germany
| | - Anna L Höving
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany.,Institute for Laboratory and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr-University Bochum, 32545, Bad Oeynhausen, Germany
| | - Cornelius Knabbe
- Forschungsverbund BioMedizin Bielefeld FBMB e.V, Bielefeld, Germany.,Institute for Laboratory and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr-University Bochum, 32545, Bad Oeynhausen, Germany
| | - Johannes F W Greiner
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany. .,Forschungsverbund BioMedizin Bielefeld FBMB e.V, Bielefeld, Germany.
| |
Collapse
|
10
|
Kaltschmidt C, Greiner JFW, Kaltschmidt B. The Transcription Factor NF-κB in Stem Cells and Development. Cells 2021; 10:2042. [PMID: 34440811 PMCID: PMC8391683 DOI: 10.3390/cells10082042] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/26/2022] Open
Abstract
NF-κB (nuclear factor kappa B) belongs to a family of transcription factors known to regulate a broad range of processes such as immune cell function, proliferation and cancer, neuroprotection, and long-term memory. Upcoming fields of NF-κB research include its role in stem cells and developmental processes. In the present review, we discuss one role of NF-κB in development in Drosophila, Xenopus, mice, and humans in accordance with the concept of evo-devo (evolutionary developmental biology). REL domain-containing proteins of the NF-κB family are evolutionarily conserved among these species. In addition, we summarize cellular phenotypes such as defective B- and T-cell compartments related to genetic NF-κB defects detected among different species. While NF-κB proteins are present in nearly all differentiated cell types, mouse and human embryonic stem cells do not contain NF-κB proteins, potentially due to miRNA-dependent inhibition. However, the mesodermal and neuroectodermal differentiation of mouse and human embryonic stem cells is hampered upon the repression of NF-κB. We further discuss NF-κB as a crucial regulator of differentiation in adult stem cells such as neural crest-derived and mesenchymal stem cells. In particular, c-REL seems to be important for neuronal differentiation and the neuroprotection of human adult stem cells, while RELA plays a crucial role in osteogenic and mesodermal differentiation.
Collapse
Affiliation(s)
- Christian Kaltschmidt
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (C.K.); (J.F.W.G.)
| | - Johannes F. W. Greiner
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (C.K.); (J.F.W.G.)
| | - Barbara Kaltschmidt
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (C.K.); (J.F.W.G.)
- Molecular Neurobiology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| |
Collapse
|
11
|
Khodabakhsh P, Pournajaf S, Mohaghegh Shalmani L, Ahmadiani A, Dargahi L. Insulin Promotes Schwann-Like Cell Differentiation of Rat Epidermal Neural Crest Stem Cells. Mol Neurobiol 2021; 58:5327-5337. [PMID: 34297315 DOI: 10.1007/s12035-021-02423-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 05/05/2021] [Indexed: 10/20/2022]
Abstract
Schwann cells (SCs) are considered potentially attractive candidates for transplantation therapies in neurodegenerative diseases. However, problems arising from the isolation and expansion of the SCs restrict their clinical applications. Establishing an alternative Schwann-like cell type is a prerequisite. Epidermal neural crest stem cells (EPI-NCSCs) are well studied for their autologous accessibility, along with the ability to produce major neural crest derivatives and neurotrophic factors. In the current study, we explored insulin influence, a well-known growth factor, on directing EPI-NCSCs into the Schwann cell (SC) lineage. EPI-NCSCs were isolated from rat hair bulge explants. The viability of cells treated with a range of insulin concentrations (0.05-100 μg/ml) was defined by MTT assay at 24, 48, and 72 h. The gene expression profiles of neurotrophic factors (BDNF, FGF-2, and IL-6), key regulators involved in the development of SC (EGR-1, SOX-10, c-JUN, GFAP, OCT-6, EGR-2, and MBP), and oligodendrocyte (PDGFR-α and NG-2) were quantified 1 and 9 days post-treatment with 0.05 and 5 μg/ml insulin. Furthermore, the protein expression of nestin (stemness marker), SOX-10, PDGFR-α, and MBP was analyzed following the long-term insulin treatment. Insulin downregulated the early-stage SC differentiation marker (EGR-1) and increased neurotrophins (BDNF and IL-6) and pro-myelinating genes, including OCT-6, SOX-10, EGR-2, and MBP, as well as oligodendrocyte differentiation markers, upon exposure for 9 days. Insulin can promote EPI-NCSC differentiation toward SC lineage and possibly oligodendrocytes. Thus, employing insulin might enhance the EPI-NCSCs efficiency in cell transplantation strategies.
Collapse
Affiliation(s)
- Pariya Khodabakhsh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Safura Pournajaf
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Mohaghegh Shalmani
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
The Implementation of Preconditioned Epidermal Neural Crest Stem Cells to Combat Ischemic Stroke. Comment on Othman, F.A.; Tan, S.C. Preconditioning Strategies to Enhance Neural Stem Cell-Based Therapy for Ischemic Stroke. Brain Sci. 2020, 10, 893. Brain Sci 2021; 11:brainsci11050653. [PMID: 34067592 PMCID: PMC8155980 DOI: 10.3390/brainsci11050653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/07/2021] [Accepted: 05/13/2021] [Indexed: 12/24/2022] Open
Abstract
In the recent review published in Brain Sciences, Othman and Tan suggested several preconditioning strategies to improve stem cell therapy after ischemic brain injury [...].
Collapse
|
13
|
Höving AL, Windmöller BA, Knabbe C, Kaltschmidt B, Kaltschmidt C, Greiner JFW. Between Fate Choice and Self-Renewal-Heterogeneity of Adult Neural Crest-Derived Stem Cells. Front Cell Dev Biol 2021; 9:662754. [PMID: 33898464 PMCID: PMC8060484 DOI: 10.3389/fcell.2021.662754] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022] Open
Abstract
Stem cells of the neural crest (NC) vitally participate to embryonic development, but also remain in distinct niches as quiescent neural crest-derived stem cell (NCSC) pools into adulthood. Although NCSC-populations share a high capacity for self-renewal and differentiation resulting in promising preclinical applications within the last two decades, inter- and intrapopulational differences exist in terms of their expression signatures and regenerative capability. Differentiation and self-renewal of stem cells in developmental and regenerative contexts are partially regulated by the niche or culture condition and further influenced by single cell decision processes, making cell-to-cell variation and heterogeneity critical for understanding adult stem cell populations. The present review summarizes current knowledge of the cellular heterogeneity within NCSC-populations located in distinct craniofacial and trunk niches including the nasal cavity, olfactory bulb, oral tissues or skin. We shed light on the impact of intrapopulational heterogeneity on fate specifications and plasticity of NCSCs in their niches in vivo as well as during in vitro culture. We further discuss underlying molecular regulators determining fate specifications of NCSCs, suggesting a regulatory network including NF-κB and NC-related transcription factors like SLUG and SOX9 accompanied by Wnt- and MAPK-signaling to orchestrate NCSC stemness and differentiation. In summary, adult NCSCs show a broad heterogeneity on the level of the donor and the donors' sex, the cell population and the single stem cell directly impacting their differentiation capability and fate choices in vivo and in vitro. The findings discussed here emphasize heterogeneity of NCSCs as a crucial parameter for understanding their role in tissue homeostasis and regeneration and for improving their applicability in regenerative medicine.
Collapse
Affiliation(s)
- Anna L. Höving
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Institute for Laboratory- and Transfusion Medicine, Heart and Diabetes Centre North Rhine-Westphalia (NRW), Ruhr University Bochum, Bad Oeynhausen, Germany
| | - Beatrice A. Windmöller
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
| | - Cornelius Knabbe
- Institute for Laboratory- and Transfusion Medicine, Heart and Diabetes Centre North Rhine-Westphalia (NRW), Ruhr University Bochum, Bad Oeynhausen, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
| | - Barbara Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
| | - Johannes F. W. Greiner
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Bielefeld, Germany
| |
Collapse
|
14
|
Vapniarsky N, Kwon H, Paschos NK, Haudenschild AK, Brown WE, DuRaine GD, Hu JC, Athanasiou KA. Adult Dermal Stem Cells for Scaffold-Free Cartilage Tissue Engineering: Exploration of Strategies. Tissue Eng Part C Methods 2020; 26:598-607. [PMID: 33198584 DOI: 10.1089/ten.tec.2020.0207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Dermis-isolated adult stem (DIAS) cells, abundantly available, are attractive for regenerative medicine. Strategies have been devised to isolate and to chondroinduce DIAS cells from various animals. This study aimed to characterize DIAS cells from human abdominal skin (human dermis-isolated adult stem [hDIAS] cells) and to compare and to refine various chondroinduction regimens to form functional neocartilage constructs. The stemness of hDIAS cells was verified (Phase I), three chondroinduction pretreatments were compared (Phase II), and, from these, one regimen was carried forward for refinement in Phase III for improving the mechanical properties of hDIAS cell-derived constructs. Multilineage differentiation and mesenchymal stem cell markers were observed. Among various chondroinduction pretreatments, the nodule formation pretreatment yielded constructs at least 72% larger in diameter, with higher glycosaminoglycan (GAG) content by 44%, compared with other pretreatments. Furthermore, it was found that culturing cells on nontissue culture-treated surfaces yielded constructs (1) on par with constructs derived from aggrecan-coated surfaces and (2) with superior mechanical properties than constructs derived from cells cultured on tissue culture-treated surfaces. After the nodule formation pretreatment, combined supplementation of TGF-β1, IGF-I, and fetal bovine serum significantly enhanced aggregate modulus and shear modulus by 75% and 69%, respectively, over the supplementation by TGF-β1 alone. In summary, human skin-derived DIAS cells are responsive to chondroinduction for forming neocartilage. Furthermore, the mechanical properties of the resultant human constructs can be improved by treatments shown to be efficacious in animal models. Advances made toward tissue-engineering cartilage using animal cells were shown to be applicable to hDIAS cells for cartilage repair and regeneration.
Collapse
Affiliation(s)
- Natalia Vapniarsky
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, Davis, California, USA
| | - Heenam Kwon
- Department of Biomedical Engineering, Samueli School of Engineering, University of California, Irvine, Irvine, California, USA
| | - Nikolaos K Paschos
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | - Anne K Haudenschild
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | - Wendy E Brown
- Department of Biomedical Engineering, Samueli School of Engineering, University of California, Irvine, Irvine, California, USA
| | - Grayson D DuRaine
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, Samueli School of Engineering, University of California, Irvine, Irvine, California, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, Samueli School of Engineering, University of California, Irvine, Irvine, California, USA
| |
Collapse
|
15
|
Höving AL, Sielemann K, Greiner JFW, Kaltschmidt B, Knabbe C, Kaltschmidt C. Transcriptome Analysis Reveals High Similarities between Adult Human Cardiac Stem Cells and Neural Crest-Derived Stem Cells. BIOLOGY 2020; 9:biology9120435. [PMID: 33271866 PMCID: PMC7761507 DOI: 10.3390/biology9120435] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022]
Abstract
For the identification of a stem cell population, the comparison of transcriptome data enables the simultaneous analysis of tens of thousands of molecular markers and thus enables the precise distinction of even closely related populations. Here, we utilized global gene expression profiling to compare two adult human stem cell populations, namely neural crest-derived inferior turbinate stem cells (ITSCs) of the nasal cavity and human cardiac stem cells (hCSCs) from the heart auricle. We detected high similarities between the transcriptomes of both stem cell populations, particularly including a range of neural crest-associated genes. However, global gene expression likewise reflected differences between the stem cell populations with regard to their niches of origin. In a broader analysis, we further identified clear similarities between ITSCs, hCSCs and other adherent stem cell populations compared to non-adherent hematopoietic progenitor cells. In summary, our observations reveal high similarities between adult human cardiac stem cells and neural crest-derived stem cells from the nasal cavity, which include a shared relation to the neural crest. The analyses provided here may help to understand underlying molecular regulators determining differences between adult human stem cell populations.
Collapse
Affiliation(s)
- Anna L. Höving
- Department of Cell Biology, Bielefeld University, 33615 Bielefeld, Germany; (J.F.W.G.); (B.K.)
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany;
- Correspondence: (A.L.H.); (C.K.)
| | - Katharina Sielemann
- Genetics and Genomics of Plants, Center for Biotechnology (CeBiTec), Bielefeld University, 33615 Bielefeld, Germany;
- Graduate School DILS, Bielefeld Institute for Bioinformatics Infrastructure (BIBI), Bielefeld University, 33615 Bielefeld, Germany
| | - Johannes F. W. Greiner
- Department of Cell Biology, Bielefeld University, 33615 Bielefeld, Germany; (J.F.W.G.); (B.K.)
| | - Barbara Kaltschmidt
- Department of Cell Biology, Bielefeld University, 33615 Bielefeld, Germany; (J.F.W.G.); (B.K.)
- AG Molecular Neurobiology, Bielefeld University, 33615 Bielefeld, Germany
| | - Cornelius Knabbe
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany;
| | - Christian Kaltschmidt
- Department of Cell Biology, Bielefeld University, 33615 Bielefeld, Germany; (J.F.W.G.); (B.K.)
- Correspondence: (A.L.H.); (C.K.)
| |
Collapse
|
16
|
Solis‐Castro OO, Boissonade FM, Rivolta MN. Establishment and neural differentiation of neural crest-derived stem cells from human dental pulp in serum-free conditions. Stem Cells Transl Med 2020; 9:1462-1476. [PMID: 32633468 PMCID: PMC7581455 DOI: 10.1002/sctm.20-0037] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/11/2020] [Accepted: 06/04/2020] [Indexed: 12/30/2022] Open
Abstract
The potential of obtaining cell cultures with neural crest resemblance (neural crest-derived stem cells [NCSCs]) from dental-related tissues, including human dental pulp cells (hDPCs), has been discussed in the literature. However, most reports include the use of serum-rich conditions and do not describe the potential for neural differentiation, slowing translation to the clinic. Therefore, we aimed to culture and characterize NCSCs from the human dental pulp in vitro and evaluate their ability to differentiate into neurons; we also investigated the effectiveness of the addition of BMP4 to enhance this potential. Cultures were established from a varied cohort of patient samples and grown, as monolayers, in serum, serum-free, and also under sphere-aggregation conditions to induce and identify a NCSC phenotype. hDPC cultures were characterized by immunocytochemistry and reverse transcription quantitative polymerase chain reaction. Monolayer cultures expressed stem cell, neural progenitor and neural crest-related markers. Culturing hDPCs as neurospheres (hDPC-NCSCs) resulted in an increased expression of neural crest-related genes, while the addition of BMP4 appeared to produce better NCSC characteristics and neural differentiation. The neural-like phenotype was evidenced by the expression of TUJ1, peripherin, NFH, TAU, SYN1, and GAP43. Our results describe the establishment of hDPC cultures from a large variety of patients in serum-free medium, as NCSC that differentiate into neural-like cells, as well as an important effect of BMP4 in enhancing the neural crest phenotype and differentiation of hDPCs.
Collapse
Affiliation(s)
- Oscar O. Solis‐Castro
- Centre for Stem Cell Biology, Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
- School of Clinical DentistryUniversity of SheffieldSheffieldUK
- The Neuroscience InstituteUniversity of SheffieldSheffieldUK
| | - Fiona M. Boissonade
- School of Clinical DentistryUniversity of SheffieldSheffieldUK
- The Neuroscience InstituteUniversity of SheffieldSheffieldUK
| | - Marcelo N. Rivolta
- Centre for Stem Cell Biology, Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
- The Neuroscience InstituteUniversity of SheffieldSheffieldUK
| |
Collapse
|
17
|
Hopf A, Schaefer DJ, Kalbermatten DF, Guzman R, Madduri S. Schwann Cell-Like Cells: Origin and Usability for Repair and Regeneration of the Peripheral and Central Nervous System. Cells 2020; 9:E1990. [PMID: 32872454 PMCID: PMC7565191 DOI: 10.3390/cells9091990] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/06/2020] [Accepted: 08/22/2020] [Indexed: 12/14/2022] Open
Abstract
Functional recovery after neurotmesis, a complete transection of the nerve fiber, is often poor and requires a surgical procedure. Especially for longer gaps (>3 mm), end-to-end suturing of the proximal to the distal part is not possible, thus requiring nerve graft implantation. Artificial nerve grafts, i.e., hollow fibers, hydrogels, chitosan, collagen conduits, and decellularized scaffolds hold promise provided that these structures are populated with Schwann cells (SC) that are widely accepted to promote peripheral and spinal cord regeneration. However, these cells must be collected from the healthy peripheral nerves, resulting in significant time delay for treatment and undesired morbidities for the donors. Therefore, there is a clear need to explore the viable source of cells with a regenerative potential similar to SC. For this, we analyzed the literature for the generation of Schwann cell-like cells (SCLC) from stem cells of different origins (i.e., mesenchymal stem cells, pluripotent stem cells, and genetically programmed somatic cells) and compared their biological performance to promote axonal regeneration. Thus, the present review accounts for current developments in the field of SCLC differentiation, their applications in peripheral and central nervous system injury, and provides insights for future strategies.
Collapse
Affiliation(s)
- Alois Hopf
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland; (A.H.); (D.F.K.)
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
| | - Dirk J. Schaefer
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Daniel F. Kalbermatten
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland; (A.H.); (D.F.K.)
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Raphael Guzman
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
- Department of Neurosurgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Srinivas Madduri
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland; (A.H.); (D.F.K.)
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| |
Collapse
|
18
|
Pournajaf S, Valian N, Mohaghegh Shalmani L, Khodabakhsh P, Jorjani M, Dargahi L. Fingolimod increases oligodendrocytes markers expression in epidermal neural crest stem cells. Eur J Pharmacol 2020; 885:173502. [PMID: 32860811 DOI: 10.1016/j.ejphar.2020.173502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/19/2020] [Accepted: 08/23/2020] [Indexed: 12/11/2022]
Abstract
Epidermal neural crest stem cells (EPI-NCSCs) are propitious candidates for cell replacement therapy and supplying neurotrophic factors in the neurological disorders. Considering the potential remyelinating and regenerative effects of fingolimod, in this study, we evaluated its effects on EPI-NCSCs viability and the expression of neurotrophic and oligodendrocyte differentiation factors. EPI-NCSCs, extracted from the bulge of rat hair follicles, were characterized and treated with fingolimod (0, 50, 100, 200, 400, 600, 1000, and 5000 nM). The cell viability was evaluated by MTT assay at 6, 24 and 72 h. The expression of neurotrophic and differentiation factors in the cells treated with 100 and 400 nM fingolimod were measured at 24 and 120 h. Fingolimod at 50-600 nM increased the cells viability after 6 h, with no change at the higher concentrations. The highest concentration (5000nM) induced toxicity at 24 and 72 h. NGF and GDNF genes expression were decreased at 120 h, but on the contrary, brain derived neurotrophic factor (BDNF) and neurotrophin 3 (NT3) were increased by both concentrations at both time points. Oligodendrocyte markers including platelet-derived growth factor receptor A (PDGFRα), neuron-glial antigen 2 (NG2) and growth associated protein 43 (GAP43) were elevated at 120 h, which was accompanied with reduce in stemness markers (Nestin and early growth response 1 (EGR1)). Fingolimod increased the expression of neurotrophic factors in EPI-NCSCs, and guided them to oligodendrocyte fate. Therefore, fingolimod in combination with EPI-NCSCs, can be considered as a promising approach for demyelinating neurological disorders.
Collapse
Affiliation(s)
- Safura Pournajaf
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Valian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Mohaghegh Shalmani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pariya Khodabakhsh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Jorjani
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Li X, Kraft P, De Vivo I, Giovannucci E, Liang L, Nan H. Height, nevus count, and risk of cutaneous malignant melanoma: Results from 2 large cohorts of US women. J Am Acad Dermatol 2020; 83:1049-1056. [PMID: 32376423 DOI: 10.1016/j.jaad.2020.04.158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 04/08/2020] [Accepted: 04/28/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Taller individuals are at higher risk of melanoma. OBJECTIVE To prospectively investigate the association of height with nevus count and melanoma and estimate the proportion of height-melanoma association explained by nevus count among white participants from the Nurses' Health Study (NHS) and Nurses' Health Study 2 (NHS2). METHODS We used Cox proportional hazards regression and multinomial logistic regression for data analyses, with adjustment of potential confounders in the multivariate model. RESULTS We included 82,468 and 106,069 women from NHS and NHS2, respectively. The hazard ratio was 1.21 (95% confidence interval [CI] 1.12-1.31) for the association between every 10-cm increase in height and melanoma. Compared with women with no nevi, the odds ratios (95% CIs) associated with a 10-cm increase in height were 1.35 (95% CI 1.23-1.48) in the NHS and 1.12 (95% CI 1.09-1.15) in the NHS2 for women with greater than or equal to 10 moles. The proportion of excess melanoma risk associated with each 10-cm increase in height explained by nevus count was 8.03% in the NHS and 10.22% in the NHS2. LIMITATION Self-reported height and nevus count. Mole counts were limited to 1 arm or both legs. CONCLUSION Nevus count is an important explanatory factor for the excess risk of melanoma among taller white women.
Collapse
Affiliation(s)
- Xin Li
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Immaculata De Vivo
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Edward Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Hongmei Nan
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana; Department of Global Health, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana.
| |
Collapse
|
20
|
Salehi MS, Pandamooz S, Safari A, Jurek B, Tamadon A, Namavar MR, Dianatpour M, Dargahi L, Azarpira N, Fattahi S, Shid Moosavi SM, Keshavarz S, Khodabandeh Z, Zare S, Nazari S, Heidari M, Izadi S, Poursadeghfard M, Borhani-Haghighi A. Epidermal neural crest stem cell transplantation as a promising therapeutic strategy for ischemic stroke. CNS Neurosci Ther 2020; 26:670-681. [PMID: 32281225 PMCID: PMC7298983 DOI: 10.1111/cns.13370] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/10/2020] [Accepted: 03/13/2020] [Indexed: 12/22/2022] Open
Abstract
Introduction Cell‐based therapy is considered as promising strategy to cure stroke. However, employing appropriate type of stem cell to fulfill many therapeutic needs of cerebral ischemia is still challenging. In this regard, the current study was designed to elucidate therapeutic potential of epidermal neural crest stem cells (EPI‐NCSCs) compared to bone marrow mesenchymal stem cells (BM‐MSCs) in rat model of ischemic stroke. Methods Ischemic stroke was induced by middle cerebral artery occlusion (MCAO) for 45 minutes. Immediately after reperfusion, EPI‐NCSCs or BM‐MSCs were transplanted via intra‐arterial or intravenous route. A test for neurological function was performed before ischemia and 1, 3, and 7 days after MCAO. Also, infarct volume ratio and relative expression of 15 selected target genes were evaluated 7 days after transplantation. Results EPI‐NCSCs transplantation (both intra‐arterial and intravenous) and BM‐MSCs transplantation (only intra‐arterial) tended to result in a better functional outcome, compared to the MCAO group; however, this difference was not statistically significant. The infarct volume ratio significantly decreased in NCSC‐intra‐arterial, NCSC‐intravenous and MSC‐intra‐arterial groups compared to the control. EPI‐NCSCs interventions led to higher expression levels of Bdnf, nestin, Sox10, doublecortin, β‐III tubulin, Gfap, and interleukin‐6, whereas neurotrophin‐3 and interleukin‐10 were decreased. On the other hand, BM‐MSCs therapy resulted in upregulation of Gdnf, β‐III tubulin, and Gfap and down‐regulation of neurotrophin‐3, interleukin‐1, and interleukin‐10. Conclusion These findings highlight the therapeutic effects of EPI‐NCSCs transplantation, probably through simultaneous induction of neuronal and glial formation, as well as Bdnf over‐expression in a rat model of ischemic stroke.
Collapse
Affiliation(s)
- Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sareh Pandamooz
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anahid Safari
- Stem cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benjamin Jurek
- Department of Behavioral and Molecular Neurobiology, Faculty of Biology and Preclinical Medicine, University of Regensburg, Regensburg, Germany
| | - Amin Tamadon
- The Persian Gulf Marine Biotechnology Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammad Reza Namavar
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Stem cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sadegh Fattahi
- Cellular & Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | - Somaye Keshavarz
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Khodabandeh
- Stem cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahrokh Zare
- Stem cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Nazari
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojdeh Heidari
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sadegh Izadi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Poursadeghfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
21
|
Pandamooz S, Jafari A, Salehi MS, Jurek B, Ahmadiani A, Safari A, Hassanajili S, Borhani-Haghighi A, Dianatpour M, Niknejad H, Azarpira N, Dargahi L. Substrate stiffness affects the morphology and gene expression of epidermal neural crest stem cells in a short term culture. Biotechnol Bioeng 2019; 117:305-317. [PMID: 31654402 DOI: 10.1002/bit.27208] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 12/20/2022]
Abstract
According to the intrinsic plasticity of stem cells, controlling their fate is a critical issue in cell-based therapies. Recently, a growing body of evidence has suggested that substrate stiffness can affect the fate decisions of various stem cells. Epidermal neural crest stem cells as one of the main neural crest cell derivatives hold great promise for cell therapies due to presenting a high level of plasticity. This study was conducted to define the influence of substrate stiffness on the lineage commitment of these cells. Here, four different polyacrylamide hydrogels with elastic modulus in the range of 0.7-30 kPa were synthesized and coated with collagen and stem cells were seeded on them for 24 hr. The obtained data showed that cells can attach faster to hydrogels compared with culture plate and cells on <1 kPa stiffness show more neuronal-like morphology as they presented several branches and extended longer neurites over time. Moreover, the transcription of actin downregulated on all hydrogels, while the expression of Nestin, Tubulin, and PDGFR-α increased on all of them and SOX-10 and doublecortin gene expression were higher only on <1 kPa. Also, it was revealed that soft hydrogels can enhance the expression of glial cell line-derived neurotrophic factor, neurotrophin-3, and vascular endothelial growth factor in these stem cells. On the basis of the results, these cells can respond to the substrate stiffness in the short term culture and soft hydrogels can alter their morphology and gene expression. These findings suggested that employing proper substrate stiffness might result in cells with more natural profiles similar to the nervous system and superior usefulness in therapeutic applications.
Collapse
Affiliation(s)
- Sareh Pandamooz
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arman Jafari
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Chemical Engineering, School of Chemical and Petroleum Engineering, Shiraz University, Shiraz, Iran
| | - Mohammad S Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benjamin Jurek
- Department of Behavioral and Molecular Neurobiology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anahid Safari
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shadi Hassanajili
- Department of Chemical Engineering, School of Chemical and Petroleum Engineering, Shiraz University, Shiraz, Iran
| | | | - Mehdi Dianatpour
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Leila Dargahi
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Greiner JFW, Merten M, Kaltschmidt C, Kaltschmidt B. Sexual dimorphisms in adult human neural, mesoderm-derived, and neural crest-derived stem cells. FEBS Lett 2019; 593:3338-3352. [PMID: 31529465 DOI: 10.1002/1873-3468.13606] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/03/2019] [Accepted: 09/05/2019] [Indexed: 12/31/2022]
Abstract
Sexual dimorphisms contribute, at least in part, to the severity and occurrence of a broad range of neurodegenerative, cardiovascular, and bone disorders. In addition to hormonal factors, increasing evidence suggests that stem cell-intrinsic mechanisms account for sex-specific differences in human physiology and pathology. Here, we discuss sex-related intrinsic mechanisms in adult stem cell populations, namely mesoderm-derived stem cells, neural stem cells (NSCs), and neural crest-derived stem cells (NCSCs), and their implications for stem cell differentiation and regeneration. We particularly focus on sex-specific differences in stem cell-mediated bone regeneration, in neuronal development, and in NSC-mediated neuroprotection. Moreover, we review our own recently published observations regarding the sex-dependent role of NF-κB-p65 in neuroprotection of human NCSC-derived neurons and sex differences in NCSC-related disorders, so-called neurocristopathies. These observations are in accordance with the increasing evidence pointing toward sex-specific differences in neurocristopathies and degenerative diseases like Parkinson's disease or osteoporosis. All findings discussed here indicate that sex-specific variability in stem cell biology may become a crucial parameter for the design of future treatment strategies.
Collapse
Affiliation(s)
| | - Madlen Merten
- Molecular Neurobiology, Bielefeld University, Germany
| | | | - Barbara Kaltschmidt
- Department of Cell Biology, Bielefeld University, Germany.,Molecular Neurobiology, Bielefeld University, Germany
| |
Collapse
|
23
|
Zhang L, Li B, Liu B, Dong Z. Co-transplantation of Epidermal Neural Crest Stem Cells and Olfactory Ensheathing Cells Repairs Sciatic Nerve Defects in Rats. Front Cell Neurosci 2019; 13:253. [PMID: 31244611 PMCID: PMC6582070 DOI: 10.3389/fncel.2019.00253] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 05/21/2019] [Indexed: 12/20/2022] Open
Abstract
Cell-based therapy is an alternative strategy to improve outcomes of peripheral nerve injury (PNI). Epidermal neural crest stem cell (EPI-NCSC) is obtained from autologous tissue without immunological rejection, which could expand quickly in vitro and is suitable candidate for cell-based therapy. Olfactory ensheathing cell (OEC) could secrete multiple neurotrophic factors (NTFs), which is often used to repair PNI individually. However, whether the combination of EPI-NCSC and OEC have better effects on PNI repair remains unclear. Here we use EPI-NCSC and OEC co-transplantation in a rat sciatic nerve defect model to ascertain the effects and potential mechanisms of cells co-transplantation on PNI. The effect of EPI-NCSC and OEC co-transplantation on PNI is assessed by using a combination of immunohistochemistry (IHC), electrophysiological recording and neural function test. Co-transplantation of EPI-NCSC and OEC exerts a beneficial effect upon PNI such as better organized structure, nerve function recovery, and lower motoneuron apoptosis. IHC and enzyme-linked immuno sorbent assay (ELISA) further demonstrate that cells co-transplantation may improve PNI via the expression of brain derived growth factor (BDNF) and nerve growth factor (NGF) up-regulated by EPI-NCSC and OEC synergistically. Eventually, the results from this study reveal that EPI-NCSC and OEC co-transplantation effectively repairs PNI through enhancing the level of BDNF and NGF, indicating that cells co-transplantation may serve as a fruitful avenue for PNI in clinic treatment.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Bingcang Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Research Institute of Surgery, Third Military Medical University, Chongqing, China
| | - Bin Liu
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Zhifang Dong
- Department of Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| |
Collapse
|
24
|
Schürmann M, Brotzmann V, Bütow M, Greiner J, Höving A, Kaltschmidt C, Kaltschmidt B, Sudhoff H. Identification of a Novel High Yielding Source of Multipotent Adult Human Neural Crest-Derived Stem Cells. Stem Cell Rev Rep 2018; 14:277-285. [PMID: 29243108 DOI: 10.1007/s12015-017-9797-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Due to their extraordinarily broad differentiation potential and persistence during adulthood, adult neural crest-derived stem cells (NCSCs) are highly promising candidates for clinical applications, particularly when facing the challenging treatment of neurodegenerative diseases or complex craniofacial injuries. Successful application of human NCSCs in regenerative medicine and pharmaceutical research mainly relies on the availability of sufficient amounts of tissue for cell isolation procedures. Facing this challenge, we here describe for the first time a novel population of NCSCs within the middle turbinate of the human nasal cavity. From a surgical point of view, high amounts of tissue are routinely and easily removed during nasal biopsies. Investigating the presence of putative stem cells in obtained middle turbinate tissue by immunohistochemistry, we observed Nestin+/p75NTR+/S100+/α smooth muscle actin (αSMA)- cells, which we successfully isolated and cultivated in vitro. Cultivated middle turbinate stem cells (MTSCs) kept their expression of neural crest and stemness markers Nestin, p75 NTR and S100 and showed the capability of sphere formation and clonal growth, indicating their stem cell character. Application of directed in vitro differentiation assays resulted in successful differentiation of MTSCs into osteogenic and neuronal cell types. Regarding the high amount of tissue obtained during surgery as well as their broad differentiation capability, MTSCs seem to be a highly promising novel neural crest stem cell population for applications in cell replacement therapy and pharmacological research.
Collapse
Affiliation(s)
- Matthias Schürmann
- Department of Otolaryngology, Head and Neck Surgery, Klinikum Bielefeld, Teutoburger Straße 50, 33604, Bielefeld, Germany
| | - Viktoria Brotzmann
- Department of Otolaryngology, Head and Neck Surgery, Klinikum Bielefeld, Teutoburger Straße 50, 33604, Bielefeld, Germany
| | - Marlena Bütow
- Department of Otolaryngology, Head and Neck Surgery, Klinikum Bielefeld, Teutoburger Straße 50, 33604, Bielefeld, Germany
| | - Johannes Greiner
- Department of Cell Biology, University of Bielefeld, 33615, Bielefeld, Germany
| | - Anna Höving
- Department of Cell Biology, University of Bielefeld, 33615, Bielefeld, Germany
| | | | - Barbara Kaltschmidt
- Department of Cell Biology, University of Bielefeld, 33615, Bielefeld, Germany
- AG Molecular Neurobiology, University of Bielefeld, 33615, Bielefeld, Germany
| | - Holger Sudhoff
- Department of Otolaryngology, Head and Neck Surgery, Klinikum Bielefeld, Teutoburger Straße 50, 33604, Bielefeld, Germany.
| |
Collapse
|
25
|
Tseropoulos G, Moghadasi Boroujeni S, Bajpai VK, Lei P, Andreadis ST. Derivation of neural crest stem cells from human epidermal keratinocytes requires FGF-2, IGF-1, and inhibition of TGF-β1. Bioeng Transl Med 2018; 3:256-264. [PMID: 30377664 PMCID: PMC6195909 DOI: 10.1002/btm2.10109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/30/2018] [Accepted: 07/30/2018] [Indexed: 11/29/2022] Open
Abstract
Neural crest (NC) cells play a central role in forming the peripheral nervous system, the craniofacial skeleton, and the pigmentation of the skin during development due to their broad multilineage differentiation potential into neurons, Schwann cells, melanocytes, and mesenchymal stem cells. Recently, we identified an easily accessible source of pluripotent NC stem cells from human inter‐follicular keratinocyte (KC) cultures (KC‐NC). In this work, we examined specific conditions for the derivation of NC from KC cultures. More specifically, we examined the role of two growth factors, FGF2 and IGF1, in NC proliferation and in expression of two potent NC transcription factors, Sox10 and FoxD3. Using specific chemical inhibitors, we uncovered that the downstream regulatory pathways AKT/PI3K, MEK/ERK, and JNK/cJun may be critical in Sox10 and FoxD3 regulation in KC‐NC. The TGF‐β1 pathway was also implicated in suppressing Sox10 expression and NC proliferation. In summary, our study shed light into the role of FGF2, IGF1, and TGF‐β1 on the induction of NC from KC cultures and the pathways that regulate Sox10 and FoxD3. We also established culture conditions for sustaining KC‐NC multipotency and, therefore, the potential of these cells for regenerative medicine and cellular therapies.
Collapse
Affiliation(s)
- Georgios Tseropoulos
- Dept. of Chemical and Biological Engineering University at Buffalo Buffalo NY 14260
| | | | - Vivek K Bajpai
- Dept. of Chemical and Biological Engineering University at Buffalo Buffalo NY 14260
| | - Pedro Lei
- Dept. of Chemical and Biological Engineering University at Buffalo Buffalo NY 14260
| | - Stelios T Andreadis
- Dept. of Chemical and Biological Engineering University at Buffalo Buffalo NY 14260.,Dept. of Biomedical Engineering University at Buffalo Buffalo NY 14228.,Center of Excellence in Bioinformatics and Life Sciences Buffalo NY 14203
| |
Collapse
|
26
|
Kosykh A, Beilin A, Sukhinich K, Vorotelyak E. Postnatal neural crest stem cells from hair follicle interact with nerve tissue in vitro and in vivo. Tissue Cell 2018; 54:94-104. [PMID: 30309515 DOI: 10.1016/j.tice.2018.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 01/05/2023]
Abstract
Neural crest stem cells that located in the postnatal hair follicle (HF-NCSC) are considered a promising tool for treatment of nervous system diseases and injuries. It is well known that HF-NCSC can be used in the spinal cord and sciatic nerve reparation but their ability to restore brain structures is poorly studied. In this article we are investigating the interaction between HF-NCSC and a nerve tissue (embryonic and adult). We have found out that HF-NCSC isolated from adult mice grow and differentiate in accordance with the mouse embryo developmental stage when co-cultured with the embryonic nerve tissue. The HF-NCSC migration is slower in the late embryonic tissue co-culture system compared to the early one. This phenomenon is related to the motor function of the cells but not to their proliferation level. We have demonstrated that the embryonic nerve tissue maintains HF-NCSC an undifferentiated status, while an adult brain tissue inhibits the cell proliferation and activates the differentiation processes. Besides, HF-NCSC pre-differentiated into the neuronal direction shows a higher survival and migration rate after the transplantation into the adult brain tissue compared to the undifferentiated HF-NCSC. Thus, we have investigated the postnatal HF-NCSC response to the nerve tissue microenvironment to analyze their possible application to the brain repair processes.
Collapse
Affiliation(s)
- Anastasiia Kosykh
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Vavilova 26, 119334, Moscow, Russian Federation; Pirogov Russian National Research Medical University, Ostrovitianova 1, 117997, Moscow, Russian Federation.
| | - Arkadii Beilin
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Vavilova 26, 119334, Moscow, Russian Federation
| | - Kirill Sukhinich
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Vavilova 26, 119334, Moscow, Russian Federation
| | - Ekaterina Vorotelyak
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Vavilova 26, 119334, Moscow, Russian Federation; Pirogov Russian National Research Medical University, Ostrovitianova 1, 117997, Moscow, Russian Federation; Lomonosov Moscow State University, Leninskie Gory 1, Moscow, Russian Federation
| |
Collapse
|
27
|
Human peptidergic nociceptive sensory neurons generated from human epidermal neural crest stem cells (hEPI-NCSC). PLoS One 2018; 13:e0199996. [PMID: 29953534 PMCID: PMC6023242 DOI: 10.1371/journal.pone.0199996] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 06/17/2018] [Indexed: 01/28/2023] Open
Abstract
Here we provide new technology for generating human peptidergic nociceptive sensory neurons in a straightforward and efficient way. The cellular source, human epidermal neural crest stem cells (hEPI-NCSC), consists of multipotent somatic stem cells that reside in the bulge of hair follicles. hEPI-NCSC and primary sensory neurons have a common origin, the embryonic neural crest. For directed differentiation, hEPI-NCSC were exposed to pertinent growth factors and small molecules in order to modulate master signalling networks involved in differentiation of neural crest cells into postmitotic peptidergic sensory neurons during embryonic development. The neuronal populations were homogenous in regard to antibody marker expression. Cells were immunoreactive for essential master regulatory genes, including NGN1/2, SOX10, and BRN3a among others, and for the pain-mediating genes substance P (SP), calcitonin gene related protein (CGRP) and the TRPV1 channel. Approximately 30% of total cells responded to capsaicin, indicating that they expressed an active TRPV1 channel. In summary, hEPI-NCSC are a biologically relevant and easily available source of somatic stem cells for generating human peptidergic nociceptive neurons without the need for genetic manipulation and cell purification. As no analgesics exist that specifically target TRPV1, a ready supply of high-quality human peptidergic nociceptive sensory neurons could open the way for new approaches, in a biologically relevant cellular context, to drug discovery and patient-specific disease modelling that is aimed at pain control, and as such is highly desirable.
Collapse
|
28
|
Zhang Q, Nguyen PD, Shi S, Burrell JC, Xu Q, Cullen KD, Le AD. Neural Crest Stem-Like Cells Non-genetically Induced from Human Gingiva-Derived Mesenchymal Stem Cells Promote Facial Nerve Regeneration in Rats. Mol Neurobiol 2018; 55:6965-6983. [PMID: 29372546 DOI: 10.1007/s12035-018-0913-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/15/2018] [Indexed: 02/07/2023]
Abstract
Non-genetic induction of somatic cells into neural crest stem-like cells (NCSCs) is promising for potential cell-based therapies for post-traumatic peripheral nerve regeneration. Here, we report that human gingiva-derived mesenchymal stem cells (GMSCs) could be reproducibly and readily induced into NCSCs via non-genetic approaches. Compared to parental GMSCs, induced NCSC population had increased expression in NCSC-related genes and displayed robust differentiation into neuronal and Schwann-like cells. Knockdown of the expression of Yes-associated protein 1 (YAP1), a critical mechanosensor and mechanotransducer, attenuated the expression of NCSC-related genes; specific blocking of RhoA/ROCK activity and non-muscle myosin II (NM II)-dependent contraction suppressed YAP1 and NCSC-related genes and concurrently abolished neural spheroid formation in NCSCs. Using a rat model of facial nerve defect, implantation of NCSC-laden nerve conduits promoted functional regeneration of the injured nerve. These promising findings demonstrate that induced NCSCs derived from GMSCs represent an easily accessible and promising source of neural stem-like cells for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Qunzhou Zhang
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA, 19104, USA
| | - Phuong D Nguyen
- Division of Plastic and Reconstructive Surgery, University of Pennsylvania Perelman School of Medicine, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Shihong Shi
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA, 19104, USA
| | - Justin C Burrell
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, 3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - Qilin Xu
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA, 19104, USA
| | - Kacy D Cullen
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, 3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - Anh D Le
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA, 19104, USA.
- Department of Oral and Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, Perelman Center for Advanced Medicine, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
29
|
Prodinger CM, Reichelt J, Bauer JW, Laimer M. Current and Future Perspectives of Stem Cell Therapy in Dermatology. Ann Dermatol 2017; 29:667-687. [PMID: 29200755 PMCID: PMC5705348 DOI: 10.5021/ad.2017.29.6.667] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 09/05/2017] [Indexed: 12/19/2022] Open
Abstract
Stem cells are undifferentiated cells capable of generating, sustaining, and replacing terminally differentiated cells and tissues. They can be isolated from embryonic as well as almost all adult tissues including skin, but are also generated through genetic reprogramming of differentiated cells. Preclinical and clinical research has recently tremendously improved stem cell therapy, being a promising treatment option for various diseases in which current medical therapies fail to cure, prevent progression or relieve symptoms. With the main goal of regeneration or sustained genetic correction of damaged tissue, advanced tissue-engineering techniques are especially applicable for many dermatological diseases including wound healing, genodermatoses (like the severe blistering disorder epidermolysis bullosa) and chronic (auto-)inflammatory diseases. This review summarizes general aspects as well as current and future perspectives of stem cell therapy in dermatology.
Collapse
Affiliation(s)
- Christine M Prodinger
- Department of Dermatology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Julia Reichelt
- Department of Dermatology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Johann W Bauer
- Department of Dermatology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Martin Laimer
- Department of Dermatology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| |
Collapse
|
30
|
Wang D, Wu F, Yuan H, Wang A, Kang GJ, Truong T, Chen L, McCallion AS, Gong X, Li S. Sox10 + Cells Contribute to Vascular Development in Multiple Organs-Brief Report. Arterioscler Thromb Vasc Biol 2017; 37:1727-1731. [PMID: 28751573 PMCID: PMC5572822 DOI: 10.1161/atvbaha.117.309774] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 07/18/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Previous genetic lineage tracing studies showed that Sox10+ cells differentiate into vascular mural cells, limited to neural crest-derived blood vessels in craniofacial tissues, aortic arch, pulmonary arch arteries, brachiocephalic, carotid arteries, and thymus. The purpose of this study was to investigate the contribution of Sox10+ cells to the vascular development in other tissues and organs and their relationship with neural crest. APPROACH AND RESULTS Using genetic lineage tracing technique based on Cre/LoxP system, we examined blood vessels in the adult organs of the mice expressing Sox10-Cre/Rosa-LoxP-red fluorescent protein or Wnt1-Cre/Rosa-LoxP-red fluorescent protein by immunohistological analysis. In addition to previously reported tissues and organs derived from neural crest, we showed that Sox10+ cells also contributed to vascular mural cells in the lung, spleen, and kidney, which are derived from non-neural crest origin as evidenced by red fluorescent protein-negative blood vessels in these 3 organs of Wnt1-Cre/Rosa-LoxP-red fluorescent protein mice. CONCLUSIONS This study demonstrates that Sox10+ cells contribute to pericytes and smooth muscle cells in most parts of the body, including those from neural crest and non-neural crest, which has significant implications in vascular remodeling under physiological and pathological conditions.
Collapse
Affiliation(s)
- Dong Wang
- From the Department of Bioengineering (D.W., F.W., A.W., S.L.) and School of Optometry and Vision Science Graduate Program (D.W., G.J.K., T.T., L.C., X.G.), University of California, Berkeley; Department of Bioengineering (D.W., H.Y., S.L.) and Department of Medicine (S.L.), University of California, Los Angeles; The Second Xiangya Hospital of Central South University (H.Y.); Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis (A.W.); and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine (A.S.M)
| | - Fan Wu
- From the Department of Bioengineering (D.W., F.W., A.W., S.L.) and School of Optometry and Vision Science Graduate Program (D.W., G.J.K., T.T., L.C., X.G.), University of California, Berkeley; Department of Bioengineering (D.W., H.Y., S.L.) and Department of Medicine (S.L.), University of California, Los Angeles; The Second Xiangya Hospital of Central South University (H.Y.); Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis (A.W.); and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine (A.S.M)
| | - Haoyong Yuan
- From the Department of Bioengineering (D.W., F.W., A.W., S.L.) and School of Optometry and Vision Science Graduate Program (D.W., G.J.K., T.T., L.C., X.G.), University of California, Berkeley; Department of Bioengineering (D.W., H.Y., S.L.) and Department of Medicine (S.L.), University of California, Los Angeles; The Second Xiangya Hospital of Central South University (H.Y.); Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis (A.W.); and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine (A.S.M)
| | - Aijun Wang
- From the Department of Bioengineering (D.W., F.W., A.W., S.L.) and School of Optometry and Vision Science Graduate Program (D.W., G.J.K., T.T., L.C., X.G.), University of California, Berkeley; Department of Bioengineering (D.W., H.Y., S.L.) and Department of Medicine (S.L.), University of California, Los Angeles; The Second Xiangya Hospital of Central South University (H.Y.); Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis (A.W.); and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine (A.S.M)
| | - Gyeong Jin Kang
- From the Department of Bioengineering (D.W., F.W., A.W., S.L.) and School of Optometry and Vision Science Graduate Program (D.W., G.J.K., T.T., L.C., X.G.), University of California, Berkeley; Department of Bioengineering (D.W., H.Y., S.L.) and Department of Medicine (S.L.), University of California, Los Angeles; The Second Xiangya Hospital of Central South University (H.Y.); Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis (A.W.); and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine (A.S.M)
| | - Tan Truong
- From the Department of Bioengineering (D.W., F.W., A.W., S.L.) and School of Optometry and Vision Science Graduate Program (D.W., G.J.K., T.T., L.C., X.G.), University of California, Berkeley; Department of Bioengineering (D.W., H.Y., S.L.) and Department of Medicine (S.L.), University of California, Los Angeles; The Second Xiangya Hospital of Central South University (H.Y.); Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis (A.W.); and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine (A.S.M)
| | - Lu Chen
- From the Department of Bioengineering (D.W., F.W., A.W., S.L.) and School of Optometry and Vision Science Graduate Program (D.W., G.J.K., T.T., L.C., X.G.), University of California, Berkeley; Department of Bioengineering (D.W., H.Y., S.L.) and Department of Medicine (S.L.), University of California, Los Angeles; The Second Xiangya Hospital of Central South University (H.Y.); Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis (A.W.); and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine (A.S.M)
| | - Andrew S McCallion
- From the Department of Bioengineering (D.W., F.W., A.W., S.L.) and School of Optometry and Vision Science Graduate Program (D.W., G.J.K., T.T., L.C., X.G.), University of California, Berkeley; Department of Bioengineering (D.W., H.Y., S.L.) and Department of Medicine (S.L.), University of California, Los Angeles; The Second Xiangya Hospital of Central South University (H.Y.); Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis (A.W.); and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine (A.S.M)
| | - Xiaohua Gong
- From the Department of Bioengineering (D.W., F.W., A.W., S.L.) and School of Optometry and Vision Science Graduate Program (D.W., G.J.K., T.T., L.C., X.G.), University of California, Berkeley; Department of Bioengineering (D.W., H.Y., S.L.) and Department of Medicine (S.L.), University of California, Los Angeles; The Second Xiangya Hospital of Central South University (H.Y.); Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis (A.W.); and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine (A.S.M)
| | - Song Li
- From the Department of Bioengineering (D.W., F.W., A.W., S.L.) and School of Optometry and Vision Science Graduate Program (D.W., G.J.K., T.T., L.C., X.G.), University of California, Berkeley; Department of Bioengineering (D.W., H.Y., S.L.) and Department of Medicine (S.L.), University of California, Los Angeles; The Second Xiangya Hospital of Central South University (H.Y.); Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis (A.W.); and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine (A.S.M).
| |
Collapse
|
31
|
Human bone marrow harbors cells with neural crest-associated characteristics like human adipose and dermis tissues. PLoS One 2017; 12:e0177962. [PMID: 28683107 PMCID: PMC5500284 DOI: 10.1371/journal.pone.0177962] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 05/05/2017] [Indexed: 12/13/2022] Open
Abstract
Adult neural crest stem-derived cells (NCSC) are of extraordinary high plasticity and promising candidates for use in regenerative medicine. Several locations such as skin, adipose tissue, dental pulp or bone marrow have been described in rodent, as sources of NCSC. However, very little information is available concerning their correspondence in human tissues, and more precisely for human bone marrow. The main objective of this study was therefore to characterize NCSC from adult human bone marrow. In this purpose, we compared human bone marrow stromal cells to human adipose tissue and dermis, already described for containing NCSC. We performed comparative analyses in terms of gene and protein expression as well as functional characterizations. It appeared that human bone marrow, similarly to adipose tissue and dermis, contains NESTIN+ / SOX9+ / TWIST+ / SLUG+ / P75NTR+/ BRN3A+/ MSI1+/ SNAIL1+ cells and were able to differentiate into melanocytes, Schwann cells and neurons. Moreover, when injected into chicken embryos, all those cells were able to migrate and follow endogenous neural crest migration pathways. Altogether, the phenotypic characterization and migration abilities strongly suggest the presence of neural crest-derived cells in human adult bone marrow.
Collapse
|
32
|
Sauer CM, Haugg AM, Chteinberg E, Rennspiess D, Winnepenninckx V, Speel EJ, Becker JC, Kurz AK, Zur Hausen A. Reviewing the current evidence supporting early B-cells as the cellular origin of Merkel cell carcinoma. Crit Rev Oncol Hematol 2017; 116:99-105. [PMID: 28693804 DOI: 10.1016/j.critrevonc.2017.05.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 02/13/2017] [Accepted: 05/28/2017] [Indexed: 12/18/2022] Open
Abstract
Merkel cell carcinoma (MCC) is a highly malignant skin cancer characterized by early metastases and poor survival. Although MCC is a rare malignancy, its incidence is rapidly increasing in the U.S. and Europe. The discovery of the Merkel cell polyomavirus (MCPyV) has enormously impacted our understanding of its etiopathogenesis and biology. MCCs are characterized by trilinear differentiation, comprising the expression of neuroendocrine, epithelial and B-lymphoid lineage markers. To date, it is generally accepted that the initial assumption of MCC originating from Merkel cells (MCs) is unlikely. This is owed to their post-mitotic character, absence of MCPyV in MCs and discrepant protein expression pattern in comparison to MCC. Evidence from mouse models suggests that epidermal/dermal stem cells might be of cellular origin in MCC. The recently formulated hypothesis of MCC originating from early B-cells is based on morphology, the consistent expression of early B-cell lineage markers and the finding of clonal immunoglobulin chain rearrangement in MCC cells. In this review we elaborate on the cellular ancestry of MCC, the identification of which could pave the way for novel and more effective therapeutic regimens.
Collapse
Affiliation(s)
- C M Sauer
- Department of Pathology, GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands; Department of Internal Medicine IV, University Hospital Aachen, Aachen, Germany
| | - A M Haugg
- Department of Pathology, GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - E Chteinberg
- Department of Pathology, GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - D Rennspiess
- Department of Pathology, GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - V Winnepenninckx
- Department of Pathology, GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - E-J Speel
- Department of Pathology, GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - J C Becker
- Department for Translational Dermato-Oncology (DKTK), Center for Medical Biotechnology (ZMB), University Hospital Essen, Essen, Germany
| | - A K Kurz
- Department of Internal Medicine IV, University Hospital Aachen, Aachen, Germany
| | - A Zur Hausen
- Department of Pathology, GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands.
| |
Collapse
|
33
|
Abstract
For decades, researchers have been fascinated by the strategy of using cell therapy for bone defects; some progress in the field has been made. Owing to its ample supply and easy access, skin, the largest organ in the body, has gained attention as a potential source of stem cells. Despite extensive applications in skin and nerve regeneration, an increasing number of reports indicate its potential use in bone tissue engineering and regeneration. Unfortunately, few review articles are available to outline current research efforts in skin-based osteogenesis. This review first summarizes the latest findings on stem cells or progenitors in skin and their niches and then discusses the strategies of skin cell-based osteogenesis. We hope this article elucidates this topic and generates new ideas for future studies.
Collapse
Affiliation(s)
- Tingliang Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA.,Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lian Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA.,Division of Exercise Physiology, West Virginia University, Morgantown, WV, USA.,Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
34
|
Hendijani F. Explant culture: An advantageous method for isolation of mesenchymal stem cells from human tissues. Cell Prolif 2017; 50:e12334. [PMID: 28144997 PMCID: PMC6529062 DOI: 10.1111/cpr.12334] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/03/2017] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cell (MSC) research progressively moves towards clinical phases. Accordingly, a wide range of different procedures were presented in the literature for MSC isolation from human tissues; however, there is not yet any close focus on the details to offer precise information for best method selection. Choosing a proper isolation method is a critical step in obtaining cells with optimal quality and yield in companion with clinical and economical considerations. In this concern, current review widely discusses advantages of omitting proteolysis step in isolation process and presence of tissue pieces in primary culture of MSCs, including removal of lytic stress on cells, reduction of in vivo to in vitro transition stress for migrated/isolated cells, reduction of price, processing time and labour, removal of viral contamination risk, and addition of supporting functions of extracellular matrix and released growth factors from tissue explant. In next sections, it provides an overall report of technical highlights and molecular events of explant culture method for isolation of MSCs from human tissues including adipose tissue, bone marrow, dental pulp, hair follicle, cornea, umbilical cord and placenta. Focusing on informative collection of molecular and methodological data about explant methods can make it easy for researchers to choose an optimal method for their experiments/clinical studies and also stimulate them to investigate and optimize more efficient procedures according to clinical and economical benefits.
Collapse
Affiliation(s)
- Fatemeh Hendijani
- Faculty of PharmacyHormozgan University of Medical SciencesBandar AbbasIran
| |
Collapse
|
35
|
|
36
|
Chen CC, Hsia CW, Ho CW, Liang CM, Chen CM, Huang KL, Kang BH, Chen YH. Hypoxia and hyperoxia differentially control proliferation of rat neural crest stem cells via distinct regulatory pathways of the HIF1α-CXCR4 and TP53-TPM1 proteins. Dev Dyn 2017; 246:162-185. [PMID: 28002632 DOI: 10.1002/dvdy.24481] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 12/09/2016] [Accepted: 12/13/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Neural crest stem cells (NCSCs) are a population of adult multipotent stem cells. We are interested in studying whether oxygen tensions affect the capability of NCSCs to self-renew and repair damaged tissues. NCSCs extracted from the hair follicle bulge region of the rat whisker pad were cultured in vitro under different oxygen tensions. RESULTS We found significantly increased and decreased rates of cell proliferation in rat NCSCs (rNCSCs) cultured, respectively, at 0.5% and 80% oxygen levels. At 0.5% oxygen, the expression of both hypoxia-inducible factor (HIF) 1α and CXCR4 was greatly enhanced in the rNCSC nuclei and was suppressed by incubation with the CXCR4-specific antagonist AMD3100. In addition, the rate of cell apoptosis in the rNCSCs cultured at 80% oxygen was dramatically increased, associated with increased nuclear expression of TP53, decreased cytoplasmic expression of TPM1 (tropomyosin-1), and increased nuclear-to-cytoplasmic translocation of S100A2. Incubation of rNCSCs with the antioxidant N-acetylcysteine (NAC) overcame the inhibitory effect of 80% oxygen on proliferation and survival of rNCSCs. CONCLUSIONS Our results show for the first time that extreme oxygen tensions directly control NCSC proliferation differentially via distinct regulatory pathways of proteins, with hypoxia via the HIF1α-CXCR4 pathway and hyperoxia via the TP53-TPM1 pathway. Developmental Dynamics 246:162-185, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Chien-Cheng Chen
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Neihu District, Taipei City, Taiwan
| | - Ching-Wu Hsia
- Department of Finance, School of Management, Shih Hsin University, Wenshan District, Taipei City, Taiwan
| | - Cheng-Wen Ho
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Neihu District, Taipei City, Taiwan
- Division of Rehabilitation Medicine, Taoyuan Armed Forces General Hospital, Longtan District, Taoyuan City, Taiwan
| | - Chang-Min Liang
- Department of Ophthalmology, Tri-Service General Hospital, Neihu District, Taipei City, Taiwan
| | - Chieh-Min Chen
- Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Neihu District, Taipei City, Taiwan
| | - Kun-Lun Huang
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Neihu District, Taipei City, Taiwan
- Department of Undersea and Hyperbaric Medicine, Tri-Service General Hospital, Neihu District, Taipei City, Taiwan
| | - Bor-Hwang Kang
- Division of Diving Medicine, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Zuoying District, Kaohsiung City, Taiwan
- Department of Otorhinolaryngology - Head and Neck Surgery, Tri-Service General Hospital, Taipei City, Taiwan
| | - Yi-Hui Chen
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Neihu District, Taipei City, Taiwan
| |
Collapse
|
37
|
Stabley JN, Towler DA. Arterial Calcification in Diabetes Mellitus: Preclinical Models and Translational Implications. Arterioscler Thromb Vasc Biol 2017; 37:205-217. [PMID: 28062508 PMCID: PMC5480317 DOI: 10.1161/atvbaha.116.306258] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus increasingly afflicts our aging and dysmetabolic population. Type 2 diabetes mellitus and the antecedent metabolic syndrome represent the vast majority of the disease burden-increasingly prevalent in children and older adults. However, type 1 diabetes mellitus is also advancing in preadolescent children. As such, a crushing wave of cardiometabolic disease burden now faces our society. Arteriosclerotic calcification is increased in metabolic syndrome, type 2 diabetes mellitus, and type 1 diabetes mellitus-impairing conduit vessel compliance and function, thereby increasing the risk for dementia, stroke, heart attack, limb ischemia, renal insufficiency, and lower extremity amputation. Preclinical models of these dysmetabolic settings have provided insights into the pathobiology of arterial calcification. Osteochondrogenic morphogens in the BMP-Wnt signaling relay and transcriptional regulatory programs driven by Msx and Runx gene families are entrained to innate immune responses-responses activated by the dysmetabolic state-to direct arterial matrix deposition and mineralization. Recent studies implicate the endothelial-mesenchymal transition in contributing to the phenotypic drift of mineralizing vascular progenitors. In this brief overview, we discuss preclinical disease models that provide mechanistic insights-and point to challenges and opportunities to translate these insights into new therapeutic strategies for our patients afflicted with diabetes mellitus and its arteriosclerotic complications.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Arteries/metabolism
- Arteries/pathology
- Atherosclerosis/etiology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetic Angiopathies/etiology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Diet, High-Fat
- Disease Models, Animal
- Female
- Genetic Predisposition to Disease
- Humans
- Hyperlipidemias/complications
- Hyperlipidemias/genetics
- Male
- Phenotype
- Plaque, Atherosclerotic
- Rats
- Signal Transduction
- Translational Research, Biomedical
- Vascular Calcification/etiology
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
Collapse
Affiliation(s)
- John N Stabley
- From the Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Dwight A Towler
- From the Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
38
|
Dunaway K, Goorha S, Matelski L, Urraca N, Lein PJ, Korf I, Reiter LT, LaSalle JM. Dental Pulp Stem Cells Model Early Life and Imprinted DNA Methylation Patterns. Stem Cells 2017; 35:981-988. [PMID: 28032673 DOI: 10.1002/stem.2563] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/27/2016] [Accepted: 12/12/2016] [Indexed: 12/20/2022]
Abstract
Early embryonic stages of pluripotency are modeled for epigenomic studies primarily with human embryonic stem cells (ESC) or induced pluripotent stem cells (iPSCs). For analysis of DNA methylation however, ESCs and iPSCs do not accurately reflect the DNA methylation levels found in preimplantation embryos. Whole genome bisulfite sequencing (WGBS) approaches have revealed the presence of large partially methylated domains (PMDs) covering 30%-40% of the genome in oocytes, preimplantation embryos, and placenta. In contrast, ESCs and iPSCs show abnormally high levels of DNA methylation compared to inner cell mass (ICM) or placenta. Here we show that dental pulp stem cells (DPSCs), derived from baby teeth and cultured in serum-containing media, have PMDs and mimic the ICM and placental methylome more closely than iPSCs and ESCs. By principal component analysis, DPSC methylation patterns were more similar to two other neural stem cell types of human derivation (EPI-NCSC and LUHMES) and placenta than were iPSCs, ESCs or other human cell lines (SH-SY5Y, B lymphoblast, IMR90). To test the suitability of DPSCs in modeling epigenetic differences associated with disease, we compared methylation patterns of DPSCs derived from children with chromosome 15q11.2-q13.3 maternal duplication (Dup15q) to controls. Differential methylation region (DMR) analyses revealed the expected Dup15q hypermethylation at the imprinting control region, as well as hypomethylation over SNORD116, and novel DMRs over 147 genes, including several autism candidate genes. Together these data suggest that DPSCs are a useful model for epigenomic and functional studies of human neurodevelopmental disorders. Stem Cells 2017;35:981-988.
Collapse
Affiliation(s)
- Keith Dunaway
- Medical Microbiology and Immunology, UC Davis, Davis, California, USA.,Genome Center, UC Davis, Davis, California, USA.,MIND Institute, UC Davis, Davis, California, USA.,Center for Children's Environmental Health, UC Davis, Davis, California, USA
| | - Sarita Goorha
- Department of Neurology, UTHSC, Memphis, Tennessee, USA.,Department of Pediatrics, UTHSC, Memphis, Tennessee, USA.,Department of Anatomy and Neurobiology, UTHSC, Memphis, Tennessee, USA
| | - Lauren Matelski
- MIND Institute, UC Davis, Davis, California, USA.,Center for Children's Environmental Health, UC Davis, Davis, California, USA.,Internal Medicine, UC Davis, Davis, California, USA
| | - Nora Urraca
- Department of Neurology, UTHSC, Memphis, Tennessee, USA
| | - Pamela J Lein
- MIND Institute, UC Davis, Davis, California, USA.,Center for Children's Environmental Health, UC Davis, Davis, California, USA.,Molecular Biosciences, UC Davis, Davis, California, USA
| | - Ian Korf
- Genome Center, UC Davis, Davis, California, USA.,Molecular and Cellular Biology, UC Davis, Davis, California, USA
| | - Lawrence T Reiter
- Department of Neurology, UTHSC, Memphis, Tennessee, USA.,Department of Pediatrics, UTHSC, Memphis, Tennessee, USA.,Department of Anatomy and Neurobiology, UTHSC, Memphis, Tennessee, USA
| | - Janine M LaSalle
- Medical Microbiology and Immunology, UC Davis, Davis, California, USA.,Genome Center, UC Davis, Davis, California, USA.,MIND Institute, UC Davis, Davis, California, USA.,Center for Children's Environmental Health, UC Davis, Davis, California, USA
| |
Collapse
|
39
|
Wang D, Wang A, Wu F, Qiu X, Li Y, Chu J, Huang WC, Xu K, Gong X, Li S. Sox10 + adult stem cells contribute to biomaterial encapsulation and microvascularization. Sci Rep 2017; 7:40295. [PMID: 28071739 PMCID: PMC5223127 DOI: 10.1038/srep40295] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/05/2016] [Indexed: 01/06/2023] Open
Abstract
Implanted biomaterials and biomedical devices generally induce foreign body reaction and end up with encapsulation by a dense avascular fibrous layer enriched in extracellular matrix. Fibroblasts/myofibroblasts are thought to be the major cell type involved in encapsulation, but it is unclear whether and how stem cells contribute to this process. Here we show, for the first time, that Sox10+ adult stem cells contribute to both encapsulation and microvessel formation. Sox10+ adult stem cells were found sparsely in the stroma of subcutaneous loose connective tissues. Upon subcutaneous biomaterial implantation, Sox10+ stem cells were activated and recruited to the biomaterial scaffold, and differentiated into fibroblasts and then myofibroblasts. This differentiation process from Sox10+ stem cells to myofibroblasts could be recapitulated in vitro. On the other hand, Sox10+ stem cells could differentiate into perivascular cells to stabilize newly formed microvessels. Sox10+ stem cells and endothelial cells in three-dimensional co-culture self-assembled into microvessels, and platelet-derived growth factor had chemotactic effect on Sox10+ stem cells. Transplanted Sox10+ stem cells differentiated into smooth muscle cells to stabilize functional microvessels. These findings demonstrate the critical role of adult stem cells in tissue remodeling and unravel the complexity of stem cell fate determination.
Collapse
Affiliation(s)
- Dong Wang
- Department of Bioengineering, University of California, Berkeley, California 94720, USA.,School of Optometry and Vision Science Program, University of California, Berkeley, California 94720, USA.,Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Aijun Wang
- Department of Bioengineering, University of California, Berkeley, California 94720, USA.,Department of Surgery, University of California, Davis, Sacramento, California 95817, USA
| | - Fan Wu
- Department of Bioengineering, University of California, Berkeley, California 94720, USA
| | - Xuefeng Qiu
- Department of Bioengineering, University of California, Berkeley, California 94720, USA.,Department of Bioengineering, University of California, Los Angeles, California 90095, USA.,Department of Cardiovascular Surgery, Union Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ye Li
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, California 94720, USA.,Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Julia Chu
- Department of Bioengineering, University of California, Berkeley, California 94720, USA
| | - Wen-Chin Huang
- Department of Bioengineering, University of California, Berkeley, California 94720, USA
| | - Kang Xu
- Department of Bioengineering, University of California, Berkeley, California 94720, USA.,Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Xiaohua Gong
- School of Optometry and Vision Science Program, University of California, Berkeley, California 94720, USA
| | - Song Li
- Department of Bioengineering, University of California, Berkeley, California 94720, USA.,Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
40
|
Liu JA, Cheung M. Neural crest stem cells and their potential therapeutic applications. Dev Biol 2016; 419:199-216. [PMID: 27640086 DOI: 10.1016/j.ydbio.2016.09.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 09/07/2016] [Accepted: 09/07/2016] [Indexed: 12/13/2022]
Abstract
The neural crest (NC) is a remarkable transient structure generated during early vertebrate development. The neural crest progenitors have extensive migratory capacity and multipotency, harboring stem cell-like characteristics such as self-renewal. They can differentiate into a variety of cell types from craniofacial skeletal tissues to the trunk peripheral nervous system (PNS). Multiple regulators such as signaling factors, transcription factors, and migration machinery components are expressed at different stages of NC development. Gain- and loss-of-function studies in various vertebrate species revealed epistatic relationships of these molecules that could be assembled into a gene regulatory network defining the processes of NC induction, specification, migration, and differentiation. These basic developmental studies led to the subsequent establishment and molecular validation of neural crest stem cells (NCSCs) derived by various strategies. We provide here an overview of the isolation and characterization of NCSCs from embryonic, fetal, and adult tissues; the experimental strategies for the derivation of NCSCs from embryonic stem cells, induced pluripotent stem cells, and skin fibroblasts; and recent developments in the use of patient-derived NCSCs for modeling and treating neurocristopathies. We discuss future research on further refinement of the culture conditions required for the differentiation of pluripotent stem cells into axial-specific NC progenitors and their derivatives, developing non-viral approaches for the generation of induced NC cells (NCCs), and using a genomic editing approach to correct genetic mutations in patient-derived NCSCs for transplantation therapy. These future endeavors should facilitate the therapeutic applications of NCSCs in the clinical setting.
Collapse
Affiliation(s)
- Jessica Aijia Liu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Martin Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
41
|
Lin SC, Gou GH, Hsia CW, Ho CW, Huang KL, Wu YF, Lee SY, Chen YH. Simulated Microgravity Disrupts Cytoskeleton Organization and Increases Apoptosis of Rat Neural Crest Stem Cells Via Upregulating CXCR4 Expression and RhoA-ROCK1-p38 MAPK-p53 Signaling. Stem Cells Dev 2016; 25:1172-93. [PMID: 27269634 DOI: 10.1089/scd.2016.0040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Neural crest stem cells (NCSCs) are a population of multipotent stem cells that are distributed broadly in many tissues and organs and are capable of differentiating into a variety of cell types that are dispersed throughout three germ layers. We are interested in studying the effects of simulated microgravity on the survival and self-renewal of NCSCs. NCSCs extracted from the hair follicle bulge region of the rat whisker pad were cultured in vitro, respectively, in a 2D adherent environment and a 3D suspension environment using the rotatory cell culture system (RCCS) to simulate microgravity. We found that rat NCSCs (rNCSCs) cultured in the RCCS for 24 h showed disrupted organization of filamentous actin, increased globular actin level, formation of plasma membrane blebbing and neurite-like artifact, as well as decreased levels of cortactin and vimentin. Interestingly, ∼70% of RCCS-cultured rNCSCs co-expressed cleaved (active) caspase-3 and neuronal markers microtubule-associated protein 2 (MAP2) and Tuj1 instead of NCSC markers, suggesting stress-induced formation of neurite-like artifact in rNCSCs. In addition, rNCSCs showed increased C-X-C chemokine receptor 4 (CXCR4) expression, RhoA GTPase activation, Rho-associated kinase 1 (ROCK1) and p38 mitogen-activated protein kinase (MAPK) phosphorylation, and p53 expression in the nucleus. Incubation of rNCSCs with the Gα protein inhibitor pertussis toxin or CXCR4 siRNA during RCCS-culturing prevented cytoskeleton disorganization and plasma membrane blebbing, and it suppressed apoptosis of rNCSCs. Taken together, we demonstrate for the first time that simulated microgravity disrupts cytoskeleton organization and increases apoptosis of rNCSCs via upregulating CXCR4 expression and the RhoA-ROCK1-p38 MAPK-p53 signaling pathway.
Collapse
Affiliation(s)
- Shing-Chen Lin
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| | - Guo-Hau Gou
- 2 Graduate Institute of Medical Sciences, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| | - Ching-Wu Hsia
- 2 Graduate Institute of Medical Sciences, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| | - Cheng-Wen Ho
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan .,3 Division of Rehabilitation Medicine, Taoyuan Armed Forces General Hospital , Longtan Township, Taoyuan County, Taiwan
| | - Kun-Lun Huang
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan .,4 Department of Undersea and Hyperbaric Medicine, Tri-Service General Hospital , Neihu District, Taipei City, Taiwan
| | - Yung-Fu Wu
- 5 Department of Medical Research, Tri-Service General Hospital , Neihu District, Taipei City, Taiwan
| | - Shih-Yu Lee
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| | - Yi-Hui Chen
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| |
Collapse
|
42
|
Azmahani A, Nakamura Y, Ishida H, McNamara KM, Fujimura T, Haga T, Hashimoto A, Aiba S, Sasano H. Estrogen receptor β in Merkel cell carcinoma: its possible roles in pathogenesis. Hum Pathol 2016; 56:128-33. [PMID: 27343835 DOI: 10.1016/j.humpath.2016.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/30/2016] [Accepted: 06/11/2016] [Indexed: 12/27/2022]
Abstract
Sex steroids have been postulated to influence skin development and functions as well as its pathogenesis. MCC occurs in both sexes; however, the specific differences in pathogenesis among sexes have yet to be conclusively defined. The detailed status of sex steroid receptors (AR, PRA and PRB, and ERα, ERβ) are also unknown in MCC patients. We first immunolocalized sex steroid receptors and compared the results with immunolocalization of relevant transcription factors including SOX2, FOXA1, and Bcl-2 and Ki-67 in 18 cases of MCCs. AR, PRA, PRB, ERα, ERβ, Bcl-2, SOX2, and FOXA1 immunoreactivity was evaluated by using the modified H score method, and Ki-67 was quantified using labeling index. ERβ immunoreactivity was markedly present in all the cases of MCC examined, with relatively weak immunoreactivity of ERα, AR, PRA, and PRB. The status of ERβ immunoreactivity was also significantly correlated with Ki-67 labeling index and Bcl-2 score. These results demonstrated that ERβ could be associated with regulation of both cell proliferation and apoptosis in MCCs.
Collapse
Affiliation(s)
- Abdullah Azmahani
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; Faculty of Health Sciences, University Sultan Zainal Abidin, 21300 Kuala Terengganu, Terengganu, Malaysia
| | - Yasuhiro Nakamura
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan.
| | - Hirotaka Ishida
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; Division of Advanced Surgical Science and Technology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Keely M McNamara
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Takahiro Haga
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Akira Hashimoto
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Setsuya Aiba
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| |
Collapse
|
43
|
Isolation, expansion and neural differentiation of stem cells from human plucked hair: a further step towards autologous nerve recovery. Cytotechnology 2015; 68:1849-58. [PMID: 26702932 PMCID: PMC5023559 DOI: 10.1007/s10616-015-9938-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/11/2015] [Indexed: 12/11/2022] Open
Abstract
Stem cells from the adult hair follicle bulge can differentiate into neurons and glia, which is advantageous for the development of an autologous cell-based therapy for neurological diseases. Consequently, bulge stem cells from plucked hair may increase opportunities for personalized neuroregenerative therapy. Hairs were plucked from the scalps of healthy donors, and the bulges were cultured without prior tissue treatment. Shortly after outgrowth from the bulge, cellular protein expression was established immunohistochemically. The doubling time was calculated upon expansion, and the viability of expanded, cryopreserved cells was assessed after shear stress. The neuroglial differentiation potential was assessed from cryopreserved cells. Shortly after outgrowth, the cells were immunopositive for nestin, SLUG, AP-2α and SOX9, and negative for SOX10. Each bulge yielded approximately 1 × 104 cells after three passages. Doubling time was 3.3 (±1.5) days. Cellular viability did not differ significantly from control cells after shear stress. The cells expressed class III β-tubulin (TUBB3) and synapsin-1 after 3 weeks of neuronal differentiation. Glial differentiation yielded KROX20- and MPZ-immunopositive cells after 2 weeks. We demonstrated that human hair follicle bulge-derived stem cells can be cultivated easily, expanded efficiently and kept frozen until needed. After cryopreservation, the cells were viable and displayed both neuronal and glial differentiation potential.
Collapse
|
44
|
McMahill BG, Spriet M, Sisó S, Manzer MD, Mitchell G, McGee J, Garcia TC, Borjesson DL, Sieber-Blum M, Nolta JA, Sturges BK. Feasibility Study of Canine Epidermal Neural Crest Stem Cell Transplantation in the Spinal Cords of Dogs. Stem Cells Transl Med 2015; 4:1173-86. [PMID: 26273065 DOI: 10.5966/sctm.2015-0018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 06/17/2015] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED This pilot feasibility study aimed to determine the outcome of canine epidermal neural crest stem cell (cEPI-NCSC) grafts in the normal spinal cords of healthy bred-for-research dogs. This included developing novel protocols for (a) the ex vivo expansion of cEPI-NCSCs, (b) the delivery of cEPI-NCSCs into the spinal cord, and (c) the labeling of the cells and subsequent tracing of the graft in the live animal by magnetic resonance imaging. A total of four million cEPI-NCSCs were injected into the spinal cord divided in two locations. Differences in locomotion at baseline and post-treatment were evaluated by gait analysis and compared with neurological outcome and behavioral exams. Histopathological analyses of the spinal cords and cEPI-NCSC grafts were performed at 3 weeks post-transplantation. Neurological and gait parameters were minimally affected by the stem cell injection. cEPI-NCSCs survived in the canine spinal cord for the entire period of investigation and did not migrate or proliferate. Subsets of cEPI-NCSCs expressed the neural crest stem cell marker Sox10. There was no detectable expression of markers for glial cells or neurons. The tissue reaction to the cell graft was predominantly vascular in addition to a degree of reactive astrogliosis and microglial activation. In the present study, we demonstrated that cEPI-NCSC grafts survive in the spinal cords of healthy dogs without major adverse effects. They persist locally in the normal spinal cord, may promote angiogenesis and tissue remodeling, and elicit a tissue response that may be beneficial in patients with spinal cord injury. SIGNIFICANCE It has been established that mouse and human epidermal neural crest stem cells are somatic multipotent stem cells with proved innovative potential in a mouse model of spinal cord injury (SCI) offering promise of a valid treatment for SCI. Traumatic SCI is a common neurological problem in dogs with marked similarities, clinically and pathologically, to the syndrome in people. For this reason, dogs provide a readily accessible, clinically realistic, spontaneous model for evaluation of epidermal neural crest stem cells therapeutic intervention. The results of this study are expected to give the baseline data for a future clinical trial in dogs with traumatic SCI.
Collapse
Affiliation(s)
- Barbara G McMahill
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Mathieu Spriet
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Sílvia Sisó
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Michael D Manzer
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Gaela Mitchell
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Jeannine McGee
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Tanya C Garcia
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Dori L Borjesson
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Maya Sieber-Blum
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Jan A Nolta
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Beverly K Sturges
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
45
|
Vapniarsky N, Arzi B, Hu JC, Nolta JA, Athanasiou KA. Concise Review: Human Dermis as an Autologous Source of Stem Cells for Tissue Engineering and Regenerative Medicine. Stem Cells Transl Med 2015; 4:1187-98. [PMID: 26253713 DOI: 10.5966/sctm.2015-0084] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/08/2015] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED The exciting potential for regenerating organs from autologous stem cells is on the near horizon, and adult dermis stem cells (DSCs) are particularly appealing because of the ease and relative minimal invasiveness of skin collection. A substantial number of reports have described DSCs and their potential for regenerating tissues from mesenchymal, ectodermal, and endodermal lineages; however, the exact niches of these stem cells in various skin types and their antigenic surface makeup are not yet clearly defined. The multilineage potential of DSCs appears to be similar, despite great variability in isolation and in vitro propagation methods. Despite this great potential, only limited amounts of tissues and clinical applications for organ regeneration have been developed from DSCs. This review summarizes the literature on DSCs regarding their niches and the specific markers they express. The concept of the niches and the differentiation capacity of cells residing in them along particular lineages is discussed. Furthermore, the advantages and disadvantages of widely used methods to demonstrate lineage differentiation are considered. In addition, safety considerations and the most recent advancements in the field of tissue engineering and regeneration using DSCs are discussed. This review concludes with thoughts on how to prospectively approach engineering of tissues and organ regeneration using DSCs. Our expectation is that implementation of the major points highlighted in this review will lead to major advancements in the fields of regenerative medicine and tissue engineering. SIGNIFICANCE Autologous dermis-derived stem cells are generating great excitement and efforts in the field of regenerative medicine and tissue engineering. The substantial impact of this review lies in its critical coverage of the available literature and in providing insight regarding niches, characteristics, and isolation methods of stem cells derived from the human dermis. Furthermore, it provides analysis of the current state-of-the-art regenerative approaches using human-derived dermal stem cells, with consideration of current guidelines, to assist translation toward therapeutic use.
Collapse
Affiliation(s)
- Natalia Vapniarsky
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| | - Boaz Arzi
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| | - Jan A Nolta
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| |
Collapse
|
46
|
Sakaue M, Sieber-Blum M. Human epidermal neural crest stem cells as a source of Schwann cells. Development 2015; 142:3188-97. [PMID: 26251357 PMCID: PMC4582175 DOI: 10.1242/dev.123034] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 05/22/2015] [Indexed: 12/16/2022]
Abstract
We show that highly pure populations of human Schwann cells can be derived rapidly and in a straightforward way, without the need for genetic manipulation, from human epidermal neural crest stem cells [hEPI-NCSC(s)] present in the bulge of hair follicles. These human Schwann cells promise to be a useful tool for cell-based therapies, disease modelling and drug discovery. Schwann cells are glia that support axons of peripheral nerves and are direct descendants of the embryonic neural crest. Peripheral nerves are damaged in various conditions, including through trauma or tumour-related surgery, and Schwann cells are required for their repair and regeneration. Schwann cells also promise to be useful for treating spinal cord injuries. Ex vivo expansion of hEPI-NCSC isolated from hair bulge explants, manipulating the WNT, sonic hedgehog and TGFβ signalling pathways, and exposure of the cells to pertinent growth factors led to the expression of the Schwann cell markers SOX10, KROX20 (EGR2), p75NTR (NGFR), MBP and S100B by day 4 in virtually all cells, and maturation was completed by 2 weeks of differentiation. Gene expression profiling demonstrated expression of transcripts for neurotrophic and angiogenic factors, as well as JUN, all of which are essential for nerve regeneration. Co-culture of hEPI-NCSC-derived human Schwann cells with rodent dorsal root ganglia showed interaction of the Schwann cells with axons, providing evidence of Schwann cell functionality. We conclude that hEPI-NCSCs are a biologically relevant source for generating large and highly pure populations of human Schwann cells. Summary: Human epidermal neural crest stem cells isolated from the bulge of hair follicles are used to derive Schwann cells that could be useful for regenerative therapies, disease modelling and drug discovery.
Collapse
Affiliation(s)
- Motoharu Sakaue
- Institute of Genetic Medicine, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Maya Sieber-Blum
- Institute of Genetic Medicine, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| |
Collapse
|
47
|
Ribero S, Glass D, Aviv A, Spector TD, Bataille V. Height and bone mineral density are associated with naevus count supporting the importance of growth in melanoma susceptibility. PLoS One 2015; 10:e0116863. [PMID: 25612317 PMCID: PMC4303431 DOI: 10.1371/journal.pone.0116863] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 12/15/2014] [Indexed: 12/21/2022] Open
Abstract
Naevus count is the strongest risk factor for melanoma. Body Mass Index (BMI) has been linked to melanoma risk. In this study, we investigate the link between naevus count and height, weight and bone mineral density (BMD) in the TwinsUK cohort (N = 2119). In addition we adjusted for leucocyte telomere length (LTL) as LTL is linked to both BMD and naevus count. Naevus count was positively associated with height (p = 0.001) but not with weight (p = 0.187) despite adjusting for age and twin relatedness. This suggests that the previously reported melanoma association with BMI may be explained by height alone. Further adjustment for LTL did not affect the significance of the association between height and naevus count so LTL does not fully explain these results. BMD was associated with naevus count at the spine (coeff 18.9, p = 0.01), hip (coeff = 18.9, p = 0.03) and forearm (coeff = 32.7, p = 0.06) despite adjusting for age, twin relatedness, weight, height and LTL. This large study in healthy individuals shows that growth via height, probably in early life, and bone mass are risk factors for melanoma via increased naevus count. The link between these two phenotypes may possibly be explained by telomere biology, differentiation genes from the neural crests but also other yet unknown factors which may influence both bones and melanocytes biology.
Collapse
Affiliation(s)
- Simone Ribero
- Department of Twin Research & Genetic Epidemiology, King's College London, United Kingdom; Section of Dermatology, Departments of Medical Sciences, University of Turin, Turin, Italy
| | - Daniel Glass
- Department of Twin Research & Genetic Epidemiology, King's College London, United Kingdom; Dermatology Department, Northwick Park Hospital, Middlesex, United Kingdom
| | - Abraham Aviv
- Centre of Human Development and Ageing, University of Medicine and Dentistry of New Jersey, Newark, New Jersey, United States of America
| | - Timothy David Spector
- Department of Twin Research & Genetic Epidemiology, King's College London, United Kingdom
| | - Veronique Bataille
- Department of Twin Research & Genetic Epidemiology, King's College London, United Kingdom; Dermatology Department, West Herts NHS Trust, Herts, United Kingdom
| |
Collapse
|
48
|
Narytnyk A, Gillinder K, Verdon B, Clewes O, Sieber-Blum M. Neural crest stem cell-specific deletion of the Pygopus2 gene modulates hair follicle development. Stem Cell Rev Rep 2015; 10:60-8. [PMID: 23955574 PMCID: PMC3907677 DOI: 10.1007/s12015-013-9466-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We show that neural crest stem cells affect mouse hair follicle development. During embryogenesis hair follicle induction is regulated by complex reciprocal and functionally redundant signals between epidermis and dermis, which remain to be fully understood. Canonical Wnt signalling is a hallmark of neural crest cells and also a prerequisite for hair follicle induction prior to hair placode formation in the epidermis. As neural crest stem cells invade the epidermis during early embryonic development we aimed at determining whether neural crest cells affect hair follicle development. To attenuate, but not silence, canonical Wnt signalling specifically in neural crest cells, we analyzed Wnt1-cre(+/−)::Pygo2(−/−) mice in which the β-catenin co-activator gene, Pygopus 2 (Pygo2), is deleted specifically in neural crest cells. Both, hair density and hair thickness were reduced in mutant mice. Furthermore, hair development was delayed and the relative ratio of hair types was affected. There was a decrease in zig-zag hairs and an increase in awl hairs. Mouse neural crest stem cells expressed ectodysplasin, an essential effector in the formation of zig-zag hair. Taken together, our data support the novel notion that neural crest cells are involved in the earliest stages of hair follicle development.
Collapse
Affiliation(s)
- Alla Narytnyk
- Institute of Genetic Medicine, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | | | | | | | | |
Collapse
|
49
|
Differentiation of human epidermal neural crest stem cells (hEPI-NCSC) into virtually homogenous populations of dopaminergic neurons. Stem Cell Rev Rep 2014; 10:316-26. [PMID: 24399192 PMCID: PMC3969515 DOI: 10.1007/s12015-013-9493-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Here we provide a protocol for the directed differentiation of hEPI-NCSC into midbrain dopaminergic neurons, which degenerate in Parkinson's disease. hEPI-NCSC are neural crest-derived multipotent stem cells that persist into adulthood in the bulge of hair follicles. The experimental design is distinctly different from conventional protocols for embryonic stem cells and induced pluripotent stem (iPS) cells. It includes pre-differentiation of the multipotent hEPI-NCSC into neural stem cell-like cells, followed by ventralizing, patterning, continued exposure to the TGFβ receptor inhibitor, SB431542, and at later stages of differentiation the presence of the WNT inhibitor, IWP-4. All cells expressed A9 midbrain dopaminergic neuron progenitor markers with gene expression levels comparable to those in normal human substantia nigra. The current study shows for the first time that virtually homogeneous populations of dopaminergic neurons can be derived ex vivo from somatic stem cells without the need for purification, with useful timeliness and high efficacy. This novel development is an important first step towards the establishment of fully functional dopaminergic neurons from an ontologically relevant stem cell type, hEPI-NCSC.
Collapse
|
50
|
Ogliari KS, Marinowic D, Brum DE, Loth F. Stem cells in dermatology. An Bras Dermatol 2014; 89:286-91. [PMID: 24770506 PMCID: PMC4008060 DOI: 10.1590/abd1806-4841.20142530] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 04/08/2013] [Indexed: 01/27/2023] Open
Abstract
Preclinical and clinical research have shown that stem cell therapy could be a
promising therapeutic option for many diseases in which current medical treatments do
not achieve satisfying results or cure. This article describes stem cells sources and
their therapeutic applications in dermatology today.
Collapse
|