1
|
Ma Z, Liu T, Liu L, Pei Y, Wang T, Wang Z, Guan Y, Zhang X, Zhang Y, Chen X. Epidermal Neural Crest Stem Cell Conditioned Medium Enhances Spinal Cord Injury Recovery via PI3K/AKT-Mediated Neuronal Apoptosis Suppression. Neurochem Res 2024; 49:2854-2870. [PMID: 39023805 PMCID: PMC11365850 DOI: 10.1007/s11064-024-04207-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 04/19/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024]
Abstract
This study aimed to assess the impact of conditioned medium from epidermal neural crest stem cells (EPI-NCSCs-CM) on functional recovery following spinal cord injury (SCI), while also exploring the involvement of the PI3K-AKT signaling pathway in regulating neuronal apoptosis. EPI-NCSCs were isolated from 10-day-old Sprague-Dawley rats and cultured for 48 h to obtain EPI-NCSC-CM. SHSY-5Y cells were subjected with H2O2 treatment to induce apoptosis. Cell viability and survival rates were evaluated using the CCK-8 assay and calcein-AM/PI staining. SCI contusion model was established in adult Sprague-Dawley rats to assess functional recovery, utilizing the Basso, Beattie and Bresnahan (BBB) scoring system, inclined test, and footprint observation. Neurological restoration after SCI was analyzed through electrophysiological recordings. Histological analysis included hematoxylin and eosin (H&E) staining and Nissl staining to evaluate tissue organization. Apoptosis and oxidative stress levels were assessed using TUNEL staining and ROS detection methods. Additionally, western blotting was performed to examine the expression of apoptotic markers and proteins related to the PI3K/AKT signaling pathway. EPI-NCSC-CM significantly facilitated functional and histological recovery in SCI rats by inhibiting neuronal apoptosis through modulation of the PI3K/AKT pathway. Administration of EPI-NCSCs-CM alleviated H2O2-induced neurotoxicity in SHSY-5Y cells in vitro. The use of LY294002, a PI3K inhibitor, underscored the crucial role of the PI3K/AKT signaling pathway in regulating neuronal apoptosis. This study contributes to the ongoing exploration of molecular pathways involved in spinal cord injury (SCI) repair, focusing on the therapeutic potential of EPI-NCSC-CM. The research findings indicate that EPI-NCSC-CM exerts a neuroprotective effect by suppressing neuronal apoptosis through activation of the PI3K/AKT pathway in SCI rats. These results highlight the promising role of EPI-NCSC-CM as a potential treatment strategy for SCI, emphasizing the significance of the PI3K/AKT pathway in mediating its beneficial effects.
Collapse
Affiliation(s)
- Ziqian Ma
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, 8 Workers Stadium South Road, Chaoyang District, Beijing, China
| | - Tao Liu
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Liang Liu
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yilun Pei
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Tianyi Wang
- Department of Orthopedics, 981st Hospital of the Chinese People's Liberation Army Joint Logistics Support Force, Chengde, 067000, Hebei Province, P.R. China
| | - Zhijie Wang
- Department of Pediatric Internal Medicine, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurological Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xinwei Zhang
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yan Zhang
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| | - Xueming Chen
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Hooshmandi E, Akbari S, Pandamooz S, Ghobadi M, Ghasemi R, Maghsoudi N, Rai SN, Borhani-Haghighi A, Salehi MS, Azarpira N, YousefiNejad A, Haghani M, Bayat M. Combined use of hair follicle stem cells and CEPO (carbamylated erythropoietin)-Fc in a rat model of chronic cerebral hypoperfusion: A behavioral, electrophysiological, and molecular study. Behav Brain Res 2023; 454:114655. [PMID: 37666305 DOI: 10.1016/j.bbr.2023.114655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/22/2023] [Accepted: 08/31/2023] [Indexed: 09/06/2023]
Abstract
BACKGROUND In dementia, synaptic dysfunction appears before neuronal loss. Stem cell therapy could potentially provide a promising strategy for the treatment of dementia models. The carbamylated erythropoietin fusion protein (CEPO-Fc) has shown synaptotrophic effects. This study aimed to determine the efficiency of the combined use of hair follicle stem cells (HFSC) and CEPO-Fc in the basal synaptic transmission (BST) and long-term plasticity (LTP) of chronic cerebral hypoperfusion (CCH) rats. METHODS We divided 64 adult rats into control, sham, CCH+vehicle, CCH+CEPO, CCH+HFSC, and CCH+HFSC+CEPO groups. The CEPO-Fc was injected three times/week for 30 days. HFSC transplantation was done on days 4, 14, and 21 after surgery. The Morris water maze test and passive avoidance were used to assess memory. BST and LTP were assessed by a field-potential recording of the CA1 region. The hippocampal mRNA expression of IGF-1, TGF-β1, β1-Catenine, NR2B, PSD-95, and GSk-3β was evaluated by quantitative RT-PCR. RESULTS Following combination therapy, spatial memory retention, and BST showed significant improvement relative to HFSC and CEPO-Fc groups. These effects were also confirmed by recovered mRNA expression of β1-catenin, TGF-β1, and NR2B. GSK-3β expression was downregulated in all treatment groups. The upregulated PSD-95 was identified in HFSC and combination groups compared to the vehicle group. CONCLUSIONS These findings indicate that the combined use of HFSC and CEPO-Fc may be more advantageous for treating memory disruption in the CCH model than CEPO-Fc or HFSC alone. This type of combination therapy may hopefully lead to a new approach to treatment for dementia.
Collapse
Affiliation(s)
- Etrat Hooshmandi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Somayeh Akbari
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran; Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Mojtaba Ghobadi
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Rasoul Ghasemi
- Neurophysiology Research Center and Physiology Department, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Nader Maghsoudi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | | | - Afshin Borhani-Haghighi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Mohammad Rasoul-Allah Research Tower, Shiraz, the Islamic Republic of Iran
| | - Amirhossein YousefiNejad
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Masoud Haghani
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran; Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Mahnaz Bayat
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran.
| |
Collapse
|
3
|
Stüfchen I, Beyer F, Staebler S, Fischer S, Kappelmann M, Beckervordersandforth R, Bosserhoff AK. Sox9 regulates melanocytic fate decision of adult hair follicle stem cells. iScience 2023; 26:106919. [PMID: 37283806 PMCID: PMC10239701 DOI: 10.1016/j.isci.2023.106919] [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] [Received: 09/24/2022] [Revised: 03/02/2023] [Accepted: 05/14/2023] [Indexed: 06/08/2023] Open
Abstract
The bulge of hair follicles harbors Nestin+ (neural crest like) stem cells, which exhibit the potential to generate various cell types including melanocytes. In this study, we aimed to determine the role of Sox9, an important regulator during neural crest development, in melanocytic differentiation of those adult Nestin+ cells. Immunohistochemical analysis after conditional Sox9 deletion in Nestin+ cells of adult mice revealed that Sox9 is crucial for melanocytic differentiation of these cells and that Sox9 acts as a fate determinant between melanocytic and glial fate. A deeper understanding of factors that regulate fate decision, proliferation and differentiation of these stem cells provides new aspects to melanoma research as melanoma cells share many similarities with neural crest cells. In summary, we here show the important role of Sox9 in melanocytic versus glial fate decision of Nestin+ stem cells in the skin of adult mice.
Collapse
Affiliation(s)
- Isabel Stüfchen
- Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Felix Beyer
- Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian Staebler
- Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Fischer
- Faculty of Computer Science, Deggendorf Institute of Technology, Deggendorf, Germany
| | - Melanie Kappelmann
- Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Faculty of Computer Science, Deggendorf Institute of Technology, Deggendorf, Germany
| | | | - Anja K. Bosserhoff
- Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
4
|
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
|
5
|
Abstract
Pathological hair loss (also known as alopecia) and shortage of hair follicle (HF) donors have posed an urgent requirement for HF regeneration. With the revelation of mechanisms in tissue engineering, the proliferation of HFs in vitro has achieved more promising trust for the treatments of alopecia and other skin impairments. Theoretically, HF organoids have great potential to develop into native HFs and attachments such as sweat glands after transplantation. However, since the rich extracellular matrix (ECM) deficiency, the induction characteristics of skin-derived cells gradually fade away along with their trichogenic capacity after continuous cell passaging in vitro. Therefore, ECM-mimicking support is an essential prelude before HF transplantation is implemented. This review summarizes the status of providing various epidermal and dermal cells with a three-dimensional (3D) scaffold to support the cell homeostasis and better mimic in vivo environments for the sake of HF regeneration. HF-relevant cells including dermal papilla cells (DPCs), hair follicle stem cells (HFSCs), and mesenchymal stem cells (MSCs) are able to be induced to form HF organoids in the vitro culture system. The niche microenvironment simulated by different forms of biomaterial scaffold can offer the cells a network of ordered growth environment to alleviate inductivity loss and promote the expression of functional proteins. The scaffolds often play the role of ECM substrates and bring about epithelial-mesenchymal interaction (EMI) through coculture to ensure the functional preservation of HF cells during in vitro passage. Functional HF organoids can be formed either before or after transplantation into the dermis layer. Here, we review and emphasize the importance of 3D culture in HF regeneration in vitro. Finally, the latest progress in treatment trials and critical analysis of the properties and benefits of different emerging biomaterials for HF regeneration along with the main challenges and prospects of HF regenerative approaches are discussed.
Collapse
Affiliation(s)
- Wei Zheng
- College of Food Science & Technology, Shanghai Ocean University, Shanghai 201306, P.R. China
| | - Chang-Hua Xu
- College of Food Science & Technology, Shanghai Ocean University, Shanghai 201306, P.R. China
- Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai 201306, China
- Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, Shanghai 201306, China
- National R&D Branch Center for Freshwater Aquatic Products Processing Technology (Shanghai), Shanghai 201306, China
| |
Collapse
|
6
|
Pandamooz S, Jurek B, Dianatpour M, Haerteis S, Limm K, Oefner PJ, Dargahi L, Borhani-Haghighi A, Miyan JA, Salehi MS. The beneficial effects of chick embryo extract preconditioning on hair follicle stem cells: A promising strategy to generate Schwann cells. Cell Prolif 2023:e13397. [PMID: 36631409 DOI: 10.1111/cpr.13397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 12/06/2022] [Accepted: 12/28/2022] [Indexed: 01/13/2023] Open
Abstract
The beneficial effects of hair follicle stem cells in different animal models of nervous system conditions have been extensively studied. While chick embryo extract (CEE) has been used as a growth medium supplement for these stem cells, this is the first study to show the effect of CEE on them. The rat hair follicle stem cells were isolated and supplemented with 10% fetal bovine serum plus 10% CEE. The migration rate, proliferative capacity and multipotency were evaluated along with morphometric alteration and differentiation direction. The proteome analysis of CEE content identified effective factors of CEE that probably regulate fate and function of stem cells. The CEE enhances the migration rate of stem cells from explanted bulges as well as their proliferation, likely due to activation of AP-1 and translationally controlled tumour protein (TCTP) by thioredoxin found in CEE. The increased length of outgrowth may be the result of cyclic AMP response element binding protein (CREB) phosphorylation triggered by active CamKII contained in CEE. Further, CEE supplementation upregulates the expression of vascular endothelial growth factor (VEGF), brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor. The elevated expression of target genes and proteins may be due to CREB, AP-1 and c-Myc activation in these stem cells. Given the increased transcript levels of neurotrophins, VEGF, and the expression of PDGFR-α, S100B, MBP and SOX-10 protein, it is possible that CEE promotes the fate of these stem cells towards Schwann cells.
Collapse
Affiliation(s)
- Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benjamin Jurek
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany.,Institute of Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
| | - Mehdi Dianatpour
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Silke Haerteis
- Institute of Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
| | - Katharina Limm
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Jaleel A Miyan
- Faculty of Biology, Medicine & Health, Division of Neuroscience & Experimental Psychology, The University of Manchester, Manchester, UK
| | - Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Hörner SJ, Couturier N, Gueiber DC, Hafner M, Rudolf R. Development and In Vitro Differentiation of Schwann Cells. Cells 2022; 11:3753. [PMID: 36497014 PMCID: PMC9739763 DOI: 10.3390/cells11233753] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
Schwann cells are glial cells of the peripheral nervous system. They exist in several subtypes and perform a variety of functions in nerves. Their derivation and culture in vitro are interesting for applications ranging from disease modeling to tissue engineering. Since primary human Schwann cells are challenging to obtain in large quantities, in vitro differentiation from other cell types presents an alternative. Here, we first review the current knowledge on the developmental signaling mechanisms that determine neural crest and Schwann cell differentiation in vivo. Next, an overview of studies on the in vitro differentiation of Schwann cells from multipotent stem cell sources is provided. The molecules frequently used in those protocols and their involvement in the relevant signaling pathways are put into context and discussed. Focusing on hiPSC- and hESC-based studies, different protocols are described and compared, regarding cell sources, differentiation methods, characterization of cells, and protocol efficiency. A brief insight into developments regarding the culture and differentiation of Schwann cells in 3D is given. In summary, this contribution provides an overview of the current resources and methods for the differentiation of Schwann cells, it supports the comparison and refinement of protocols and aids the choice of suitable methods for specific applications.
Collapse
Affiliation(s)
- Sarah Janice Hörner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Nathalie Couturier
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Daniele Caroline Gueiber
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Department of Electronics Engineering, Federal University of Technology Paraná, Ponta Grossa 84017-220, Brazil
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| |
Collapse
|
8
|
Stüfchen I, Beckervordersandforth R, Fischer S, Kappelmann M, Bosserhoff AK, Beyer F. Two novel CreER T2 transgenic mouse lines to study melanocytic cells in vivo. Pigment Cell Melanoma Res 2022; 35:613-621. [PMID: 35920064 DOI: 10.1111/pcmr.13061] [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: 09/01/2021] [Revised: 05/20/2022] [Accepted: 08/01/2022] [Indexed: 11/29/2022]
Abstract
The skin of adult mammals protects from radiation, physical and chemical insults. While melanocytes and melanocyte-producing stem cells contribute to proper skin function in healthy organisms, dysfunction of these cells can lead to the generation of malignant melanoma - the deadliest type of skin cancer. Addressing cells of the melanocyte lineage in vivo represents a prerequisite for the understanding of melanoma on cellular level and the development of preventive and treatment strategies. Here, the inducible Cre-loxP-system has emerged as a promising tool to specifically target, monitor and modulate cells in adult mice. Re-analysis of existing sequencing data sets of melanocytic cells revealed that genes with a known function in neural cells, including neural stem cells (Aldh1L1 and Nestin), are also expressed in melanocytic cells. Therefore, in this study we explored whether the promoter activity of Nestin and Aldh1L1 can serve to target cells of the melanocyte lineage using the inducible CreERT2 -loxP-system. Using an immunohistochemical approach and different time-points of analysis, we were able to map the melanocytic fate of recombined stem cells in the adult hair follicle of Nestin-CreERT2 and Aldh1L1-CreERT2 transgenic mice. Thus, we here present two new mouse models and propose their use to study and putatively modulate adult melanocytic cells in vivo.
Collapse
Affiliation(s)
- Isabel Stüfchen
- Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | | | - Stefan Fischer
- Faculty of Computer Science, Deggendorf Institute of Technology, Deggendorf, Germany
| | - Melanie Kappelmann
- Faculty of Computer Science, Deggendorf Institute of Technology, Deggendorf, Germany
| | - Anja K Bosserhoff
- Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Felix Beyer
- Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
9
|
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
|
10
|
Erdoğan A, Mutlu HS, Solakoğlu S. Autologously transplanted dermis-derived cells alleviated monobenzone-induced vitiligo in mouse. Exp Dermatol 2022; 31:1355-1363. [PMID: 35538739 DOI: 10.1111/exd.14603] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/26/2022] [Accepted: 05/08/2022] [Indexed: 12/01/2022]
Abstract
Vitiligo is a depigmentation disease which affects skin and hair follicles with a prevalence of 0.5-1% worldwide. In this study, we aimed to investigate treatmental potential of dermis-derived cells in monobenzone (MBEH)-induced mouse vitiligo model with light and electron microscopy. MBEH (40%) cream was topically applied to C57BL/6 mice until depigmentation occured in vitiligo and experimental groups. In experimental groups, dermis-derived cells obtained from back skin biopsy samples before induction of vitiligo, were injected intradermally to vitiligo mice. On day 3 and 15 after cell transplantation to experimental groups, skin biopsies were compared with biopsies of control and vitiligo groups. Dermis-derived cells obtained from back skin biopsy samples of experimental groups showed nestin and versican immunoreactivity. Melanin in hair follicles of control group was detected by histochemical stainings (Hematoxylin&Eosin and Fontana-Masson) whereas sparse melanin granules were observed in hair follicles of vitiligo group. In experimental groups, there was an increase in the number of hair follicles with melanin compared to vitiligo group. We observed MART-1 immunoreactive cells mostly around the hair follicles in control group and within dermis in vitiligo group. Electron microscopic investigation showed presence of melanosomes in hair follicles of control group and lacking in vitiligo group. In experimental groups, both type of hair follicles were observed with electron microscope. Our data suggest that autologously transplanted dermis-derived cells may be effective in vitiligo treatment by contrubuting to melanin production.
Collapse
Affiliation(s)
- Aslı Erdoğan
- İstanbul University, İstanbul Faculty of Medicine, Department of Histology and Embryology, İstanbul, Turkey.,İstanbul University, Graduate School of Health Sciences, İstanbul, Turkey.,İzmir Kȃtip Çelebi University, Faculty of Medicine, Department of Histology and Embryology, İzmir, Turkey
| | - Hasan Serdar Mutlu
- İstanbul University, İstanbul Faculty of Medicine, Department of Histology and Embryology, İstanbul, Turkey.,Giresun University, Faculty of Medicine, Department of Histology and Embryology, Giresun, Turkey
| | - Seyhun Solakoğlu
- İstanbul University, İstanbul Faculty of Medicine, Department of Histology and Embryology, İstanbul, Turkey
| |
Collapse
|
11
|
ECM1 modified HF-MSCs targeting HSC attenuate liver cirrhosis by inhibiting the TGF-β/Smad signaling pathway. Cell Death Dis 2022; 8:51. [PMID: 35136027 PMCID: PMC8827057 DOI: 10.1038/s41420-022-00846-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/05/2022] [Accepted: 01/20/2022] [Indexed: 12/13/2022]
Abstract
Hair follicle-derived mesenchymal stem cells (HF-MSCs) show considerable therapeutic potential for liver cirrhosis (LC). To improve the effectiveness of naïve HF-MSC treatments on LC, we used bioinformatic tools to identify an exogenous gene targeting HSCs among the differentially expressed genes (DEGs) in LC to modify HF-MSCs. Extracellular matrix protein 1 (ECM1) was identified as a DEG that was significantly downregulated in the cirrhotic liver. Then, ECM1-overexpressing HF-MSCs (ECM1-HF-MSCs) were transplanted into mice with LC to explore the effectiveness and correlated mechanism of gene-overexpressing HF-MSCs on LC. The results showed that ECM1-HF-MSCs significantly improved liver function and liver pathological injury in LC after cell therapy relative to the other treatment groups. Moreover, we found that ECM1-HF-MSCs homed to the injured liver and expressed the hepatocyte-specific surface markers ALB, CK18, and AFP. In addition, hepatic stellate cell (HSC) activation was significantly inhibited in the cell treatment groups in vivo and in vitro, especially in the ECM1-HF-MSC group. Additionally, TGF-β/Smad signal inhibition was the most significant in the ECM1-HF-MSC group in vivo and in vitro. The findings indicate that the genetic modification of HF-MSCs with bioinformatic tools may provide a broad perspective for precision treatment of LC.
Collapse
|
12
|
Li H, Ziemer M, Stojanovic I, Saksida T, Maksimovic-Ivanic D, Mijatovic S, Djmura G, Gajic D, Koprivica I, Krajnovic T, Draca D, Simon JC, Lethaus B, Savkovic V. Mesenchymal Stem Cells From Mouse Hair Follicles Reduce Hypertrophic Scarring in a Murine Wound Healing Model. Stem Cell Rev Rep 2022; 18:2028-2044. [PMID: 35080748 PMCID: PMC9391240 DOI: 10.1007/s12015-021-10288-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2021] [Indexed: 12/11/2022]
Abstract
Wound healing of acute full-thickness injuries and chronic non-healing ulcers leads to delayed wound closure, prolonged recovery period and hypertrophic scarring, generating a demand for an autologous cell therapy and a relevant pre-clinical research models for wound healing. In this study, an immunocompetent model for wound healing was employed using a syngeneic murine cell line of mesenchymal stem cells cultured from the mouse whisker hair follicle outer root sheath (named moMSCORS). moMSCORS were isolated using an air-liquid interface method, expanded in vitro and characterized according to the MSC definition criteria - cell viability, in vitro proliferation, MSC phenotype and multi-lineage differentiations. Moreover, upon applying moMSCORS in an in vivo full-thickness wound model in the syngeneic C57BL/6 mice, the treated wounds displayed different morphology to that of the untreated wound beds. Quantitative evaluation of angiogenesis, granulation and wound closure involving clinical scoring and software-based quantification indicated a lower degree of inflammation in the treated wounds. Histological staining of treated wounds by the means of H&E, Alcian Blue, PicroSirius Red and αSMA immune labelling showed lower cellularity, less collagen filaments as well as thinner dermal and epidermal layers compared with the untreated wounds, indicating a general reduction of hypertrophic scars. The decreased inflammation, accelerated wound closure and non-hypertrophic scarring, which were facilitated by moMSCORS, hereby address a common problem of hypertrophic scars and non-physiological tissue properties upon wound closure, and additionally offer an in vivo model for the autologous cell-based wound healing.
Collapse
Affiliation(s)
- Hanluo Li
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Provincial Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, 430068, Hubei Province, China.,Department of Cranial Maxillofacial Plastic Surgery, University Clinic Leipzig, 04103, Leipzig, Germany
| | - Mirjana Ziemer
- Clinic for Dermatology, Venereology and Allergology, University Hospital Leipzig, 04103, Leipzig, Germany
| | - Ivana Stojanovic
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Saksida
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Danijela Maksimovic-Ivanic
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Sanja Mijatovic
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Goran Djmura
- Clinic for Dermatology, Venereology and Allergology, University Hospital Leipzig, 04103, Leipzig, Germany
| | - Dragica Gajic
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ivan Koprivica
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Krajnovic
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Dijana Draca
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jan-Christoph Simon
- Clinic for Dermatology, Venereology and Allergology, University Hospital Leipzig, 04103, Leipzig, Germany
| | - Bernd Lethaus
- Department of Cranial Maxillofacial Plastic Surgery, University Clinic Leipzig, 04103, Leipzig, Germany
| | - Vuk Savkovic
- Department of Cranial Maxillofacial Plastic Surgery, University Clinic Leipzig, 04103, Leipzig, Germany.
| |
Collapse
|
13
|
Cerrizuela S, Vega-Lopez GA, Méndez-Maldonado K, Velasco I, Aybar MJ. The crucial role of model systems in understanding the complexity of cell signaling in human neurocristopathies. WIREs Mech Dis 2022; 14:e1537. [PMID: 35023327 DOI: 10.1002/wsbm.1537] [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: 03/30/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 11/07/2022]
Abstract
Animal models are useful to study the molecular, cellular, and morphogenetic mechanisms underlying normal and pathological development. Cell-based study models have emerged as an alternative approach to study many aspects of human embryonic development and disease. The neural crest (NC) is a transient, multipotent, and migratory embryonic cell population that generates a diverse group of cell types that arises during vertebrate development. The abnormal formation or development of the NC results in neurocristopathies (NCPs), which are characterized by a broad spectrum of functional and morphological alterations. The impaired molecular mechanisms that give rise to these multiphenotypic diseases are not entirely clear yet. This fact, added to the high incidence of these disorders in the newborn population, has led to the development of systematic approaches for their understanding. In this article, we have systematically reviewed the ways in which experimentation with different animal and cell model systems has improved our knowledge of NCPs, and how these advances might contribute to the development of better diagnostic and therapeutic tools for the treatment of these pathologies. This article is categorized under: Congenital Diseases > Genetics/Genomics/Epigenetics Congenital Diseases > Stem Cells and Development Congenital Diseases > Molecular and Cellular Physiology Neurological Diseases > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Santiago Cerrizuela
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina
| | - Guillermo A Vega-Lopez
- Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina.,Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Karla Méndez-Maldonado
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.,Departamento de Fisiología y Farmacología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.,Laboratorio de Reprogramación Celular del Instituto de Fisiología Celular, UNAM en el Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Ciudad de México, Mexico
| | - Manuel J Aybar
- Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina.,Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| |
Collapse
|
14
|
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
|
15
|
Peripheral Nerve Regeneration Using Different Germ Layer-Derived Adult Stem Cells in the Past Decade. Behav Neurol 2021; 2021:5586523. [PMID: 34539934 PMCID: PMC8448597 DOI: 10.1155/2021/5586523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/27/2021] [Accepted: 08/09/2021] [Indexed: 12/15/2022] Open
Abstract
Peripheral nerve injuries (PNIs) are some of the most common types of traumatic lesions affecting the nervous system. Although the peripheral nervous system has a higher regenerative ability than the central nervous system, delayed treatment is associated with disturbances in both distal sensory and functional abilities. Over the past decades, adult stem cell-based therapies for peripheral nerve injuries have drawn attention from researchers. This is because various stem cells can promote regeneration after peripheral nerve injuries by differentiating into neural-line cells, secreting various neurotrophic factors, and regulating the activity of in situ Schwann cells (SCs). This article reviewed research from the past 10 years on the role of stem cells in the repair of PNIs. We concluded that adult stem cell-based therapies promote the regeneration of PNI in various ways.
Collapse
|
16
|
Olszewski C, Maassen J, Guenther R, Skazik-Voogt C, Gutermuth A. Mechanotransductive Differentiation of Hair Follicle Stem Cells Derived from Aged Eyelid Skin into Corneal Endothelial-Like Cells. Stem Cell Rev Rep 2021; 18:1668-1685. [PMID: 34515937 PMCID: PMC9209348 DOI: 10.1007/s12015-021-10249-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2021] [Indexed: 11/25/2022]
Abstract
Corneal endothelial insufficiency is one of the leading causes of blindness. The main contemporary treatment for corneal blindness is endothelial keratoplasty, which, however, is unsatisfactory as a medical therapy due to the lack of donor corneas and graft rejection. Therefore, autologous stem cell-based corneal endothelial tissue substitutes may be a promising alternative to conventional grafts in the future. To address the age of most patients suffering from corneal endothelial deficiencies, we investigated the presence and potential of hair-derived stem cells from older tissue donors. Our studies revealed the presence of pluripotency- and neural crest-associated markers in tissue sections from blepharoplasty patients aged 50 to 80 years. In vitro outgrowths from eyelid hair follicles on collagen-coated tissue culture plates revealed a weak decrease in stem-cell potency. In contrast, cells within the spheres that spontaneously formed from the adherent cell layer retained full stem-cell potency and could be differentiated into cells of the ecto- meso and endodermal lineages. Although these highly potent hair follicle derived stem cells (HFSC) were only very slightly expandable, they were able to recognize the biomimicry of the Descemet’s-like topography and differentiate into corneal endothelial-like cells. In conclusion, HFSCs derived from epidermal skin of eyelid biopsies are a promising cell source to provide autologous corneal endothelial replacement for any age group of patients.
Collapse
Affiliation(s)
- Christian Olszewski
- Fraunhofer Institute for Production Technology, Steinbachstraße 17, 52074, Aachen, Germany
| | - Jessika Maassen
- Fraunhofer Institute for Production Technology, Steinbachstraße 17, 52074, Aachen, Germany
| | - Rebecca Guenther
- Fraunhofer Institute for Production Technology, Steinbachstraße 17, 52074, Aachen, Germany
| | - Claudia Skazik-Voogt
- Fraunhofer Institute for Production Technology, Steinbachstraße 17, 52074, Aachen, Germany
| | - Angela Gutermuth
- Fraunhofer Institute for Production Technology, Steinbachstraße 17, 52074, Aachen, Germany.
| |
Collapse
|
17
|
Saha D, Thannimangalath S, Budamakuntla L, Loganathan E, Jamora C. Hair Follicle Grafting Therapy Promotes Re-Emergence of Critical Skin Components in Chronic Nonhealing Wounds. JID INNOVATIONS 2021; 1:100041. [PMID: 34909736 PMCID: PMC8659399 DOI: 10.1016/j.xjidi.2021.100041] [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] [Received: 03/30/2021] [Revised: 05/26/2021] [Accepted: 06/11/2021] [Indexed: 12/03/2022] Open
Abstract
An exploding public health crisis is the exponential growth in the incidence of chronic nonhealing ulcers associated with diseases such as diabetes. Various modalities have been developed to stimulate wound closure that is otherwise recalcitrant to standard clinical treatments. However, these approaches primarily focus on the process of re-epithelialization and are often deficient in regenerating the full spectrum of structures necessary for normal skin function. Autologous hair follicle grafting is a recent therapy to stimulate the closure of such nonhealing wounds, and we observed effects beyond the epidermis to other important components of the dermis. We found that hair follicle grafting facilitated the reappearance of various undifferentiated and differentiated layers of the epidermis with the restoration of epidermal junctions. In addition, other important structures that are critical for cutaneous health and function such as the blood and lymph vasculature, nerve fibers, and sweat gland structures were restored in postgrafted wounds. Interestingly, both immune cells and inflammatory signals were substantially decreased, indicating a reduction in the chronic inflammation that is a hallmark of nonhealing wounds. Our observation that punch wounds created on the postgrafted area likewise healed suggests that this is a self-sustaining long-term therapy for patients with chronic wounds.
Collapse
Affiliation(s)
- Dyuti Saha
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
- Department of Biology, Manipal Academy of Higher Education, Manipal, India
| | - Sujaya Thannimangalath
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Leelavathy Budamakuntla
- Department of Dermatology, Bangalore Medical College and Research Institute, Bangalore, India
| | - Eswari Loganathan
- Department of Dermatology, Bangalore Medical College and Research Institute, Bangalore, India
| | - Colin Jamora
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| |
Collapse
|
18
|
Peterson A, Nair L. Hair Follicle Stem Cells for Tissue Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:695-706. [PMID: 34238037 PMCID: PMC9419938 DOI: 10.1089/ten.teb.2021.0098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
With the positive outcomes of various cell therapies currently under pre-clinical and clinical studies, there is a significant interest in novel stem cell sources with unique therapeutic properties. Studies over the past two decades or so demonstrated the feasibility to isolate multipotent/pluripotent stem cells from hair follicles. The easy accessibility, high proliferation and differentiation ability as well as lack of ethical concerns associated with this stem cell source make hair follicle stem cells (HFSCs) attractive candidate for cell therapy and tissue engineering. This review discusses the various stem cell types identified in rodent and human hair follicles and ongoing studies on the potential use of HFSCs for skin, bone, cardio-vascular, and nerve tissue engineering.
Collapse
Affiliation(s)
- Alyssa Peterson
- University of Connecticut, 7712, Storrs, Connecticut, United States;
| | - Lakshmi Nair
- University of Connecticut Health Center, 21654, Orthopaedic Surgery, Farmington, Connecticut, United States;
| |
Collapse
|
19
|
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
|
20
|
Tian DH, Qin CH, Xu WY, Pan WK, Zhao YY, Zheng BJ, Chen XL, Liu Y, Gao Y, Yu H. Phenotypic and functional comparison of rat enteric neural crest-derived cells during fetal and early-postnatal stages. Neural Regen Res 2021; 16:2310-2315. [PMID: 33818517 PMCID: PMC8354115 DOI: 10.4103/1673-5374.310701] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In our previous study, we showed that with increasing time in culture, the growth characteristics of enteric neural crest-derived cells (ENCCs) change, and that the proliferation, migration and neural differentiation potential of these cells in vitro notably diminish. However, there are no studies on the developmental differences in these characteristics between fetal and early-postnatal stages in vitro or in vivo. In this study, we isolated fetal (embryonic day 14.5) and postnatal (postnatal day 2) ENCCs from the intestines of rats. Fetal ENCCs had greater maximum cross-sectional area of the neurospheres, stronger migration ability, and reduced apoptosis, compared with postnatal ENCCs. However, fetal and postnatal ENCCs had a similar differentiation ability. Fetal and postnatal ENCCs both survived after transplant into a rat model of Hirschsprung's disease. In these rats with Hirschsprung's disease, the number of ganglionic cells in the myenteric plexus was higher and the distal intestinal pressure change was greater in animals treated with fetal ENCCs compared with those treated with postnatal ENCCs. These findings suggest that, compared with postnatal ENCCs, fetal ENCCs exhibit higher survival and proliferation and migration abilities, and are therefore a more appropriate seed cell for the treatment of Hirschsprung's disease. This study was approved by the Animal Ethics Committee of the Second Affiliated Hospital of Xi'an Jiaotong University (approval No. 2016086) on March 3, 2016.
Collapse
Affiliation(s)
- Dong-Hao Tian
- Department of Pediatric Surgery, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Chuan-Hui Qin
- Department of Anorectal, Suizhou Central Hospital, Hubei University of Medicine, Suizhou, Hubei Province, China
| | - Wen-Yao Xu
- Department of Pediatric Surgery, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Wei-Kang Pan
- Department of Pediatric Surgery, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yu-Ying Zhao
- Department of Pediatric Surgery, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Bai-Jun Zheng
- Department of Pediatric Surgery, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Xin-Lin Chen
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Chinese Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yong Liu
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Chinese Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Ya Gao
- Department of Pediatric Surgery, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Hui Yu
- Department of Pediatric Surgery, the Second Affiliated Hospital; Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Chinese Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| |
Collapse
|
21
|
Ahmadi S, Nabiuni M, Tahmaseb M, Amini E. Enhanced Neural Differentiation of Epidermal Neural Crest Stem Cell by Synergistic Effect of Lithium carbonate and Crocin on BDNF and GDNF Expression as Neurotrophic Factors. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:95-106. [PMID: 34567149 PMCID: PMC8457715 DOI: 10.22037/ijpr.2019.15561.13176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Neurodegenerative diseases are incurable and debilitating conditions that result in progressive degeneration of nerve cells. Due to the complexity of conditions in neurodegenerative diseases, combination therapy, including cell and drug therapy is important as a new therapeutic strategy. Epidermal neural crest stem cells (EPI-NCSCs) are among the best choices in cell therapy for various neurological diseases. In this study, the effect of Lithium carbonate and Crocin, considering their effects on cellular signaling pathways and neuroprotective properties were investigated on the expression of neurotrophic factors BDNF and GDNF in EPI-NCSCs. EPI-NCSCs were isolated from the hair follicle and treated with different concentrations of drugs [Lithium, Crocin, and lithium + Crocin] for 72h. Then, trial concentrations were selected by MTT assay. The cells were treated with selected concentrations (Lithium 1 mM, Crocin 1.5 mM, and for co-treatment Lithium 1 mM and Crocin 1 mM) for 7 days. The Real-Time PCR results indicated an increasing in expression of BDNF and GDNF in treated cells as compared with control (* p < 0.05, ** p < 0.01 and *** p < 0.001). The results in this study confirmed and supported the neuroprotective/neurogenesis effects of Lithium and Crocin. It also showed that the proposed protocol could be used to increase EPI-NCSCs differentiation potential into neural cells in cell therapy and combination therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Shirin Ahmadi
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Mohammad Nabiuni
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Mohammad Tahmaseb
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Elaheh Amini
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| |
Collapse
|
22
|
Vanzo RJ, Prasad A, Staunch L, Hensel CH, Serrano MA, Wassman ER, Kaplun A, Grandin T, Boles RG. The Temple Grandin Genome: Comprehensive Analysis in a Scientist with High-Functioning Autism. J Pers Med 2020; 11:21. [PMID: 33383702 PMCID: PMC7824360 DOI: 10.3390/jpm11010021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/31/2022] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous condition with a complex genetic etiology. The objective of this study is to identify the complex genetic factors that underlie the ASD phenotype and other clinical features of Professor Temple Grandin, an animal scientist and woman with high-functioning ASD. Identifying the underlying genetic cause for ASD can impact medical management, personalize services and treatment, and uncover other medical risks that are associated with the genetic diagnosis. Prof. Grandin underwent chromosomal microarray analysis, whole exome sequencing, and whole genome sequencing, as well as a comprehensive clinical and family history intake. The raw data were analyzed in order to identify possible genotype-phenotype correlations. Genetic testing identified variants in three genes (SHANK2, ALX1, and RELN) that are candidate risk factors for ASD. We identified variants in MEFV and WNT10A, reported to be disease-associated in previous studies, which are likely to contribute to some of her additional clinical features. Moreover, candidate variants in genes encoding metabolic enzymes and transporters were identified, some of which suggest potential therapies. This case report describes the genomic findings in Prof. Grandin and it serves as an example to discuss state-of-the-art clinical diagnostics for individuals with ASD, as well as the medical, logistical, and economic hurdles that are involved in clinical genetic testing for an individual on the autism spectrum.
Collapse
Affiliation(s)
- Rena J. Vanzo
- Lineagen, Inc., Salt Lake City, UT 84109, USA; (A.P.); (L.S.); (C.H.H.); (M.A.S.); (E.R.W.)
| | - Aparna Prasad
- Lineagen, Inc., Salt Lake City, UT 84109, USA; (A.P.); (L.S.); (C.H.H.); (M.A.S.); (E.R.W.)
| | - Lauren Staunch
- Lineagen, Inc., Salt Lake City, UT 84109, USA; (A.P.); (L.S.); (C.H.H.); (M.A.S.); (E.R.W.)
| | - Charles H. Hensel
- Lineagen, Inc., Salt Lake City, UT 84109, USA; (A.P.); (L.S.); (C.H.H.); (M.A.S.); (E.R.W.)
| | - Moises A. Serrano
- Lineagen, Inc., Salt Lake City, UT 84109, USA; (A.P.); (L.S.); (C.H.H.); (M.A.S.); (E.R.W.)
| | - E. Robert Wassman
- Lineagen, Inc., Salt Lake City, UT 84109, USA; (A.P.); (L.S.); (C.H.H.); (M.A.S.); (E.R.W.)
| | | | - Temple Grandin
- Department of Animal Science, Colorado State University, Fort Collins, CO 80523, USA;
| | - Richard G. Boles
- The Center for Neurological and Neurodevelopmental Health, Voorhees, NJ 08043, USA;
| |
Collapse
|
23
|
Diener J, Sommer L. Reemergence of neural crest stem cell-like states in melanoma during disease progression and treatment. Stem Cells Transl Med 2020; 10:522-533. [PMID: 33258291 PMCID: PMC7980219 DOI: 10.1002/sctm.20-0351] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/28/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Melanoma is the deadliest of all skin cancers due to its high metastatic potential. In recent years, advances in targeted therapy and immunotherapy have contributed to a remarkable progress in the treatment of metastatic disease. However, intrinsic or acquired resistance to such therapies remains a major obstacle in melanoma treatment. Melanoma disease progression, beginning from tumor initiation and growth to acquisition of invasive phenotypes and metastatic spread and acquisition of treatment resistance, has been associated with cellular dedifferentiation and the hijacking of gene regulatory networks reminiscent of the neural crest (NC)—the developmental structure which gives rise to melanocytes and hence melanoma. This review summarizes the experimental evidence for the involvement of NC stem cell (NCSC)‐like cell states during melanoma progression and addresses novel approaches to combat the emergence of stemness characteristics that have shown to be linked with aggressive disease outcome and drug resistance.
Collapse
Affiliation(s)
- Johanna Diener
- University of Zurich, Institute of Anatomy, Zürich, Switzerland
| | - Lukas Sommer
- University of Zurich, Institute of Anatomy, Zürich, Switzerland
| |
Collapse
|
24
|
Perera SN, Kerosuo L. On the road again: Establishment and maintenance of stemness in the neural crest from embryo to adulthood. STEM CELLS (DAYTON, OHIO) 2020; 39:7-25. [PMID: 33017496 PMCID: PMC7821161 DOI: 10.1002/stem.3283] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 12/22/2022]
Abstract
Unique to vertebrates, the neural crest (NC) is an embryonic stem cell population that contributes to a greatly expanding list of derivatives ranging from neurons and glia of the peripheral nervous system, facial cartilage and bone, pigment cells of the skin to secretory cells of the endocrine system. Here, we focus on what is specifically known about establishment and maintenance of NC stemness and ultimate fate commitment mechanisms, which could help explain its exceptionally high stem cell potential that exceeds the "rules set during gastrulation." In fact, recent discoveries have shed light on the existence of NC cells that coexpress commonly accepted pluripotency factors like Nanog, Oct4/PouV, and Klf4. The coexpression of pluripotency factors together with the exceptional array of diverse NC derivatives encouraged us to propose a new term "pleistopotent" (Greek for abundant, a substantial amount) to be used to reflect the uniqueness of the NC as compared to other post-gastrulation stem cell populations in the vertebrate body, and to differentiate them from multipotent lineage restricted stem cells. We also discuss studies related to the maintenance of NC stemness within the challenging context of being a transient and thus a constantly changing population of stem cells without a permanent niche. The discovery of the stem cell potential of Schwann cell precursors as well as multiple adult NC-derived stem cell reservoirs during the past decade has greatly increased our understanding of how NC cells contribute to tissues formed after its initial migration stage in young embryos.
Collapse
Affiliation(s)
- Surangi N Perera
- Neural Crest Development and Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Laura Kerosuo
- Neural Crest Development and Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
25
|
Zhang X, Tang H, Mao S, Li B, Zhou Y, Yue H, Wang D, Wang Y, Fu J. Transplanted hair follicle stem cells migrate to the penumbra and express neural markers in a rat model of cerebral ischaemia/reperfusion. Stem Cell Res Ther 2020; 11:413. [PMID: 32967732 PMCID: PMC7510278 DOI: 10.1186/s13287-020-01927-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/16/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Ischaemic stroke has become the main cause of death and severe neurological disorders, for which effective restorative treatments are currently limited. While stem cell transplantation offers therapeutic potential through neural regeneration, this approach is associated with the challenges of limited applicable sources. Hair follicle stem cells (HFSCs) are multipotential cells that can differentiate into ectodermal and mesodermal lineages and proliferate for long periods. The therapeutic potentials of HFSCs have not been investigated in ischaemic stroke models, and therefore, in this study, we aimed to determine whether they could survive and migrate to ischaemic areas after a stroke attack. METHODS A rat model of middle cerebral artery ischaemia/reperfusion was established and intravenously administered HFSCs. The potential of HFSCs to migrate and differentiate into neuron-like cells as well as their ability to reduce the infarct size was evaluated. Rat brain tissue samples were collected 2 weeks after cell transplantation and analysed via TTC staining, immunofluorescence and immunohistochemistry methods. The data were statistically analysed and presented as the means ± standard deviations. RESULTS Intravenously administrated rat HFSCs were able to migrate to the penumbra where they expressed neuron-specific markers, reduced the infarct volume and promoted neurological recovery. CONCLUSION HFSC transplantation has therapeutic potential for ischaemic stroke and is, therefore, worthy of further investigation toward possible clinical development for treating stroke patients.
Collapse
Affiliation(s)
- Xuemei Zhang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
| | - Hao Tang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
| | - Senlin Mao
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
| | - Bing Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
| | - Yinglian Zhou
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
| | - Hui Yue
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
| | - Duo Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
| | - Yifei Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China
| | - Jin Fu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, No.246 Xuefu Road, Nangang District, Harbin, 150086, Heilongjiang Province, China.
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Zhang Y, Hu W, Ma K, Zhang C, Fu X. Reprogramming of Keratinocytes as Donor or Target Cells Holds Great Promise for Cell Therapy and Regenerative Medicine. Stem Cell Rev Rep 2020; 15:680-689. [PMID: 31197578 DOI: 10.1007/s12015-019-09900-8] [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: 12/30/2022]
Abstract
One of the most crucial branches of regenerative medicine is cell therapy, in which cellular material is injected into the patient to initiate the regenerative process. Cells obtained by reprogramming of the patient's own cells offer ethical and clinical advantages could provide a new source of material for therapeutic applications. Studies to date have shown that only a subset of differentiated cell types can be reprogrammed. Among these, keratinocytes, which are the most abundant proliferating cell type in the epidermis, have gained increasing attention as both donor and target cells for reprogramming and have become a new focus of regenerative medicine. As target cells for the treatment of skin defects, keratinocytes can be differentiated or reprogrammed from embryonic stem cells, induced pluripotent stem cells, fibroblasts, adipose tissue stem cells, and mesenchymal cells. As donor cells, keratinocytes can be reprogrammed or direct reprogrammed into a number of cell types, including induced pluripotent stem cells, neural cells, and Schwann cells. In this review, we discuss recent advances in keratinocyte reprogramming, focusing on the induction methods, potential molecular mechanisms, conversion efficiency, and safety for clinical applications. Graphical Abstract KCs as target cells can be reprogrammed or differentiated from fibroblasts, iPSCs, ATSCs, and mesenchymal cells. And as donor cells, KCs can be reprogrammed or directly reprogrammded into iPSCs, neural cells, Schwann cells, and epidermal stem cells.
Collapse
Affiliation(s)
- Yuehou Zhang
- School of Medicine, NanKai University, 94 Wei Jin Road, NanKai District, Tianjin, 300071, People's Republic of China.,Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center of General Hospital of PLA, 51 Fu Cheng Road, HaiDian District, Beijing, 100048, People's Republic of China
| | - Wenzhi Hu
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center of General Hospital of PLA, 51 Fu Cheng Road, HaiDian District, Beijing, 100048, People's Republic of China
| | - Kui Ma
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center of General Hospital of PLA, 51 Fu Cheng Road, HaiDian District, Beijing, 100048, People's Republic of China
| | - Cuiping Zhang
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center of General Hospital of PLA, 51 Fu Cheng Road, HaiDian District, Beijing, 100048, People's Republic of China.
| | - Xiaobing Fu
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center of General Hospital of PLA, 51 Fu Cheng Road, HaiDian District, Beijing, 100048, People's Republic of China.
| |
Collapse
|
29
|
Dash S, Trainor PA. The development, patterning and evolution of neural crest cell differentiation into cartilage and bone. Bone 2020; 137:115409. [PMID: 32417535 DOI: 10.1016/j.bone.2020.115409] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022]
Abstract
Neural crest cells are a vertebrate-specific migratory, multipotent cell population that give rise to a diverse array of cells and tissues during development. Cranial neural crest cells, in particular, generate cartilage, bone, tendons and connective tissue in the head and face as well as neurons, glia and melanocytes. In this review, we focus on the chondrogenic and osteogenic potential of cranial neural crest cells and discuss the roles of Sox9, Runx2 and Msx1/2 transcription factors and WNT, FGF and TGFβ signaling pathways in regulating neural crest cell differentiation into cartilage and bone. We also describe cranioskeletal defects and disorders arising from gain or loss-of-function of genes that are required for patterning and differentiation of cranial neural crest cells. Finally, we discuss the evolution of skeletogenic potential in neural crest cells and their function as a conduit for intraspecies and interspecies variation, and the evolution of craniofacial novelties.
Collapse
Affiliation(s)
- Soma Dash
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
30
|
Kempfle JS, Luu NNC, Petrillo M, Al-Asad R, Zhang A, Edge ASB. Lin28 reprograms inner ear glia to a neuronal fate. Stem Cells 2020; 38:890-903. [PMID: 32246510 PMCID: PMC10908373 DOI: 10.1002/stem.3181] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/05/2020] [Accepted: 02/08/2020] [Indexed: 12/16/2022]
Abstract
Sensorineural hearing loss is irreversible and can be caused by loss of auditory neurons. Regeneration of neural cells from endogenous cells may offer a future tool to restore the auditory circuit and to enhance the performance of implantable hearing devices. Neurons and glial cells in the peripheral nervous system are closely related and originate from a common progenitor. Prior work in our lab indicated that in the early postnatal mouse inner ear, proteolipid protein 1 (Plp1) expressing glial cells could act as progenitor cells for neurons in vitro. Here, we used a transgenic mouse model to transiently overexpress Lin28, a neural stem cell regulator, in Plp1-positive glial cells. Lin28 promoted proliferation and conversion of auditory glial cells into neurons in vitro. To study the effects of Lin28 on endogenous glial cells after loss of auditory neurons in vivo, we produced a model of auditory neuropathy by selectively damaging auditory neurons with ouabain. After neural damage was confirmed by the auditory brainstem response, we briefly upregulated the Lin28 in Plp1-expressing inner ear glial cells. One month later, we analyzed the cochlea for neural marker expression by quantitative RT-PCR and immunohistochemistry. We found that transient Lin28 overexpression in Plp1-expressing glial cells induced expression of neural stem cell markers and subsequent conversion into neurons. This suggests the potential for inner ear glia to be converted into neurons as a regeneration therapy for neural replacement in auditory neuropathy.
Collapse
Affiliation(s)
- Judith S. Kempfle
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts
- University Department of Otolaryngology, Head and Neck Surgery, Tübingen, Germany
| | - Ngoc-Nhi C. Luu
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts
- University Department of Otolaryngology, Head and Neck Surgery, Zürich, Switzerland
| | - Marco Petrillo
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Reef Al-Asad
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Andrea Zhang
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Albert S. B. Edge
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| |
Collapse
|
31
|
Schomann T, Iljas JD, Que I, Li Y, Suidgeest E, Cruz LJ, Frijns JHM, Chan A, Löwik CMWG, Huisman MA, Mezzanotte L. Multimodal imaging of hair follicle bulge-derived stem cells in a mouse model of traumatic brain injury. Cell Tissue Res 2020; 381:55-69. [PMID: 32036485 PMCID: PMC7306043 DOI: 10.1007/s00441-020-03173-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 01/20/2020] [Indexed: 01/01/2023]
Abstract
Traumatic brain injury (TBI) is a devastating event for which current therapies are limited. Stem cell transplantation may lead to recovery of function via different mechanisms, such as cell replacement through differentiation, stimulation of angiogenesis and support to the microenvironment. Adult hair follicle bulge-derived stem cells (HFBSCs) possess neuronal differentiation capacity, are easy to harvest and are relatively immune-privileged, which makes them potential candidates for autologous stem cell-based therapy. In this study, we apply in vivo multimodal, optical and magnetic resonance imaging techniques to investigate the behavior of mouse HFBSCs in a mouse model of TBI. HFBSCs expressed Luc2 and copGFP and were examined for their differentiation capacity in vitro. Subsequently, transduced HFBSCs, preloaded with ferumoxytol, were transplanted next to the TBI lesion (cortical region) in nude mice, 2 days after injury. Brains were fixed for immunohistochemistry 58 days after transplantation. Luc2- and copGFP-expressing, ferumoxytol-loaded HFBSCs showed adequate neuronal differentiation potential in vitro. Bioluminescence of the lesioned brain revealed survival of HFBSCs and magnetic resonance imaging identified their localization in the area of transplantation. Immunohistochemistry showed that transplanted cells stained for nestin and neurofilament protein (NF-Pan). Cells also expressed laminin and fibronectin but extracellular matrix masses were not detected. After 58 days, ferumoxytol could be detected in HFBSCs in brain tissue sections. These results show that HFBSCs are able to survive after brain transplantation and suggest that cells may undergo differentiation towards a neuronal cell lineage, which supports their potential use for cell-based therapy for TBI.
Collapse
Affiliation(s)
- Timo Schomann
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Percuros B.V, Leiden, the Netherlands
| | - Juvita D Iljas
- Percuros B.V, Leiden, the Netherlands
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Ivo Que
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Yuedan Li
- Percuros B.V, Leiden, the Netherlands
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ernst Suidgeest
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Johan H M Frijns
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Institute for Brain and Cognition, Leiden University, Leiden, the Netherlands
| | - Alan Chan
- Percuros B.V, Leiden, the Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Clemens M W G Löwik
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Margriet A Huisman
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Hair Science Institute, Maastricht, the Netherlands
| | - Laura Mezzanotte
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, the Netherlands.
- Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
32
|
Dong D, Chen S, Feng C, Xiong H, Xu X. NB-UVB Induces Melanocytic Differentiation of Human Hair Follicle Neural Crest Stem Cells. Ann Dermatol 2020; 32:289-297. [PMID: 33911756 PMCID: PMC7992648 DOI: 10.5021/ad.2020.32.4.289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/15/2020] [Accepted: 03/16/2020] [Indexed: 11/08/2022] Open
Abstract
Background Phototherapy is an important method to treat vitiligo. However, it is unclear how phototherapy affects melanocyte precursors and skin neural crest stem cells. Objective To investigate the underlying mechanisms of narrow-band ultraviolet B (NB-UVB) induced melanocyte lineage differentiated from human scalp-derived neural crest stem cells (HS-NCSCs). Methods HS-NCSCs were expanded from scalp hair follicles. The c-Kit-/CD57- HS-NCSCs were isolated by cell sorting. Different doses of NB-UVB were used to irradiate these HS-NCSCs. Cell ultrastructure was examined by transmission electron microscope. Melanocyte marker expression was analyzed by Quantitative RT-PCR and Western blot. Cell proliferation and migration were also evaluated. Results The c-Kit-/CD57- HS-NCSCs expressed embryonic NCSC biomarkers. NB-UVB at a dose of 100 mJ of NB-UVB had little effect on the cell proliferation of differentiated melanocytes from c-Kit-/CD57- HS-NCSCs, while 700 mJ inhibited cell proliferation significantly. The dendritic processes of differentiated melanocytes increased after radiation. The tyrosinase and Melanocortin 1 receptor (Mc1R) expression of differentiated melanocytes increased after NB-UVB exposure. The effect of NB-UVB on tyrosinase expression was modulated by signaling inhibitors H89 and PD98059 as well as Mc1R level in the cells. The migration ability of differentiated melanocytes was enhanced under 100 mJ exposure. Conclusion These data demonstrate that NB-UVB facilitates melanocytic differentiation of the HS-NCSCs and enhances migration of these cells. Mc1R and cAMP pathway play a critical role in NB-UVB induced melanocytic differentiation.
Collapse
Affiliation(s)
- Dake Dong
- Department of Dermatology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shujun Chen
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Cheng Feng
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Dermatology, the Second Affiliated Hospital of Xi'anJiaoTong University, Xi'an, Shanxi, China
| | - Huizi Xiong
- Department of Dermatology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
33
|
Abstract
Wild sheep and many primitive domesticated breeds have two coats: coarse hairs covering shorter, finer fibres. Both are shed annually. Exploitation of wool for apparel in the Bronze Age encouraged breeding for denser fleeces and continuously growing white fibres. The Merino is regarded as the culmination of this process. Archaeological discoveries, ancient images and parchment records portray this as an evolutionary progression, spanning millennia. However, examination of the fleeces from feral, two-coated and woolled sheep has revealed a ready facility of the follicle population to change from shedding to continuous growth and to revert from domesticated to primitive states. Modifications to coat structure, colour and composition have occurred in timeframes and to sheep population sizes that exclude the likelihood of variations arising from mutations and natural selection. The features are characteristic of the domestication phenotype: an assemblage of developmental, physiological, skeletal and hormonal modifications common to a wide variety of species under human control. The phenotypic similarities appeared to result from an accumulation of cryptic genetic changes early during vertebrate evolution. Because they did not affect fitness in the wild, the mutations were protected from adverse selection, becoming apparent only after exposure to a domestic environment. The neural crest, a transient embryonic cell population unique to vertebrates, has been implicated in the manifestations of the domesticated phenotype. This hypothesis is discussed with reference to the development of the wool follicle population and the particular roles of Notch pathway genes, culminating in the specific cell interactions that typify follicle initiation.
Collapse
|
34
|
Aran S, Zahri S, Asadi A, Khaksar F, Abdolmaleki A. Hair follicle stem cells differentiation into bone cells on collagen scaffold. Cell Tissue Bank 2020; 21:181-188. [PMID: 32016616 DOI: 10.1007/s10561-020-09812-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 01/24/2020] [Indexed: 12/17/2022]
Abstract
The hair follicle is a dynamic structure which contains different niches for stem cells, therefore; it has been considered as valuable and rich sources of stem cells, due to easy access, multipotency and non-oncogenic properties. In the present study, the differentiation capacities of hair follicle stem cells into bone cells on the natural collagen scaffolds were investigated. The stem cells were extracted from the hair follicle bulge area of male Wistar rats' whisker and cultured until 3rd passage, then osteogenic differentiations were induced by culturing the cells in the specific osteogenic medium. After 3 weeks, the differentiation parameters, including morphological changes, levels of calcification and expression of the bone specific genes were detected. The hydrogel preparation and scaffold fabrication was carried out using the extracted collagen and was studied by scanning electron microscope. Comparison of the stem cells' growth and changes on the scaffold and non-scaffold conditions showed that, in the both situation, the cells revealed differentiation signs of osteocytes, including large and cubic morphology with a star-shaped nucleus. Staining by Alizarin-red and Von-Kossa methods showed the presence of red and black calcium mass on the scaffold. Expression of the osteopontin and alkaline phosphatase genes confirmed the differentiation. Considerable porosity in the surface of the scaffold was recorded by scanning electron microscopy, which made it convenient for cells' attachment and growth. The data showed that the bulge stem cells possess significant capacity for osteoblastic differentiation and collagen scaffolds were found to be an appropriate matrix for growth and differentiation of the cell.
Collapse
Affiliation(s)
- Saeideh Aran
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Saber Zahri
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran.
| | - Asadollah Asadi
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Fatemeh Khaksar
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Arash Abdolmaleki
- Department of Engineering Sciences, Faculty of Advanced Technologies, University of Mohaghegh Ardabili, Namin, Iran
- Bio Science and Biotechnology Research Center (BBRC), Sabalan University of Advanced Technologies (SUAT), Namin, Iran
| |
Collapse
|
35
|
In Vitro Differentiation of Human Skin-Derived Cells into Functional Sensory Neurons-Like. Cells 2020; 9:cells9041000. [PMID: 32316463 PMCID: PMC7226083 DOI: 10.3390/cells9041000] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
Skin-derived precursor cells (SKPs) are neural crest stem cells that persist in certain adult tissues, particularly in the skin. They can generate a large type of cell in vitro, including neurons. SKPs were induced to differentiate into sensory neurons (SNs) by molecules that were previously shown to be important for the generation of SNs: purmorphamine, CHIR99021, BMP4, GDNF, BDNF, and NGF. We showed that the differentiation of SKPs induced the upregulation of neurogenins. At the end of the differentiation protocol, transcriptional analysis was performed on BRN3A and a marker of pain-sensing nerve cell PRDM12 genes: 1000 times higher for PRDM12 and 2500 times higher for BRN3A in differentiated cells than they were in undifferentiated SKPs. Using immunostaining, we showed that 65% and 80% of cells expressed peripheral neuron markers BRN3A and PERIPHERIN, respectively. Furthermore, differentiated cells expressed TRPV1, PAR2, TRPA1, substance P, CGRP, HR1. Using calcium imaging, we observed that a proportion of cells responded to histamine, SLIGKV (a specific agonist of PAR2), polygodial (a specific agonist of TRPA1), and capsaicin (a specific agonist of TRPV1). In conclusion, SKPs are able to differentiate directly into functional SNs. These differentiated cells will be very useful for further in vitro studies.
Collapse
|
36
|
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
|
37
|
Cerrizuela S, Vega-Lopez GA, Aybar MJ. The role of teratogens in neural crest development. Birth Defects Res 2020; 112:584-632. [PMID: 31926062 DOI: 10.1002/bdr2.1644] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/11/2019] [Accepted: 12/22/2019] [Indexed: 12/13/2022]
Abstract
The neural crest (NC), discovered by Wilhelm His 150 years ago, gives rise to a multipotent migratory embryonic cell population that generates a remarkably diverse and important array of cell types during the development of the vertebrate embryo. These cells originate in the neural plate border (NPB), which is the ectoderm between the neural plate and the epidermis. They give rise to the neurons and glia of the peripheral nervous system, melanocytes, chondrocytes, smooth muscle cells, odontoblasts and neuroendocrine cells, among others. Neurocristopathies are a class of congenital diseases resulting from the abnormal induction, specification, migration, differentiation or death of NC cells (NCCs) during embryonic development and have an important medical and societal impact. In general, congenital defects affect an appreciable percentage of newborns worldwide. Some of these defects are caused by teratogens, which are agents that negatively impact the formation of tissues and organs during development. In this review, we will discuss the teratogens linked to the development of many birth defects, with a strong focus on those that specifically affect the development of the NC, thereby producing neurocristopathies. Although increasing attention is being paid to the effect of teratogens on embryonic development in general, there is a strong need to critically evaluate the specific role of these agents in NC development. Therefore, increased understanding of the role of these factors in NC development will contribute to the planning of strategies aimed at the prevention and treatment of human neurocristopathies, whose etiology was previously not considered.
Collapse
Affiliation(s)
- Santiago Cerrizuela
- Área Biología Experimental, Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina.,Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Guillermo A Vega-Lopez
- Área Biología Experimental, Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina.,Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Manuel J Aybar
- Área Biología Experimental, Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina.,Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| |
Collapse
|
38
|
Baharvand Z, Nabiuni M, Tahmaseb M, Amini E, Pandamooz S. Investigating the synergic effects of valproic acid and crocin on BDNF and GDNF expression in epidermal neural crest stem cells. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2020-004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
39
|
Kubiak CA, Grochmal J, Kung TA, Cederna PS, Midha R, Kemp SWP. Stem-cell-based therapies to enhance peripheral nerve regeneration. Muscle Nerve 2019; 61:449-459. [PMID: 31725911 DOI: 10.1002/mus.26760] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 10/31/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022]
Abstract
Peripheral nerve injury remains a major cause of morbidity in trauma patients. Despite advances in microsurgical techniques and improved understanding of nerve regeneration, obtaining satisfactory outcomes after peripheral nerve injury remains a difficult clinical problem. There is a growing body of evidence in preclinical animal studies demonstrating the supportive role of stem cells in peripheral nerve regeneration after injury. The characteristics of both mesoderm-derived and ectoderm-derived stem cell types and their role in peripheral nerve regeneration are discussed, specifically focusing on the presentation of both foundational laboratory studies and translational applications. The current state of clinical translation is presented, with an emphasis on both ethical considerations of using stems cells in humans and current governmental regulatory policies. Current advancements in cell-based therapies represent a promising future with regard to supporting nerve regeneration and achieving significant functional recovery after debilitating nerve injuries.
Collapse
Affiliation(s)
- Carrie A Kubiak
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, Michigan
| | - Joey Grochmal
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Theodore A Kung
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, Michigan
| | - Paul S Cederna
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Rajiv Midha
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Stephen W P Kemp
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
40
|
Mehrotra P, Tseropoulos G, Bronner ME, Andreadis ST. Adult tissue-derived neural crest-like stem cells: Sources, regulatory networks, and translational potential. Stem Cells Transl Med 2019; 9:328-341. [PMID: 31738018 PMCID: PMC7031649 DOI: 10.1002/sctm.19-0173] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 12/15/2022] Open
Abstract
Neural crest (NC) cells are a multipotent stem cell population that give rise to a diverse array of cell types in the body, including peripheral neurons, Schwann cells (SC), craniofacial cartilage and bone, smooth muscle cells, and melanocytes. NC formation and differentiation into specific lineages takes place in response to a set of highly regulated signaling and transcriptional events within the neural plate border. Premigratory NC cells initially are contained within the dorsal neural tube from which they subsequently emigrate, migrating to often distant sites in the periphery. Following their migration and differentiation, some NC‐like cells persist in adult tissues in a nascent multipotent state, making them potential candidates for autologous cell therapy. This review discusses the gene regulatory network responsible for NC development and maintenance of multipotency. We summarize the genes and signaling pathways that have been implicated in the differentiation of a postmigratory NC into mature myelinating SC. We elaborate on the signals and transcription factors involved in the acquisition of immature SC fate, axonal sorting of unmyelinated neuronal axons, and finally the path toward mature myelinating SC, which envelope axons within myelin sheaths, facilitating electrical signal propagation. The gene regulatory events guiding development of SC in vivo provides insights into means for differentiating NC‐like cells from adult human tissues into functional SC, which have the potential to provide autologous cell sources for the treatment of demyelinating and neurodegenerative disorders.
Collapse
Affiliation(s)
- Pihu Mehrotra
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, New York
| | - Georgios Tseropoulos
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, New York
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, New York.,Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York.,Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| |
Collapse
|
41
|
Higa K, Higuchi J, Kimoto R, Satake Y, Yamaguchi T, Tomida D, Shimazaki J. Effects of Amniotic Membrane–Derived Fibroblast Supernatant on Corneal Epithelium. ACTA ACUST UNITED AC 2019; 60:3718-3726. [DOI: 10.1167/iovs.19-27041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Kazunari Higa
- Cornea Center Eye Bank, Tokyo Dental College Ichikawa General Hospital, Ichikawa, Chiba, Japan
| | - Junko Higuchi
- Cornea Center Eye Bank, Tokyo Dental College Ichikawa General Hospital, Ichikawa, Chiba, Japan
| | - Reona Kimoto
- Cornea Center Eye Bank, Tokyo Dental College Ichikawa General Hospital, Ichikawa, Chiba, Japan
| | - Yoshiyuki Satake
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Ichikawa, Chiba, Japan
| | - Takefumi Yamaguchi
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Ichikawa, Chiba, Japan
| | - Daisuke Tomida
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Ichikawa, Chiba, Japan
| | - Jun Shimazaki
- Cornea Center Eye Bank, Tokyo Dental College Ichikawa General Hospital, Ichikawa, Chiba, Japan
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Ichikawa, Chiba, Japan
| |
Collapse
|
42
|
Khalil S, Ariel Gru A, Saavedra AP. Cutaneous extramedullary haematopoiesis: Implications in human disease and treatment. Exp Dermatol 2019; 28:1201-1209. [DOI: 10.1111/exd.14013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 06/26/2019] [Accepted: 07/11/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Shadi Khalil
- Department of Dermatology University of Virginia School of Medicine Charlottesville Virginia
| | - Alejandro Ariel Gru
- Department of Pathology University of Virginia School of Medicine Charlottesville Virginia
| | - Arturo P. Saavedra
- Department of Dermatology University of Virginia School of Medicine Charlottesville Virginia
| |
Collapse
|
43
|
Lee YH, Lee HT, Chen CL, Chang CH, Hsu CY, Shyu WC. Role of FOXC1 in regulating APSCs self-renewal via STI-1/PrP C signaling. Am J Cancer Res 2019; 9:6443-6465. [PMID: 31588228 PMCID: PMC6771253 DOI: 10.7150/thno.35619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/20/2019] [Indexed: 01/03/2023] Open
Abstract
Forkhead box protein C1 (FOXC1) is known to regulate developmental processes in the skull and brain. Methods: The unique multipotent arachnoid-pia stem cells (APSCs) isolated from human and mouse arachnoid-pia membranes of meninges were grown as 3D spheres and displayed a capacity for self-renewal. Additionally, APSCs also expressed the surface antigens as mesenchymal stem cells. By applying the FOXC1 knockout mice and mouse brain explants, signaling cascade of FOXC1-STI-1-PrPC was investigated to demonstrate the molecular regulatory pathway for APSCs self-renewal. Moreover, APSCs implantation in stroke model was also verified whether neurogenic property of APSCs could repair the ischemic insult of the stroke brain. Results: Activated FOXC1 regulated the proliferation of APSCs in a cell cycle-dependent manner, whereas FOXC1-mediated APSCs self-renewal was abolished in FOXC1 knockout mice (FOXC1-/- mice). Moreover, upregulation of STI-1 regulated by FOXC1 enhanced cell survival and self-renewal of APSCs through autocrine signaling of cellular prion protein (PrPC). Mouse brain explants STI-1 rescues the cortical phenotype in vitro and induces neurogenesis in the FOXC1 -/- mouse brain. Furthermore, administration of APSCs in ischemic brain restored the neuroglial microenvironment and improved neurological dysfunction. Conclusion: We identified a novel role for FOXC1 in the direct regulation of the STI-1-PrPC signaling pathway to promote cell proliferation and self-renewal of APSCs.
Collapse
|
44
|
Kim DH, Kim BY, Kim DH, Hur J, Baek CH. Rabbit palatum-derived mesenchymal progenitor cells tri-lineage differentiation on 2D substrates and 3D printed constructs. J Appl Biomater Funct Mater 2019; 17:2280800019834520. [PMID: 31291802 DOI: 10.1177/2280800019834520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Hard palate, developed by embryo neural crest stem cells, is a tissue with strong regenerative abilities. It is considered an abundant source of progenitor cells, forming various mesenchymal tissues. Rabbits are more suitable models than murine animals for regenerative preclinical study of the head and neck, owing to their larger size. However, there are no reports of the existence or characteristics of neural crest stem cells in the hard palate of rabbits. In this study, we demonstrate for the first time the presence of nestin-, Sox2-, and p75-positive neural crest stem cells obtained from the hard palate of rabbits and the properties of these cells. Flow cytometry analysis revealed that CD29, CD44, and CD81 were positive; and CD11b, CD34, and CD90 were negative on the ex vivo expanded palatal progenitor cells. Finally, we differentiated them into cells of mesenchymal lineages (bone, cartilage, and fat) in vitro, and in three-dimensional fabricated polycaprolactone and polycaprolactone-tricalcium phosphate scaffolds. Taken together, our data showed the existence of rabbit palatum-derived mesenchymal progenitor cells, and successful fabrication of progenitor cell-loaded biodegradable scaffold using three-dimensional printing. This study will open avenues for new tissue engineering strategies for cell therapy using three-dimensional printing with scaffolds for reconstruction of head and neck defects.
Collapse
Affiliation(s)
- Dong Hwan Kim
- 1 Department of Orthopedic Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Bo Young Kim
- 2 Department of Otorhinolaryngology Head and Neck Surgery, Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Dong Hyun Kim
- 3 Department of Pediatrics, Hematology and Oncology, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Hur
- 4 Department of Convergence Medical Science, Pusan National University School of Medicine, Republic of Korea
| | - Chung-Hwan Baek
- 5 Department of Otorhinolaryngology-Head and Neck Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Republic of Korea
| |
Collapse
|
45
|
Neural crest stem cells from human epidermis of aged donors maintain their multipotency in vitro and in vivo. Sci Rep 2019; 9:9750. [PMID: 31278326 PMCID: PMC6611768 DOI: 10.1038/s41598-019-46140-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/24/2019] [Indexed: 11/08/2022] Open
Abstract
Neural crest (NC) cells are multipotent stem cells that arise from the embryonic ectoderm, delaminate from the neural tube in early vertebrate development and migrate throughout the developing embryo, where they differentiate into various cell lineages. Here we show that multipotent and functional NC cells can be derived by induction with a growth factor cocktail containing FGF2 and IGF1 from cultures of human inter-follicular keratinocytes (KC) isolated from elderly donors. Adult NC cells exhibited longer doubling times as compared to neonatal NC cells, but showed limited signs of cellular senescence despite the advanced age of the donors and exhibited significantly younger epigenetic age as compared to KC. They also maintained their multipotency, as evidenced by their ability to differentiate into all NC-specific lineages including neurons, Schwann cells, melanocytes, and smooth muscle cells (SMC). Notably, upon implantation into chick embryos, adult NC cells behaved similar to their embryonic counterparts, migrated along stereotypical pathways and contributed to multiple NC derivatives in ovo. These results suggest that KC-derived NC cells may provide an easily accessible, autologous source of stem cells that can be used for treatment of neurodegenerative diseases or as a model system for studying disease pathophysiology and drug development.
Collapse
|
46
|
The Use and Delivery of Stem Cells in Nerve Regeneration: Preclinical Evidence and Regulatory Considerations. Ann Plast Surg 2019; 80:448-456. [PMID: 29166311 DOI: 10.1097/sap.0000000000001259] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Outcomes following peripheral nerve injury remain poor despite the regenerative capacity displayed by the peripheral nervous system. Current therapies are limited and do not provide satisfactory functional recovery in a multitude of cases. Biomaterials have decreased the need for nerve autograft across small nerve gaps in small-caliber nerves, but the lack of a cellular substrate presents a limiting factor to the effectiveness of this therapy. Schwann cells are the supportive cells in the peripheral nervous system and play an integral role in the physiological response and regeneration following nerve injury. Limitations to autologous Schwann cells include donor site morbidity during harvesting, limited expansion capability, and finite source. Stem cells are multipotent or pluripotent cells with self-renewing capabilities that show promise to improve functional recovery following nerve injury. Differentiation of stem cells into supportive Schwann cells could provide additional trophic support without the disadvantages of autologous Schwann cells, providing an avenue to improve existing therapies. A variety of stem cells have been evaluated in animal models for this clinical application; the current options, along with their clinical feasibility, are summarized in this article.
Collapse
|
47
|
Kim YH, Kim BJ, Kim SM, Kim SU, Ryu BY. Induction of cardiomyocyte‑like cells from hair follicle cells in mice. Int J Mol Med 2019; 43:2230-2240. [PMID: 30864673 DOI: 10.3892/ijmm.2019.4133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 03/08/2019] [Indexed: 11/05/2022] Open
Abstract
Hair follicles (HFs) are a well‑characterized niche for adult stem cells (SCs), and include epithelial and melanocytic SCs. HF cells are an accessible source of multipotent adult SCs for the generation of the interfollicular epidermis, HF structures and sebaceous glands in addition to the reconstitution of novel HFs in vivo. In the present study, it was demonstrated that HF cells are able to be induced to differentiate into cardiomyocyte‑like cells in vitro under specific conditions. It was determined that HF cells cultured on OP9 feeder cells in KnockOut‑Dulbecco's modified Eagle's medium/B27 in the presence of vascular endothelial growth factors differentiated into cardiomyocyte‑like cells that express markers specific to cardiac lineage, but do not express non‑cardiac lineage markers including neural stem/progenitor cell, HF bulge cells or undifferentiated spermatogonia markers. These cardiomyocyte‑like cells exhibited a spindle‑ and filament‑shaped morphology similar to that presented by cardiac muscles and exhibited spontaneous beating that persisted for over 3 months. These results demonstrate that SC reprogramming and differentiation may be induced without resulting in any genetic modification, which is important for the clinical applications of SCs including tissue and organ regeneration.
Collapse
Affiliation(s)
- Yong-Hee Kim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung‑Ang University, Anseong, Gyeonggi‑do 17546, Republic of Korea
| | - Bang-Jin Kim
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seok-Man Kim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung‑Ang University, Anseong, Gyeonggi‑do 17546, Republic of Korea
| | - Sun-Uk Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| | - Buom-Yong Ryu
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung‑Ang University, Anseong, Gyeonggi‑do 17546, Republic of Korea
| |
Collapse
|
48
|
Comparative Analysis of Biological Properties of Large-Scale Expanded Adult Neural Crest-Derived Stem Cells Isolated from Human Hair Follicle and Skin Dermis. Stem Cells Int 2019; 2019:9640790. [PMID: 30915126 PMCID: PMC6399535 DOI: 10.1155/2019/9640790] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/14/2018] [Accepted: 11/22/2018] [Indexed: 12/14/2022] Open
Abstract
Introduction The adult neural crest-derived stem cells (NCSCs) have significant perspectives for use in regenerative medicine. The most attractive sources for adult NCSC isolation are the hair follicles (HF) and skin dermis (SD) because of easy access and minimally invasive biopsy. The aim of this study was to compare the biological properties of HF- and SD-derived NCSCs after their large-scale expansion. Methods The conventional explant method was used to obtain HF NCSCs. For the isolation of SD NCSCs, a new combined technique consisting of preplating and subsequent culturing in 3D blood plasma-derived fibrin hydrogel was applied. The studied cells were characterized by flow cytometry, ICC, qPCR, Bio-Plex multiplex assay, and directed multilineage differentiation assays. Results We have obtained both adult SD and HF NCSCs from each skin sample (n = 5). Adult SD and HF NCSCs were positive for key neural crest markers: SOX10, P75 (CD271), NESTIN, SOX2, and CD349. SD NCSCs showed a higher growth rate during the large-scale expansion compared to HF NCSCs (p < 0.01). Final population of SD NCSCs also contained more clonogenic cells (p < 0.01) and SOX10+, CD271+, CD105+, CD140a+, CD146+, CD349+ cells (p < 0.01). Both HF and SD NCSCs had similar gene expression profiling and produced growth factors, but some quantitative differences were detected. Adult HF and SD NCSCs were able to undergo directed differentiation into neurons, Schwann cells, adipocytes, and osteoblasts. Conclusion The HF and SD are suitable sources for large-scale manufacturing of adult NCSCs with similar biological properties. We demonstrated that the NCSC population from SD was homogenous and displayed significantly higher growth rate than HF NCSCs. Moreover, SD NCSC isolation is cheaper, easier, and minimally time-consuming method.
Collapse
|
49
|
Salehi MS, Borhani-Haghighi A, Pandamooz S, Safari A, Dargahi L, Dianatpour M, Tanideh N. Dimethyl fumarate up-regulates expression of major neurotrophic factors in the epidermal neural crest stem cells. Tissue Cell 2019; 56:114-120. [PMID: 30736899 DOI: 10.1016/j.tice.2019.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/20/2019] [Accepted: 01/21/2019] [Indexed: 02/07/2023]
Abstract
There is an agreement that combining treatments can lead to substantial improvement, therefore the present study assessed the effects of different concentrations of dimethyl fumarate (DMF) on viability of epidermal neural crest stem cells (EPI-NCSCs). In addition, this investigation was designed to evaluate the effects of DMF on relative expression of major trophic factors mainly the ones with neurotrophic effects, expressed in EPI-NCSCs in order to enhance their therapeutic potential. To determine the appropriate concentration of DMF for EPI-NCSCs treatment, the MTT assay was employed and based on the obtained data, EPI-NCSCs treated with 10μM DMF for 6, 24, 72 or 168 h. In each time point, quantitative RT-PCR technique was used to evaluate NGF, NT-3, BDNF, GDNF and VEGF transcripts. The acquired data showed that 10μM DMF significantly increased the mRNA expression of NGF, NT-3 and BDNF, 72 h following treatment; however, DMF inhibitory effect on GDNF mRNA expression was observed in various time points. No significant changes were detected for VEGF transcript. Our findings reveled that expression of major neurotrophic factors were up-regulated by dimethyl fumarate treatment. Therefore, combining EPI-NCSCs with DMF treatment might be a valuable strategy to improve their therapeutic functions in vivo.
Collapse
Affiliation(s)
- Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Sareh Pandamooz
- 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.
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Dianatpour
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nader Tanideh
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
50
|
Head to Knee: Cranial Neural Crest-Derived Cells as Promising Candidates for Human Cartilage Repair. Stem Cells Int 2019; 2019:9310318. [PMID: 30766608 PMCID: PMC6350557 DOI: 10.1155/2019/9310318] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 11/04/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022] Open
Abstract
A large array of therapeutic procedures is available to treat cartilage disorders caused by trauma or inflammatory disease. Most are invasive and may result in treatment failure or development of osteoarthritis due to extensive cartilage damage from repeated surgery. Despite encouraging results of early cell therapy trials that used chondrocytes collected during arthroscopic surgery, these approaches have serious disadvantages, including morbidity associated with cell harvesting and low predictive clinical outcomes. To overcome these limitations, adult stem cells derived from bone marrow and subsequently from other tissues are now considered as preferred sources of cells for cartilage regeneration. Moreover, with new evidence showing that the choice of cell source is one of the most important factors for successful cell therapy, there is growing interest in neural crest-derived cells in both the research and clinical communities. Neural crest-derived cells such as nasal chondrocytes and oral stem cells that exhibit chondrocyte-like properties seem particularly promising in cartilage repair. Here, we review the types of cells currently available for cartilage cell therapy, including articular chondrocytes and various mesenchymal stem cells, and then highlight recent developments in the use of neural crest-derived chondrocytes and oral stem cells for repair of cartilage lesions.
Collapse
|