1
|
Song H, Zhao XB, Chu QS, Zhang J, Gao L, Liao XH. Expression dynamics of lymphoid enhancer-binding factor 1 in terminal Schwann cells, dermal papilla, and interfollicular epidermis. Dev Dyn 2022; 252:527-535. [PMID: 36576725 DOI: 10.1002/dvdy.562] [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: 09/19/2022] [Revised: 12/24/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Transcription factor lymphoid enhancer-binding factor 1 (LEF1) is a downstream mediator of the Wnt/β-catenin signaling pathway. It is expressed in dermal papilla and surrounding cells in the hair follicle, promoting cell proliferation, and differentiation. RESULTS Here, we report that LEF1 is also expressed all through the hair cycle in the terminal Schwann cells (TSCs), a component of the lanceolate complex located at the isthmus. The timing of LEF1 appearance at the isthmus coincides with that of hair follicle innervation. LEF1 is not found at the isthmus in the aberrant hair follicles in nude mice. Instead, LEF1 in TSCs is found in the de novo hair follicles reconstituted on nude mice by stem cells chamber graft assay. Cutaneous denervation experiment demonstrates that the LEF1 expression in TSCs is independent of nerve endings. At last, LEF1 expression in the interfollicular epidermis during the early stage of skin development is significantly suppressed in transgenic mice with T-cell factor 3 (TCF3) overexpression. CONCLUSION We reveal the expression dynamics of LEF1 in skin during development and hair cycle. LEF1 expression in TSCs indicates that the LEF1/Wnt signal might help to establish a niche at the isthmus region for the lanceolate complex, the bulge stem cells and other neighboring cells.
Collapse
Affiliation(s)
- Hongzhi Song
- School of Medicine, Shanghai University, Shanghai, China.,School of Life Sciences, Shanghai University, Shanghai, China.,School of Environmental and Chemical Engineering, Shanghai University, Shanghai, China
| | - Xu-Bo Zhao
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Qing-Song Chu
- School of Life Sciences, Shanghai University, Shanghai, China.,Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Jianyu Zhang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Lipeng Gao
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xin-Hua Liao
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
2
|
Lu Q, Gao Y, Fan Z, Xiao X, Chen Y, Si Y, Kong D, Wang S, Liao M, Chen X, Wang X, Chu W. Amphiregulin promotes hair regeneration of skin-derived precursors via the PI3K and MAPK pathways. Cell Prolif 2021; 54:e13106. [PMID: 34382262 PMCID: PMC8450126 DOI: 10.1111/cpr.13106] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 12/13/2022] Open
Abstract
Objectives There are significant clinical challenges associated with alopecia treatment, including poor efficiency of related drugs and insufficient hair follicles (HFs) for transplantation. Skin‐derived precursors (SKPs) exhibit great potential as stem cell‐based therapies for hair regeneration; however, the proliferation and hair‐inducing capacity of SKPs gradually decrease during culturing. Materials and Methods We describe a 3D co‐culture system accompanied by kyoto encyclopaedia of genes and genomes and gene ontology enrichment analyses to determine the key factors and pathways that enhance SKP stemness and verified using alkaline phosphatase assays, Ki‐67 staining, HF reconstitution, Western blot and immunofluorescence staining. The upregulated genes were confirmed utilizing corresponding recombinant protein or small‐interfering RNA silencing in vitro, as well as the evaluation of telogen‐to‐anagen transition and HF reconstitution in vivo. Results The 3D co‐culture system revealed that epidermal stem cells and adipose‐derived stem cells enhanced SKP proliferation and HF regeneration capacity by amphiregulin (AREG), with the promoted stemness allowing SKPs to gain an earlier telogen‐to‐anagen transition and high‐efficiency HF reconstitution. By contrast, inhibitors of the phosphoinositide 3‐kinase (PI3K) and mitogen‐activated protein kinase (MAPK) pathways downstream of AREG signalling resulted in diametrically opposite activities. Conclusions By exploiting a 3D co‐culture model, we determined that AREG promoted SKP stemness by enhancing both proliferation and hair‐inducing capacity through the PI3K and MAPK pathways. These findings suggest AREG therapy as a potentially promising approach for treating alopecia.
Collapse
Affiliation(s)
- Qiumei Lu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Ying Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China.,Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Zhimeng Fan
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Xing Xiao
- Center of Scientific Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yu Chen
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuan Si
- Department of Dermatology, Guangzhou First People's Hospital, Guangzhou, China
| | - Deqiang Kong
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Shuai Wang
- The Yonghe Medical Beauty Clinic Limited Company, Guangzhou, China
| | - Meijian Liao
- School of basic medicine, Xuzhou Medical University, Xuzhou, China
| | - Xiaodong Chen
- Department of Burn Surgery, The First People's Hospital of Foshan, Foshan, China
| | - Xusheng Wang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Weiwei Chu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
3
|
Kobayashi M, Zochodne DW. Diabetic polyneuropathy: Bridging the translational gap. J Peripher Nerv Syst 2021; 25:66-75. [PMID: 32573914 DOI: 10.1111/jns.12392] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/22/2022]
Abstract
Clinical trials for diabetic polyneuropathy (DPN) have failed to identify therapeutic impacts that have arrested or reversed the disorder, despite a long history. This review considers DPN in the context of a unique neurodegenerative disorder that targets peripheral neurons and their companion glial cells. The approach is to examine what cells, cell substructures, and pathways are implicated in causing DPN and how they might be addressed therapeutically. These include axonopathy, neuronopathy, hyperglycemia, polyol flux, advanced glycation endproduct (AGE)-receptor AGE signaling, growth factor disruption, abnormal insulin signaling, and abnormalities of other intrinsic neuron pathways. Mitochondrial dysfunction and lipid toxicity are largely delegated to the companion review in this issue by Stino and Feldman. Finally, the linkage between axon plasticity of cutaneous nerves, peripheral neuroregenerative pathways, and diabetes are discussed.
Collapse
Affiliation(s)
- Masaki Kobayashi
- Department of Neurology, Nissan Tamagawa Hospital, Tokyo, Japan.,Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Douglas W Zochodne
- Division of Neurology and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
4
|
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: 4.0] [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
|
5
|
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: 1.4] [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
|
6
|
Yoshizumi T, Kubo A, Murata H, Shinonaga M, Kanno H. BC-Box Motif in SOCS6 Induces Differentiation of Epidermal Stem Cells into GABAnergic Neurons. Int J Mol Sci 2020; 21:ijms21144947. [PMID: 32668737 PMCID: PMC7403999 DOI: 10.3390/ijms21144947] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 12/11/2022] Open
Abstract
The BC-box motif in suppressor of cytokine signaling 6 (SOCS6) promotes the neuronal differentiation of somatic stem cells, including epidermal stem cells. SOCS6 protein belongs to the group of SOCS proteins and inhibits cytokine signaling. Here we showed that epidermal stem cells were induced to differentiate into GABAnergic neurons by the intracellular delivery of a peptide composed of the amino-acid sequences encoded by the BC-box motif in SOCS6 protein. The BC-box motif (SLQYLCRFVI) in SOCS6 corresponded to the binding site of elongin BC. GABAnergic differentiation mediated by the BC-box motif in SOCS6 protein was caused by ubiquitination of JAK2 and inhibition of the JAK2-STAT3 pathway. Furthermore, GABAnergic neuron-like cells generated from epidermal stem cells were transplanted into the brain of a rodent ischemic model. Then, we demonstrated that these transplanted cells were GAD positive and that the cognitive function of the ischemic model rodents with the transplanted cells was improved. This study could contribute to not only elucidating the mechanism of GABAnergic neuronal differentiation but also to neuronal regenerative medicine utilizing GABAnergic neurons.
Collapse
Affiliation(s)
- Tetsuya Yoshizumi
- Department of Neurosurgery, International University of Health and Welfare Atami Hospital, Atami 413-0012, Japan; (T.Y.); (M.S.)
| | - Atsuhiko Kubo
- Nerve Care Clinic, Yokosuka 238-0012, Japan;
- Department of Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan;
| | - Hidetoshi Murata
- Department of Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan;
| | - Masamichi Shinonaga
- Department of Neurosurgery, International University of Health and Welfare Atami Hospital, Atami 413-0012, Japan; (T.Y.); (M.S.)
| | - Hiroshi Kanno
- Department of Neurosurgery, International University of Health and Welfare Atami Hospital, Atami 413-0012, Japan; (T.Y.); (M.S.)
- Department of Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan;
- Correspondence: ; Tel.: +81-557-81-9171; Fax: +81-557-83-6632
| |
Collapse
|
7
|
Chen Y, Shen J, Ma C, Cao M, Yan J, Liang J, Ke K, Cao M, Xiaosu G. Skin-derived precursor Schwann cells protect SH-SY5Y cells against 6-OHDA-induced neurotoxicity by PI3K/AKT/Bcl-2 pathway. Brain Res Bull 2020; 161:84-93. [PMID: 32360763 DOI: 10.1016/j.brainresbull.2020.03.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 03/22/2020] [Accepted: 03/31/2020] [Indexed: 02/08/2023]
Abstract
Skin-derived precursors (SKPs) are self-renewing and pluripotent adult stem cell sources that have been successfully obtained and cultured from adult tissues of rodents and humans. Skin-derived precursor Schwann cells (SKP-SCs), derived from SKPs when cultured in a neuro stromal medium supplemented with some appropriate neurotrophic factors, have been reported to play a neuroprotective effect in the peripheral nervous system. This proves our previous studies that SKP-SCs' function to bridge sciatic nerve gap in rats. However, the function of SKP-SCs in Parkinson disease (PD) remains unknown. This study was aimed to investigate the possible neuroprotective effects of SKP-SCs in 6-OHDA-induced Parkinson's disease (PD) model. Our results showed that the treatment with SKP-SCs prevented SH-SY5Y cells from 6-OHDA-induced apoptosis, accompanied by modulation of apoptosis-related proteins (Bcl-2 and Bax) and the decreased expression of active caspase-3. Furthermore, we confirmed that SKP-SCs might exert protective effects and increase the mitochondrial membrane potential (MMP) through PI3K/AKT/Bcl-2 pathway. Taken together, our results demonstrated that SKP-SCs protect against 6-OHDA-induced cytotoxicity through PI3K/AKT/Bcl-2 pathway in PD model in vitro, which provides a new theoretical basis for the treatment of PD.
Collapse
Affiliation(s)
- Ying Chen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jiabing Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Chengxiao Ma
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Maosheng Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jianan Yan
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jingjing Liang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Kaifu Ke
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Maohong Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China.
| | - Gu Xiaosu
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, China.
| |
Collapse
|
8
|
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: 2.4] [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
|
9
|
Bergeron L, Busuttil V, Botto JM. Multipotentiality of skin-derived precursors: application to the regeneration of skin and other tissues. Int J Cosmet Sci 2020; 42:5-15. [PMID: 31612512 DOI: 10.1111/ics.12587] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/12/2019] [Indexed: 12/13/2022]
Abstract
Skin-derived precursors (SKPs) have been described as multipotent dermal precursors. Here, we provide a review of the breadth and depth of scientific literature and studies regarding SKPs, accounting for a large number of scientific publications. Interestingly, these progenitors can be isolated from embryonic and adult skin, as well as from a population of dermal cells cultured in vitro in monolayer. Gathering information from different authors, this review explores different aspects of the SKP theme, such as the potential distinct origins of SKPs in rodents and in humans, and also their ability to differentiate in vitro and in vivo into multiple lineages of different progeny. This remarkable capacity makes SKPs an interesting endogenous source of precursors to explore in the framework of experimental and therapeutic applications in different domains. SKPs are not only involved in the skin's dermal maintenance and support as well as wound healing, but also in hair follicle morphogenesis. This review points out the interests of future researches on SKPs for innovative perspectives that may be helpful in many different types of scientific and medical domains.
Collapse
Affiliation(s)
- L Bergeron
- Ashland Specialties France, Global Skin Research Center, 655, route du Pin Montard, 06904, Sophia Antipolis, France
| | - V Busuttil
- Ashland Specialties France, Global Skin Research Center, 655, route du Pin Montard, 06904, Sophia Antipolis, France
| | - J-M Botto
- Ashland Specialties France, Global Skin Research Center, 655, route du Pin Montard, 06904, Sophia Antipolis, France
| |
Collapse
|
10
|
Castro AR, Logarinho E. Tissue engineering strategies for human hair follicle regeneration: How far from a hairy goal? Stem Cells Transl Med 2019; 9:342-350. [PMID: 31876379 PMCID: PMC7031632 DOI: 10.1002/sctm.19-0301] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022] Open
Abstract
The demand for an efficient therapy for alopecia disease has fueled the hair research field in recent decades. However, despite significant improvements in the knowledge of key processes of hair follicle biology such as genesis and cycling, translation into hair follicle replacement therapies has not occurred. Great expectation has been recently put on hair follicle bioengineering, which is based on the development of fully functional hair follicles with cycling activity from an expanded population of hair‐inductive (trichogenic) cells. Most bioengineering approaches focus on in vitro reconstruction of folliculogenesis by manipulating key regulatory molecular/physical features of hair follicle growth/cycling in vivo. Despite their great potential, no cell‐based product is clinically available for hair regeneration therapy to date. This is mainly due to demanding issues that still hinder the functionality of cultured human hair cells. The present review comprehensively compares emergent strategies using different cell sources and tissue engineering approaches, aiming to successfully achieve a clinical cure for hair loss. The hurdles of these strategies are discussed, as well as the future directions to overcome the obstacles and fulfill the promise of a “hairy” feat.
Collapse
Affiliation(s)
- Ana Rita Castro
- Aging and Aneuploidy Group, IBMC, Instituto de Biologia Molecular e Celular, Porto, Portugal.,i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Programa Doutoral em Engenharia Biomédica, Faculdade de Engenharia, Universidade do Porto, Porto, Portugal.,Saúde Viável - Clínica de Microtransplante Capilar, Porto, Portugal
| | - Elsa Logarinho
- Aging and Aneuploidy Group, IBMC, Instituto de Biologia Molecular e Celular, Porto, Portugal.,i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Saúde Viável - Clínica de Microtransplante Capilar, Porto, Portugal
| |
Collapse
|
11
|
Dai R, Hua W, Chen W, Xiong L, Li L, Li Y. Isolation, Characterization, and Safety Evaluation of Human Skin-Derived Precursors from an Adherent Monolayer Culture System. Stem Cells Int 2019; 2019:9194560. [PMID: 31531027 PMCID: PMC6721512 DOI: 10.1155/2019/9194560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/18/2019] [Accepted: 07/16/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Skin-derived precursors (SKPs) are promising candidates for regenerative medicine. Several studies have transcultured human SKPs (termed tSKPs) from fibroblasts (FBs) expanded in monolayer culture. Herein, we optimized the procedure by treating flasks with poly-2-hydroxyethyl methacrylate (poly-HEMA). METHODS tSKPs generated from our adherent monolayer culture system were investigated for protein expression and differentiation capacity. The aggregated cells and the proliferative cells within tSKP spheres were detected by mix-culturing FBs expressing two different fluorescent proteins and BrdU- or EdU-positive cells, respectively. To distinguish tSKPs from FBs, we compared their phenotypes and transcriptomes. The tumorigenicity of tSKPs and FBs was also assessed in our study. RESULTS tSKPs expressed Versican, Fibronectin, Vimentin, Sox2, and Nestin. Under appropriate stimuli, tSKPs could differentiate to mesenchymal or neural lineages. While these spheres were heterogeneous populations consisting of both proliferative and aggregated cells, the rate of proliferative cells correlated with a seeding density. tSKPs, isolated from FBs, were distinctive from FBs in cell cycle, marker expression, neural differentiation potential, and transcript profiles despite the two sharing partial similarity in certain properties. As for tumorigenesis, both tSKPs and FBs could be considered as nontumorigenic ex vivo and in vivo. CONCLUSION tSKPs were heterogeneous populations presenting similar characteristics as traditional SKPs, while being different from FBs. The potential mixture of FBs in spheres did not affect the biosafety of tSKPs, as both of which had normal karyotype and nontumorigenicity. Taken together, we suggested tSKPs had potential applications in regenerative medicine.
Collapse
Affiliation(s)
- Ru Dai
- Department of Dermatology, Ningbo First Hospital, Zhejiang University, No. 59, Liuting Street, Ningbo, Zhejiang 315010, China
- Department of Dermatology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Wei Hua
- Department of Dermatology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Wei Chen
- Department of Dermatology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Lidan Xiong
- Department of Dermatology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Li Li
- Department of Dermatology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Yiming Li
- Department of Dermatology, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan 610041, China
| |
Collapse
|
12
|
Joshi SS, Tandukar B, Pan L, Huang JM, Livak F, Smith BJ, Hodges T, Mahurkar AA, Hornyak TJ. CD34 defines melanocyte stem cell subpopulations with distinct regenerative properties. PLoS Genet 2019; 15:e1008034. [PMID: 31017901 PMCID: PMC6481766 DOI: 10.1371/journal.pgen.1008034] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/18/2019] [Indexed: 12/16/2022] Open
Abstract
Melanocyte stem cells (McSCs) are the undifferentiated melanocytic cells of the mammalian hair follicle (HF) responsible for recurrent generation of a large number of differentiated melanocytes during each HF cycle. HF McSCs reside in both the CD34+ bulge/lower permanent portion (LPP) and the CD34- secondary hair germ (SHG) regions of the HF during telogen. Using Dct-H2BGFP mice, we separate bulge/LPP and SHG McSCs using FACS with GFP and anti-CD34 to show that these two subsets of McSCs are functionally distinct. Genome-wide expression profiling results support the distinct nature of these populations, with CD34- McSCs exhibiting higher expression of melanocyte differentiation genes and with CD34+ McSCs demonstrating a profile more consistent with a neural crest stem cell. In culture and in vivo, CD34- McSCs regenerate pigmentation more efficiently whereas CD34+ McSCs selectively exhibit the ability to myelinate neurons. CD34+ McSCs, and their counterparts in human skin, may be useful for myelinating neurons in vivo, leading to new therapeutic opportunities for demyelinating diseases and traumatic nerve injury.
Collapse
Affiliation(s)
- Sandeep S. Joshi
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Bishal Tandukar
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Li Pan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jennifer M. Huang
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Ferenc Livak
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Marlene and Stuart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Barbara J. Smith
- Institute for Basic Biomedical Sciences, John Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Theresa Hodges
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Anup A. Mahurkar
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Thomas J. Hornyak
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Marlene and Stuart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Research & Development Service, VA Maryland Health Care System, United States Department of Veterans Affairs, Baltimore, Maryland, United States of America
- Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
13
|
Park S, Kim H, Kim K, Roh S. Sonic hedgehog signalling regulates the self-renewal and proliferation of skin-derived precursor cells in mice. Cell Prolif 2018; 51:e12500. [PMID: 30151845 PMCID: PMC6528853 DOI: 10.1111/cpr.12500] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES The sonic hedgehog (Shh) signalling pathway has an important role in the maintenance of various stem cells and organogenesis during development. However, the effect of Shh in skin-derived precursors (SKPs), which have the capacity for multipotency and self-renewal, is not yet clear. The present study investigated the effects of the Shh signalling pathway on the proliferation and self-renewal of murine SKPs (mSKPs). METHODS The Shh signalling pathway was activated by treatment with purmorphamine (Shh agonist) or recombinant Shh in mSKPs. Cyclopamine (Shh antagonist) or GANT-61 (Gli inhibitor) was used to inhibit the pathway. Western blot, qPCR, and immunofluorescence were used to analyse the expression of genes related to self-renewal, stemness, epithelial-mesenchymal transition (EMT) and the Shh signalling pathway. In addition, cell proliferation and apoptosis were examined. RESULTS Inhibiting the Shh signalling pathway reduced mSKP proliferation and sphere formation, but increased apoptosis. Activating this signalling pathway produced opposite results. The Shh signalling pathway also controlled the EMT phenotype in mSKPs. Moreover, purmorphamine recovered the self-renewal and proliferation of aged mSKPs. CONCLUSION Our results suggest that the Shh signalling pathway has an important role in the proliferation, self-renewal and apoptosis of mSKPs. These findings also provide a better understanding of the cellular mechanisms underlying SKP self-renewal and apoptosis that allow more efficient expansion of SKPs.
Collapse
Affiliation(s)
- Sangkyu Park
- Cellular Reprogramming and Embryo Biotechnology LaboratoryDental Research Institute, BK21, Seoul National University School of DentistrySeoulKorea
| | - Hyewon Kim
- Cellular Reprogramming and Embryo Biotechnology LaboratoryDental Research Institute, BK21, Seoul National University School of DentistrySeoulKorea
| | - Kichul Kim
- Cellular Reprogramming and Embryo Biotechnology LaboratoryDental Research Institute, BK21, Seoul National University School of DentistrySeoulKorea
| | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology LaboratoryDental Research Institute, BK21, Seoul National University School of DentistrySeoulKorea
| |
Collapse
|
14
|
Yousefi F, Lavi Arab F, Saeidi K, Amiri H, Mahmoudi M. Various strategies to improve efficacy of stem cell transplantation in multiple sclerosis: Focus on mesenchymal stem cells and neuroprotection. J Neuroimmunol 2018; 328:20-34. [PMID: 30557687 DOI: 10.1016/j.jneuroim.2018.11.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 11/30/2018] [Indexed: 02/09/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) which predominantly affect young adults and undergo heavy socioeconomic burdens. Conventional therapeutic modalities for MS mostly downregulate aggressive immune responses and are almost insufficient for management of progressive course of the disease. Mesenchymal stem cells (MSCs), due to both immunomodulatory and neuroprotective properties have been known as practical cells for treatment of neurodegenerative diseases like MS. However, clinical translation of MSCs is associated with some limitations such as short-life engraftment duration, little in vivo trans-differentiation and restricted accessibility into damaged sites. Therefore, laboratory manipulation of MSCs can improve efficacy of MSCs transplantation in MS patients. In this review, we discuss several novel approaches, which can potentially enhance MSCs capabilities for treating MS.
Collapse
Affiliation(s)
- Forouzan Yousefi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Lavi Arab
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kolsoum Saeidi
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Houshang Amiri
- Neurology Research Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
15
|
Matsushita T, Steinfeld J, Fujihara A, Urayama S, Taketani S, Araki M. Regulation of neuronal and photoreceptor cell differentiation by Wnt signaling from iris-derived stem/progenitor cells of the chick in flat vs. Matrigel-embedding cultures. Brain Res 2018; 1704:207-218. [PMID: 30347217 DOI: 10.1016/j.brainres.2018.10.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/17/2018] [Accepted: 10/17/2018] [Indexed: 01/03/2023]
Abstract
Previously we developed a simple culture method of the iris tissues and reported novel properties of neural stem/progenitor-like cells in the iris tissues of the chick and pig. When the iris epithelium or connective tissue (stroma) was treated with dispase, embedded in Matrigel, and cultured, neuronal cells extended from the explants within 24 h of culture, and cells positively stained for photoreceptor cell markers were also observed within a few days of culturing. In ordinary flat tissue culture conditions, explants had the same differentiation properties to those in tissue environments. Previously, we suggested that iris neural stem/progenitor cells are simply suppressed from neuronal differentiation within tissue, and that separation from the tissue releases the cells from this suppression mechanism. Here, we examined whether Wnt signaling suppressed neuronal differentiation of iris tissue cells in tissue environments because the lens, which has direct contact with the iris, is a rich source of Wnt proteins. When the Wnt signaling activator 6-bromoindirubin-3'-oxime (BIO) was administered to Matrigel culture, neuronal differentiation was markedly suppressed, but cell proliferation was not affected. When Wnt signaling inhibitors, such as DKK-1 and IWR-1, were applied to the same culture, they did not have any effect on cell differentiation and proliferation. However, when the inhibitors were applied to flat tissue culture, cells with neural properties emerged. These results indicate that the interaction of iris tissue with neighboring tissues and the environment regulates the stemness nature of iris tissue cells, and that Wnt signaling is a major factor.
Collapse
Affiliation(s)
- Tamami Matsushita
- Developmental Neurobiology Laboratory, Nara Women's University, Nara 630-8506, Japan
| | | | - Ai Fujihara
- Developmental Neurobiology Laboratory, Nara Women's University, Nara 630-8506, Japan
| | - Satoshi Urayama
- Unit of Neural Development and Regeneration, Nara Medical University, Kashihara 634-8521, Japan
| | - Shigeru Taketani
- Department of Biotechnology, Kyoto Institute of Technology, Kyoto 606-8585, Japan
| | - Masasuke Araki
- Developmental Neurobiology Laboratory, Nara Women's University, Nara 630-8506, Japan; Unit of Neural Development and Regeneration, Nara Medical University, Kashihara 634-8521, Japan.
| |
Collapse
|
16
|
Biochemical re-programming of human dermal stem cells to neurons by increasing mitochondrial membrane potential. Cell Death Differ 2018; 26:1048-1061. [PMID: 30154448 DOI: 10.1038/s41418-018-0182-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/15/2018] [Accepted: 07/22/2018] [Indexed: 01/07/2023] Open
Abstract
Stem cells are generally believed to contain a small number of mitochondria, thus accounting for their glycolytic phenotype. We demonstrate here, however, that despite an indispensable glucose dependency, human dermal stem cells (hDSCs) contain very numerous mitochondria. Interestingly, these stem cells segregate into two distinct subpopulations. One exhibits high, the other low-mitochondrial membrane potentials (Δψm). We have made the same observations with mouse neural stem cells (mNSCs) which serve here as a complementary model to hDSCs. Strikingly, pharmacologic inhibition of phosphoinositide 3-kinase (PI3K) increased the overall Δψm, decreased the dependency on glycolysis and led to formation of TUJ1 positive, electrophysiologically functional neuron-like cells in both mNSCs and hDSCs, even in the absence of any neuronal growth factors. Furthermore, of the two, it was the Δψm-high subpopulation which produced more mitochondrial reactive oxygen species (ROS) and showed an enhanced neuronal differentiation capacity as compared to the Δψm-low subpopulation. These data suggest that the Δψm-low stem cells may function as the dormant stem cell population to sustain future neuronal differentiation by avoiding excessive ROS production. Thus, chemical modulation of PI3K activity, switching the metabotype of hDSCs to neurons, may have potential as an autologous transplantation strategy for neurodegenerative diseases.
Collapse
|
17
|
Iwasa SN, Popovic MR, Morshead CM. Skin-derived precursor cells undergo substrate-dependent galvanotaxis that can be modified by neighbouring cells. Stem Cell Res 2018; 31:95-101. [PMID: 30059907 DOI: 10.1016/j.scr.2018.07.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/13/2018] [Accepted: 07/18/2018] [Indexed: 12/28/2022] Open
Abstract
Many cell types respond to electric fields (EFs) through cell migration, a process termed galvanotaxis. The galvanotactic response is critical for development and wound healing. Here we investigate whether skin-derived precursor cells (SKPs), which have the potential to differentiate into mesodermal and peripheral neural cell types, undergo directed migration in the presence of an EF. We found that EF application promotes SKP migration towards the anode. The migratory response is substrate-dependent as SKPs undergo directed migration on laminin and Matrigel, but not collagen. The majority of SKPs express the undifferentiated cell markers nestin, fibronectin and Sox2, after both EF application and in sister cultures with no EF application, suggesting that EFs do not promote cell differentiation. Co-cultures of SKPs and brain-derived neural precursor cells (NPCs), a population of cells that undergo rapid, cathode-directed migration, reveal that in the presence of NPCs an increased percentage of SKPs undergo galvanotaxis, providing evidence that cells can provide cues to modify the galvanotactic response. We propose that a better understanding of SKP migration in the presence of EFs may provide insight into improved strategies for wound repair.
Collapse
Affiliation(s)
- Stephanie N Iwasa
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3E1, Canada; Toronto Rehabilitation Institute - University Health Network, Toronto, Ontario M4G 3V9, Canada.
| | - Milos R Popovic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3E1, Canada; Toronto Rehabilitation Institute - University Health Network, Toronto, Ontario M4G 3V9, Canada.
| | - Cindi M Morshead
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3E1, Canada; Toronto Rehabilitation Institute - University Health Network, Toronto, Ontario M4G 3V9, Canada; Department of Surgery, University of Toronto, Toronto, Ontario M5S 3E1, Canada.
| |
Collapse
|
18
|
The Human Skin-Derived Precursors for Regenerative Medicine: Current State, Challenges, and Perspectives. Stem Cells Int 2018; 2018:8637812. [PMID: 30123295 PMCID: PMC6079335 DOI: 10.1155/2018/8637812] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/29/2018] [Accepted: 06/13/2018] [Indexed: 02/05/2023] Open
Abstract
Skin-derived precursors (SKPs) are an adult stem cell source with self-renewal and multipotent differentiation. Although rodent SKPs have been discussed in detail in substantial studies, human SKPs (hSKPs) are rarely reported. Understanding the biological properties and possible mechanisms underlying hSKPs has important implications for regenerative medicine particularly clinical applications, as human-derived sources are more suitable for clinical transplantation. The finding that hSKPs derivatives, such as neural and mesodermal progeny, have both in vitro and in vivo function without any genetical modification makes hSKPs a trustable, secure, and accessible resource for cell-based therapy. Here, we provide an overview of hSKPs, describing their characteristics, originations and niches, and potential applications. A comparison between traditional and innovative culture methods used for hSKPs is also introduced. Furthermore, we discuss the challenges and the future perspectives towards the field of hSKPs. With this review, we hope to point out the current stage of hSKPs and highlight the problems that remain in this field.
Collapse
|
19
|
Dental Pulp Stem Cells - Exploration in a Novel Animal Model: the Tasmanian Devil (Sarcophilus harrisii). Stem Cell Rev Rep 2018; 14:500-509. [PMID: 29737458 DOI: 10.1007/s12015-018-9814-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Dental pulp stem cells (DPSC) are a heterogeneous population of highly proliferative stem cells located in the soft inner pulp tissue of the tooth. Demonstrated to have an affinity for neural differentiation, DPSC have been reported to generate functional Schwann cells (SC) through in vitro differentiation. Both DPSC and SC have neural crest origins, recently a significant population of DPSC have been reported to derive from peripheral nerve-associated glia. The predisposition DPSC have towards the SC lineage is not only a very useful tool for neural regenerative therapies in the medical field, it also holds great promise in the veterinary field. Devil Facial Tumour (DFT) is a clonally transmissible cancer of SC origin responsible for devastating wild populations of the Tasmanian devil. Very few studies have investigated the healthy Tasmanian devil SC (tdSC) for comparative studies between tdSC and DFT cells, and the development and isolation of a tdSC population is yet to be undertaken. A Tasmanian devil DPSC model offers a promising new outlook for DFT research, and the link between SC and DPSC may provide a potential explanation as to how a cancerous SC initially arose in a single Tasmanian devil to then go on to infect others as a parasitic clonal cell line. In this review we explore the current role of DPSC in human regenerative medicine, provide an overview of the Tasmanian devil and the devastating effect of DFT, and highlight the promising potential DPSC techniques pose for DFT research and our current understanding of DFT.
Collapse
|
20
|
Calderone A. The Biological Role of Nestin (+)-Cells in Physiological and Pathological Cardiovascular Remodeling. Front Cell Dev Biol 2018; 6:15. [PMID: 29492403 PMCID: PMC5817075 DOI: 10.3389/fcell.2018.00015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 01/31/2018] [Indexed: 01/02/2023] Open
Abstract
The intermediate filament protein nestin was identified in diverse populations of cells implicated in cardiovascular remodeling. Cardiac resident neural progenitor/stem cells constitutively express nestin and following an ischemic insult migrate to the infarct region and participate in angiogenesis and neurogenesis. A modest number of normal adult ventricular fibroblasts express nestin and the intermediate filament protein is upregulated during the progression of reparative and reactive fibrosis. Nestin depletion attenuates cell cycle re-entry suggesting that increased expression of the intermediate filament protein in ventricular fibroblasts may represent an activated phenotype accelerating the biological impact during fibrosis. Nestin immunoreactivity is absent in normal adult rodent ventricular cardiomyocytes. Following ischemic damage, the intermediate filament protein is induced in a modest population of pre-existing adult ventricular cardiomyocytes bordering the peri-infarct/infarct region and nestin(+)-ventricular cardiomyocytes were identified in the infarcted human heart. The appearance of nestin(+)-ventricular cardiomyocytes post-myocardial infarction (MI) recapitulates an embryonic phenotype and depletion of the intermediate filament protein inhibits cell cycle re-entry. Recruitment of the serine/threonine kinase p38 MAPK secondary to an overt inflammatory response after an ischemic insult may represent a seminal event limiting the appearance of nestin(+)-ventricular cardiomyocytes and concomitantly suppressing cell cycle re-entry. Endothelial and vascular smooth muscle cells (VSMCs) express nestin and upregulation of the intermediate filament protein may directly contribute to vascular remodeling. This review will highlight the biological role of nestin(+)-cells during physiological and pathological remodeling of the heart and vasculature and discuss the phenotypic advantage attributed to the intermediate filament protein.
Collapse
Affiliation(s)
- Angelino Calderone
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada.,Montreal Heart Institute, Montréal, QC, Canada
| |
Collapse
|
21
|
Therapy of corneal endothelial dysfunction with corneal endothelial cell-like cells derived from skin-derived precursors. Sci Rep 2017; 7:13400. [PMID: 29042661 PMCID: PMC5645363 DOI: 10.1038/s41598-017-13787-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 10/03/2017] [Indexed: 12/13/2022] Open
Abstract
Corneal endothelial dysfunction occurs when corneal endothelial cells (CECs) are dramatically lost and eventually results in vision loss. Corneal transplantation is the only solution at present. However, corneal transplantation requires a fresh human cornea and there is a worldwide shortage of donors. Therefore, finding new functional CECs to replace human CECs is urgent. Skin-derived precursors (SKPs) can be easily acquired and have multiple differential potential. We co-cultured human SKPs with B4G12 cells in serum-free medium and obtained abundant CEC-like cells which had similar morphology and characteristic to human CECs. CEC-like cells exerted excellent therapeutic effect when they were transplanted into rabbit and monkey corneal endothelial dysfunction models by injection method. This protocol enables efficient production of CEC-like cells from SKPs. The renewable cell source, novel derivation method and simple treatment strategy may lead to potential applications in cell replacement therapy for corneal endothelial dysfunction.
Collapse
|
22
|
Xian D, Gao X, Xiong X, Xu J, Yang L, Pan L, Zhong J. Photoprotection against UV-induced damage by skin-derived precursors in hairless mice. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2017; 175:73-82. [PMID: 28865317 DOI: 10.1016/j.jphotobiol.2017.08.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/20/2017] [Accepted: 08/21/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Skin photodamage is associated with UV-induced overproduction of reactive oxygen species (ROS) and the inactivation of NF-E2-related factor 2 (Nrf2). Skin-derived precursor cells (SKPs), a population of dermal stem cells, are considered to be involved in wound repair and skin regeneration through the activation of Nrf2. However, no reports concentrate on the treatment of skin photodamage with SKPs. OBJECTIVE To investigate the photoprotective role of SKPs against UV-induced damage in mice. METHODS Fifty Balb/c hairless mice were divided into five groups (n=10), namely, normal (no intervention), model, prevention, treatment, and control groups. The latter four groups were dorsally exposed to UVA+UVB irradiation over a 2-week period. Mice in the prevention group received weekly SKP injections for 2weeks the day before irradiation. Mice in the treatment and Hanks groups received a two-time injection of SKPs and Hanks, respectively, after irradiation. One week after final intervention, skin appearance, pathological alterations, and oxidative indicators were evaluated by enzyme-linked immunosorbent assay, immunohistochemical analysis, and western blotting. RESULTS After irradiation, lesions were observed on the dorsal skin of mice, including erythema, edema, scales, and wrinkles; however, these were significantly ameliorated by subcutaneous SKP injection. Hyperkeratosis, acanthosis, and spongiosis in the epidermis, as well as dermal papillae edema and inflammatory cell infiltration, were observed in both model and control groups; however, these conditions resolved with either pretreatment or posttreatment with SKPs. In addition, SKPs increased Nrf2, heme oxygenase-1, glutathione peroxidase, superoxide dismutase, catalase, and gluthathione expression, while decreasing levels of ROS, MDA, and H2O2. CONCLUSIONS These findings suggest that SKPs have a photoprotective role against UV-induced damage in mice, which may be associated with their ability to scavenge photo-oxidative insults and activate Nrf2.
Collapse
Affiliation(s)
- Dehai Xian
- Department of Neurobiology, Southwest Medical University, Luzhou 646000, China
| | - Xiaoqing Gao
- Department of Neurobiology, Southwest Medical University, Luzhou 646000, China
| | - Xia Xiong
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jixiang Xu
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Lingyu Yang
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Lun Pan
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jianqiao Zhong
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
23
|
Human bone marrow harbors cells with neural crest-associated characteristics like human adipose and dermis tissues. PLoS One 2017; 12:e0177962. [PMID: 28683107 PMCID: PMC5500284 DOI: 10.1371/journal.pone.0177962] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 05/05/2017] [Indexed: 12/13/2022] Open
Abstract
Adult neural crest stem-derived cells (NCSC) are of extraordinary high plasticity and promising candidates for use in regenerative medicine. Several locations such as skin, adipose tissue, dental pulp or bone marrow have been described in rodent, as sources of NCSC. However, very little information is available concerning their correspondence in human tissues, and more precisely for human bone marrow. The main objective of this study was therefore to characterize NCSC from adult human bone marrow. In this purpose, we compared human bone marrow stromal cells to human adipose tissue and dermis, already described for containing NCSC. We performed comparative analyses in terms of gene and protein expression as well as functional characterizations. It appeared that human bone marrow, similarly to adipose tissue and dermis, contains NESTIN+ / SOX9+ / TWIST+ / SLUG+ / P75NTR+/ BRN3A+/ MSI1+/ SNAIL1+ cells and were able to differentiate into melanocytes, Schwann cells and neurons. Moreover, when injected into chicken embryos, all those cells were able to migrate and follow endogenous neural crest migration pathways. Altogether, the phenotypic characterization and migration abilities strongly suggest the presence of neural crest-derived cells in human adult bone marrow.
Collapse
|
24
|
Inagaki E, Hatou S, Higa K, Yoshida S, Shibata S, Okano H, Tsubota K, Shimmura S. Skin-Derived Precursors as a Source of Progenitors for Corneal Endothelial Regeneration. Stem Cells Transl Med 2017; 6:788-798. [PMID: 28186681 PMCID: PMC5442762 DOI: 10.1002/sctm.16-0162] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 09/16/2016] [Accepted: 09/30/2016] [Indexed: 12/13/2022] Open
Abstract
Corneal blindness is the fourth leading cause of blindness in the world. Current treatment is allogenic corneal transplantation, which is limited by shortage of donors and immunological rejection. Skin-derived precursors (SKPs) are postnatal stem cells, which are self-renewing, multipotent precursors that can be isolated and expanded from the dermis. Facial skin may therefore be an accessible autologous source of neural crest derived cells. SKPs were isolated from facial skin of Wnt1-Cre/Floxed EGFP mouse. After inducing differentiation with medium containing retinoic acid and GSK 3-β inhibitor, SKPs formed polygonal corneal endothelial-like cells (sTECE). Expression of major corneal endothelial markers were confirmed by Reverse transcription polymerase chain reaction (RT-PCR) and quantitative Real time polymerase chain reaction (qRT-PCR). Western blots confirmed the expression of Na, K-ATPase protein, the major functional marker of corneal endothelial cells. Immunohistochemistry revealed the expression of zonular occludens-1 and Na, K-ATPase in cell-cell junctions. In vitro functional analysis of Na, K-ATPase pump activity revealed that sTECE had significantly high pump function compared to SKPs or control 3T3 cells. Moreover, sTECE transplanted into a rabbit model of bullous keratopathy successfully maintained corneal thickness and transparency. Furthermore, we successfully induced corneal endothelial-like cells from human SKPs, and showed that transplanted corneas also maintained corneal transparency and thickness. Our findings suggest that SKPs may be used as a source of autologous cells for the treatment of corneal endothelial disease. Stem Cells Translational Medicine 2017;6:788-798.
Collapse
Affiliation(s)
- Emi Inagaki
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Shin Hatou
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Kazunari Higa
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Ichikawa, Japan
| | - Satoru Yoshida
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Shinsuke Shibata
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Shigeto Shimmura
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
25
|
Bayati V, Abbaspour MR, Neisi N, Hashemitabar M. Skin-derived precursors possess the ability of differentiation into the epidermal progeny and accelerate burn wound healing. Cell Biol Int 2016; 41:187-196. [PMID: 27981666 DOI: 10.1002/cbin.10717] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 12/10/2016] [Indexed: 12/31/2022]
Abstract
Skin-derived precursors (SKPs) are remnants of the embryonic neural crest stem cells that reside in the dermis until adulthood. Although they possess a wide range of differentiation potentials, their differentiation into keratinocyte-like cells and their roles in skin wound healing are obscure. The present study aimed to investigate the differentiation of SKPs into keratinocyte-like cells and evaluate their role in healing of third degree burn wounds. To this aim, SKPs were differentiated into keratinocyte-like cells on tissue culture plate and collagen-chitosan scaffold prepared by freeze-drying. Their differentiation capability was detected by real-time RT-PCR and immunofluorescence. Thereafter, they were cultured on scaffold and implanted in a rat model of burn wound. Histopathological and immunohistochemical analyses were employed to examine the reconstituted skin. The research findings revealed that SKPs were able to differentiate along the epidermal lineage and this ability can be enhanced on a suitable scaffold. Additionally, the results indicated that SKPs apparently promoted wound healing process and accelerate its transition from proliferating stage to maturational phase, especially if they were differentiated into keratinocyte-like cells. Regarding the results, it is concluded that SKPs are able to differentiate into keratinocyte-like cells, particularly when they are cultured on collagen-chitosan scaffold. Moreover, they can regenerate epidermal and dermal layers including thick collagen bundles, possibly through differentiation into keratinocyte-like cells.
Collapse
Affiliation(s)
- Vahid Bayati
- Cellular and Molecular Research Centre, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, 61357-15794, Iran.,Department of Anatomical Sciences, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, 61357-15794, Iran
| | - Mohammad Reza Abbaspour
- Targeted Drug Delivery Research Centre, Mashhad University of Medical Sciences, Mashhad, 91775-1365, Iran
| | - Niloofar Neisi
- Department of Medical Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, 61357-15794, Iran
| | - Mahmoud Hashemitabar
- Cellular and Molecular Research Centre, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, 61357-15794, Iran.,Department of Anatomical Sciences, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, 61357-15794, Iran
| |
Collapse
|
26
|
Xiao J, Li Q, Qu P, Zhang Z, Pan S, Wang Y, Zhang Y. Isolation of Bovine Skin-Derived Precursor Cells and Their Developmental Potential After Nuclear Transfer. Cell Reprogram 2016; 18:411-418. [PMID: 27906583 DOI: 10.1089/cell.2016.0021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Nuclei from less differentiated stem cells yield high cloning efficiency. However, pluripotent stem cells are rather difficult to obtain from bovines. Skin-derived precursor (SKPs) cells exhibit a certain degree of pluripotency, which has been shown to enhance the efficiency of nuclear transfer (NT) in pigs. In this study, bovine SKPs were isolated and characterized. Results showed that bovine SKPs expressed nestin, fibronectin, vimentin, pluripotency-related genes, and characteristic neural crest markers, such as NGFR, PAX3, SOX9, SNAI2, and OCT4. Bovine SKPs and fibroblasts were used as NT donor cells to examine and compare the preimplantation developmental potential of reconstructed embryos after somatic cell nuclear transfer (SCNT). Bovine SKP-cloned embryos displayed higher developmental competence in terms of blastocyst formation rate and total cell number in blastocysts compared with the bovine embryonic fibroblast-cloned embryos. This study revealed that bovine SKPs may be considered excellent candidate nuclear donors for SCNT and may provide a promising platform for transgenic cattle generation.
Collapse
Affiliation(s)
- Jiajia Xiao
- 1 College of Veterinary Medicine, Northwest A&F University , Yangling, Shaanxi, 712100 China .,2 Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University , Yangling, Shaanxi, 712100 China
| | - Qiaoqiao Li
- 1 College of Veterinary Medicine, Northwest A&F University , Yangling, Shaanxi, 712100 China .,2 Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University , Yangling, Shaanxi, 712100 China
| | - Pengxiang Qu
- 1 College of Veterinary Medicine, Northwest A&F University , Yangling, Shaanxi, 712100 China .,2 Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University , Yangling, Shaanxi, 712100 China
| | - Zihan Zhang
- 1 College of Veterinary Medicine, Northwest A&F University , Yangling, Shaanxi, 712100 China .,2 Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University , Yangling, Shaanxi, 712100 China
| | - Shaohui Pan
- 1 College of Veterinary Medicine, Northwest A&F University , Yangling, Shaanxi, 712100 China .,2 Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University , Yangling, Shaanxi, 712100 China
| | - Yongsheng Wang
- 1 College of Veterinary Medicine, Northwest A&F University , Yangling, Shaanxi, 712100 China .,2 Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University , Yangling, Shaanxi, 712100 China
| | - Yong Zhang
- 1 College of Veterinary Medicine, Northwest A&F University , Yangling, Shaanxi, 712100 China .,2 Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University , Yangling, Shaanxi, 712100 China
| |
Collapse
|
27
|
Bayati V, Gazor R, Nejatbakhsh R, Negad Dehbashi F. Enrichment of skin-derived neural precursor cells from dermal cell populations by altering culture conditions. Stem Cell Investig 2016; 3:83. [PMID: 28066785 DOI: 10.21037/sci.2016.10.10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 10/25/2016] [Indexed: 01/26/2023]
Abstract
BACKGROUND As stem cells play a critical role in tissue repair, their manipulation for being applied in regenerative medicine is of great importance. Skin-derived precursors (SKPs) may be good candidates for use in cell-based therapy as the only neural stem cells which can be isolated from an accessible tissue, skin. Herein, we presented a simple protocol to enrich neural SKPs by monolayer adherent cultivation to prove the efficacy of this method. METHODS To enrich neural SKPs from dermal cell populations, we have found that a monolayer adherent cultivation helps to increase the numbers of neural precursor cells. Indeed, we have cultured dermal cells as monolayer under serum-supplemented (control) and serum-supplemented culture, followed by serum free cultivation (test) and compared. Finally, protein markers of SKPs were assessed and compared in both experimental groups and differentiation potential was evaluated in enriched culture. RESULTS The cells of enriched culture concurrently expressed fibronectin, vimentin and nestin, an intermediate filament protein expressed in neural and skeletal muscle precursors as compared to control culture. In addition, they possessed a multipotential capacity to differentiate into neurogenic, glial, adipogenic, osteogenic and skeletal myogenic cell lineages. CONCLUSIONS It was concluded that serum-free adherent culture reinforced by growth factors have been shown to be effective on proliferation of skin-derived neural precursor cells (skin-NPCs) and drive their selective and rapid expansion.
Collapse
Affiliation(s)
- Vahid Bayati
- Cellular and Molecular Research Center, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran;; Department of Anatomical Sciences, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Rohoullah Gazor
- Department of Anatomy and Cell Biology, Gilan University of Medical Sciences, Rasht, Iran
| | - Reza Nejatbakhsh
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Fereshteh Negad Dehbashi
- Cellular and Molecular Research Center, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
28
|
Skin-Derived Precursors against UVB-Induced Apoptosis via Bcl-2 and Nrf2 Upregulation. BIOMED RESEARCH INTERNATIONAL 2016; 2016:6894743. [PMID: 27635399 PMCID: PMC5011201 DOI: 10.1155/2016/6894743] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/10/2016] [Accepted: 07/31/2016] [Indexed: 02/05/2023]
Abstract
Bcl-2 and Nrf2 are critical factors in protecting cells against UVB-induced apoptosis. Hair-follicle-bulge stem cells could resist ionization through Bcl-2 upregulation. Skin-derived precursors (SKPs) dwelling on the bulge may be against UVB irradiation. Initially, SKPs were isolated and identified. Then, SKPs were exposed to UVB and grew in medium for 24 hours. CCK-8 assay, TUNEL, and Ki67 staining evaluated cells apoptosis/proliferation, while SA-βgal staining evaluated cells senescence and pH2AX immunostaining evaluated DNA damage. Meanwhile, Bcl-2, Nrf2, HO-1, Bax, and Bak expressions were assessed by qRT-PCR and western blot. Two weeks later, floating spheres appeared and were identified as SKPs. After UVB radiation, SKPs maintained spherical colonies and outnumbered unirradiated ones, showing high Ki67 expression and low TUNEL, SA-βgal, and pH2AX expression. Fibroblasts (FBs), however, displayed deformation, senescence, and reduction, with increased TUNEL, SA-βgal, and pH2AX expression. Moreover, Bcl-2 and Nrf2 mRNA expression were significantly higher than Bak and Bax in irradiated SKPs. Conversely, Bcl-2 and Nrf2 mRNA levels greatly decreased compared with Bax and Bak in irradiated FBs. Interestingly, SKPs showed higher protein levels of Bcl-2, Nrf2, and HO-1 than FBs. SKPs exert a beneficial effect on resisting UVB-induced apoptosis, which may be associated with Bcl-2 and Nrf2 upregulation.
Collapse
|
29
|
Adult skin-derived precursor Schwann cells exhibit superior myelination and regeneration supportive properties compared to chronically denervated nerve-derived Schwann cells. Exp Neurol 2016; 278:127-42. [DOI: 10.1016/j.expneurol.2016.02.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 02/01/2016] [Accepted: 02/04/2016] [Indexed: 01/09/2023]
|
30
|
Steinbach SK, Husain M. Vascular smooth muscle cell differentiation from human stem/progenitor cells. Methods 2015; 101:85-92. [PMID: 26678794 DOI: 10.1016/j.ymeth.2015.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 12/03/2015] [Accepted: 12/07/2015] [Indexed: 01/16/2023] Open
Abstract
Transplantation of vascular smooth muscle cells (VSMCs) is a promising cellular therapy to promote angiogenesis and wound healing. However, VSMCs are derived from diverse embryonic sources which may influence their role in the development of vascular disease and in its therapeutic modulation. Despite progress in understanding the mechanisms of VSMC differentiation, there remains a shortage of robust methods for generating lineage-specific VSMCs from pluripotent and adult stem/progenitor cells in serum-free conditions. Here we describe a method for differentiating pluripotent stem cells, such as embryonic and induced pluripotent stem cells, as well as skin-derived precursors, into lateral plate-derived VSMCs including 'coronary-like' VSMCs and neural crest-derived VSMC, respectively. We believe this approach will have broad applications in modeling origin-specific disease vulnerability and in developing personalized cell-based vascular grafts for regenerative medicine.
Collapse
Affiliation(s)
- Sarah K Steinbach
- McEwen Centre for Regenerative Medicine, Toronto General Research Institute, 101 College St., Toronto, Ontario M5G-1L7, Canada; Division of Experimental Therapeutics, Toronto General Research Institute, 101 College St., Toronto, Ontario M5G-1L7, Canada
| | - Mansoor Husain
- McEwen Centre for Regenerative Medicine, Toronto General Research Institute, 101 College St., Toronto, Ontario M5G-1L7, Canada; Division of Experimental Therapeutics, Toronto General Research Institute, 101 College St., Toronto, Ontario M5G-1L7, Canada; Departments of Medicine, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Departments of Physiology, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Departments of Laboratory Medicine & Pathobiology, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Ted Rogers Centre for Heart Research, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Peter Munk Cardiac Centre, University Health Network, 200 Elizabeth St., Toronto, Ontario M5G-2C4, Canada.
| |
Collapse
|
31
|
Omidi A, Ragerdi Kashani I, Akbari M, Mortezaee K, Ghasemi S, Beyer C, Zendedel A. Homing of allogeneic nestin-positive hair follicle-associated pluripotent stem cells after maternal transplantation in experimental model of cortical dysplasia. Biochem Cell Biol 2015; 93:619-25. [DOI: 10.1139/bcb-2015-0098] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
An embryo has the capability to accept allo- or xeno-geneic cells, which probably makes it an ideal candidate for stem cell transplantation of various cerebral cortex abnormalities, such as cortical dysplasia. The aim of this study was to determine hair follicle-associated pluripotent (HAP) stem cells homing into various organs of mother and fetus. Cells were obtained, analyzed for immunophenotypic features, and then labelled with CM-Dil; nestin+HAP stem cells or media phosphate-buffered saline (PBS) were intravenously delivered on day 16 of gestation in BALB/c mice, which intraperitoneally received methylazoxymethanol (MAM) one day in advance, and homing was assessed at 24 h after cell injection. Flow cytometry and immunocytochemistry manifested positive expression of nestin in HAP stem cells. For both mother and fetus, brain, lungs, liver, and spleen were the host organs for cell implants. For the brain, the figure was considerably higher in fetus, 4.05 ± 0.5% (p ≤ 0.05 vs. mother). MAM-injected mice had a downward trend for SDF-1α and CXCR4 (p ≤ 0.05 vs. control), but HAP stem cells group showed an upward trend for CXCR4 (p ≤ 0.05 vs. MAM). We conclude the HAP stem cells show homing potential in experimental cortical dysplasia, which may permit these cells to be a target in future work on prenatal therapy of neural disorders.
Collapse
Affiliation(s)
- Ameneh Omidi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, 16 Azar Street, Pour Sina Street, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, 16 Azar Street, Pour Sina Street, Tehran, Iran
| | - Mohammad Akbari
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, 16 Azar Street, Pour Sina Street, Tehran, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, 16 Azar Street, Pour Sina Street, Tehran, Iran
| | - Soudabeh Ghasemi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, 16 Azar Street, Pour Sina Street, Tehran, Iran
| | - Cordian Beyer
- Institute of Neuroanatomy, School of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Adib Zendedel
- Institute of Neuroanatomy, School of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
32
|
Oriented cell division: new roles in guiding skin wound repair and regeneration. Biosci Rep 2015; 35:BSR20150225. [PMID: 26582817 PMCID: PMC4708010 DOI: 10.1042/bsr20150225] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/12/2015] [Indexed: 01/14/2023] Open
Abstract
Tissue morphogenesis depends on precise regulation and timely co-ordination of cell division and also on the control of the direction of cell division. Establishment of polarity division axis, correct alignment of the mitotic spindle, segregation of fate determinants equally or unequally between daughter cells, are essential for the realization of oriented cell division. Furthermore, oriented cell division is regulated by intrinsic cues, extrinsic cues and other cues, such as cell geometry and polarity. However, dysregulation of cell division orientation could lead to abnormal tissue development and function. In the present study, we review recent studies on the molecular mechanism of cell division orientation and explain their new roles in skin repair and regeneration.
Collapse
|
33
|
Vapniarsky N, Arzi B, Hu JC, Nolta JA, Athanasiou KA. Concise Review: Human Dermis as an Autologous Source of Stem Cells for Tissue Engineering and Regenerative Medicine. Stem Cells Transl Med 2015; 4:1187-98. [PMID: 26253713 DOI: 10.5966/sctm.2015-0084] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/08/2015] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED The exciting potential for regenerating organs from autologous stem cells is on the near horizon, and adult dermis stem cells (DSCs) are particularly appealing because of the ease and relative minimal invasiveness of skin collection. A substantial number of reports have described DSCs and their potential for regenerating tissues from mesenchymal, ectodermal, and endodermal lineages; however, the exact niches of these stem cells in various skin types and their antigenic surface makeup are not yet clearly defined. The multilineage potential of DSCs appears to be similar, despite great variability in isolation and in vitro propagation methods. Despite this great potential, only limited amounts of tissues and clinical applications for organ regeneration have been developed from DSCs. This review summarizes the literature on DSCs regarding their niches and the specific markers they express. The concept of the niches and the differentiation capacity of cells residing in them along particular lineages is discussed. Furthermore, the advantages and disadvantages of widely used methods to demonstrate lineage differentiation are considered. In addition, safety considerations and the most recent advancements in the field of tissue engineering and regeneration using DSCs are discussed. This review concludes with thoughts on how to prospectively approach engineering of tissues and organ regeneration using DSCs. Our expectation is that implementation of the major points highlighted in this review will lead to major advancements in the fields of regenerative medicine and tissue engineering. SIGNIFICANCE Autologous dermis-derived stem cells are generating great excitement and efforts in the field of regenerative medicine and tissue engineering. The substantial impact of this review lies in its critical coverage of the available literature and in providing insight regarding niches, characteristics, and isolation methods of stem cells derived from the human dermis. Furthermore, it provides analysis of the current state-of-the-art regenerative approaches using human-derived dermal stem cells, with consideration of current guidelines, to assist translation toward therapeutic use.
Collapse
Affiliation(s)
- Natalia Vapniarsky
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| | - Boaz Arzi
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| | - Jan A Nolta
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| |
Collapse
|
34
|
Li P, Zhang L. Exogenous Nkx2.5- or GATA-4-transfected rabbit bone marrow mesenchymal stem cells and myocardial cell co-culture on the treatment of myocardial infarction in rabbits. Mol Med Rep 2015; 12:2607-21. [PMID: 25975979 PMCID: PMC4464300 DOI: 10.3892/mmr.2015.3775] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 02/23/2015] [Indexed: 02/06/2023] Open
Abstract
The present study aimed to investigate the effects of Nkx2.5 or GATA-4 transfection with myocardial extracellular environment co-culture on the transformation of bone marrow mesenchymal stem cells (BMSCs) into differentiated cardiomyocytes. Nkx2.5 or GATA-4 were transfected into myocardial extracellular environment co-cultured BMSCs, and then injected into the periphery of infarcted myocardium of a myocardial infarction rabbit model. The effects of these gene transfections and culture on the infarcted myocardium were observed and the results may provide an experimental basis for the efficient myocardial cell differentiation of BMSCs. The present study also suggested that these cells may provide a source and clinical basis for myocardial injury repair via stem cell transplantation. The present study examined whether Nkx2.5 or GATA-4 exogenous gene transfection with myocardial cell extracellular environment co-culture were able to induce the differentiation of BMSCs into cardiac cells. In addition, the effect of these transfected BMSCs on the repair of the myocardium following myocardial infarction was determined using New Zealand rabbit models. The results demonstrated that myocardial cell differentiation was significantly less effective following exogenous gene transfection of Nkx2.5 or GATA-4 alone compared with that of transfection in combination with extracellular environment co-culture. In addition, the results of the present study showed that exogenous gene transfection of Nkx2.5 or GATA-4 into myocardial cell extracellular environment co-cultured BMSCs was able to significantly enhance the ability to repair, mitigating the death of myocardial cells and activation of the myocardium in rabbits with myocardial infarction compared with those of the rabbits transplanted with untreated BMSCs. In conclusion, the exogenous Nkx2.5 and GATA-4 gene transfection into myocardial extracellular environment co-cultured BMSCs induced increased differentiation into myocardial cells compared with that of gene transfection alone. Furthermore, significantly enhanced reparative effects were observed in the myocardium of rabbits following treatment with Nkx2.5- or GATA-4-transfected myocardial cell extracellular environment co-cultured BMSCs compared with those treated with untreated BMSCs.
Collapse
Affiliation(s)
- Pu Li
- Department of Cardiac Surgery, The Third Hospital of Hebei Medical University, Hebei, Shijiazhuang 050017, P.R. China
| | - Lei Zhang
- Department of Histology and Embryology, Hebei Medical University, Hebei, Shijiazhuang 050017, P.R. China
| |
Collapse
|
35
|
Skin-derived precursor cells promote angiogenesis and stimulate proliferation of endogenous neural stem cells after cerebral infarction. BIOMED RESEARCH INTERNATIONAL 2015; 2015:945846. [PMID: 25883981 PMCID: PMC4391522 DOI: 10.1155/2015/945846] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 01/03/2015] [Accepted: 01/05/2015] [Indexed: 01/10/2023]
Abstract
Stroke is one of the most common diseases that caused high mortality and has become burden to the health care systems. Stem cell transplantation has shown therapeutic effect in ameliorating ischemic damage after cerebral artery occlusion mainly due to their neurogenesis, immune regulation, or effects on the plasticity, proliferation, and survival of host cells. Recent studies demonstrated that skin-derived precursor cells (SKPs) could promote central nervous system regeneration in spinal cord injury model or the neonatal peripheral neuron. Here, we investigated the therapeutic potential of SKPs in a rat model of cerebral ischemia. SKPs were isolated, expanded, and transplanted into rat cortex and striatum after transient middle cerebral artery occlusion. Our results revealed that SKPs transplantation could improve the behavioral measures of neurological deficit. Moreover, immunohistology confirmed that SKPs could secrete basic FGF and VEGF in the ischemic region and further markedly increase the proliferation of endogenous nestin+ and βIII-tubulin+ neural stem cells. Furthermore, increased angiogenesis induced by SKPs was observed by vWF and α-SMA staining. These data suggest that SKPs induced endogenous neurogenesis and angiogenesis and protected neuron from hypoxic-ischemic environment. In conclusion, SKPs transplantation may be a promising approach in treatment of stroke.
Collapse
|
36
|
Differentiation of human skin-derived precursor cells into functional islet-like insulin-producing cell clusters. In Vitro Cell Dev Biol Anim 2015; 51:595-603. [DOI: 10.1007/s11626-015-9866-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/01/2015] [Indexed: 01/09/2023]
|
37
|
White Matter Repair: Skin-Derived Precursors as a Source of Myelinating Cells. Can J Neurol Sci 2014; 37 Suppl 2:S34-41. [DOI: 10.1017/s0317167100022411] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
ABSTRACT:Stem cell based therapies hold great promise for repair and functional restoration following neurological injury and disease. Skin-derived precursors (or “SKPs”) are a novel, multipotent somatic stem cell that resides within the mammalian dermis. SKPs persist within the skin throughout adulthood and yet intriguingly, exhibit many similarities to embryonic neural crest stem cells (NCSCs). For example, SKPs give rise to both neural and mesodermal cell types, and the former appear biased to peripheral nervous system fates. As such, SKPs are capable of generating Schwann cells, the myelinating glial cell of the peripheral nervous system. Here we discuss our current understanding of the biological origin of SKPs and specifically the potential therapeutic utility of SKPs as a highly accessible and autologous source of Schwann cells for remyelination and repair of the injured or diseased nervous system.
Collapse
|
38
|
Zhang P, Lu X, Chen J, Chen Z. Schwann cells originating from skin-derived precursors promote peripheral nerve regeneration in rats. Neural Regen Res 2014; 9:1696-702. [PMID: 25374591 PMCID: PMC4211190 DOI: 10.4103/1673-5374.141805] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2014] [Indexed: 02/04/2023] Open
Abstract
Artificial guidance channels containing Schwann cells can promote the regeneration of injured peripheral nerve over long distances. However, primary Schwann cells are not suitable for autotransplantation. Under specific conditions, skin-derived progenitors can be induced to differentiate into Schwann cells. Therefore, adult rat dorsal skin (dermis)-derived progenitors were isolated and induced to differentiate with DMEM/F12 containing B27, neuregulin 1, and forskolin. Immunofluorescence staining and reverse transcription polymerase chain reaction (RT-PCR) confirmed that the resultant cells were indeed Schwann cells. Artificial guidance channels containing skin-derived progenitors, Schwann cells originating from skin-derived progenitors, or primary Schwann cells were used to bridge 5 mm sciatic nerve defects. Schwann cells originating from skin-derived progenitors significantly promoted sciatic nerve axonal regeneration. The significant recovery of injured rat sciatic nerve function after the transplantation of Schwann cells originating from skin-derived progenitors was confirmed by electromyogram. The therapeutic effect of Schwann cells originating from skin-derived progenitors was better than that of skin-derived progenitors. These findings indicate that Schwann cells originating from skin-derived precursors can promote peripheral nerve regeneration in rats.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Orthopedics, Wuhan Women and Children Health Care Center, Wuhan, Hubei Province, China
| | - Xiaocheng Lu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jianghai Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
39
|
Kunisada T, Tezulka KI, Aoki H, Motohashi T. The stemness of neural crest cells and their derivatives. ACTA ACUST UNITED AC 2014; 102:251-62. [DOI: 10.1002/bdrc.21079] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/22/2014] [Indexed: 01/22/2023]
Affiliation(s)
- Takahiro Kunisada
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science; Gifu University Graduate School of Medicine, 1-1, Yanagido; Gifu 501-1194 Japan
| | - Ken-Ichi Tezulka
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science; Gifu University Graduate School of Medicine, 1-1, Yanagido; Gifu 501-1194 Japan
| | - Hitomi Aoki
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science; Gifu University Graduate School of Medicine, 1-1, Yanagido; Gifu 501-1194 Japan
| | - Tsutomu Motohashi
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science; Gifu University Graduate School of Medicine, 1-1, Yanagido; Gifu 501-1194 Japan
| |
Collapse
|
40
|
Häkkinen L, Larjava H, Fournier BPJ. Distinct phenotype and therapeutic potential of gingival fibroblasts. Cytotherapy 2014; 16:1171-86. [PMID: 24934304 DOI: 10.1016/j.jcyt.2014.04.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 03/13/2014] [Accepted: 04/04/2014] [Indexed: 01/15/2023]
Abstract
Gingiva of the oral mucosa provides a practical source to isolate fibroblasts for therapeutic purposes because the tissue is easily accessible, tissue discards are common during routine clinical procedures and wound healing after biopsy is fast and results in complete wound regeneration with very little morbidity or scarring. In addition, gingival fibroblasts have unique traits, including neural crest origin, distinct gene expression and synthetic properties and potent immunomodulatory functions. These characteristics may provide advantages for certain therapeutic approaches over other more commonly used cells, including skin fibroblasts, both in intraoral and extra-oral sites. However, identity and phenotype of gingival fibroblasts, like other fibroblasts, are still not completely understood. Gingival fibroblasts are phenotypically heterogeneous, and these…fibroblast subpopulations may play different roles in tissue maintenance, regeneration and pathologies. The purpose of this review is to summarize what is currently known about gingival fibroblasts, their distinct potential for tissue regeneration and their potential therapeutic uses in the future.
Collapse
Affiliation(s)
- Lari Häkkinen
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada.
| | - Hannu Larjava
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Benjamin P J Fournier
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada; Paris Diderot University, Dental School, Rotschild Hospital, AP-HP, Paris, France; UMRS872, Team 5, Molecular Oral Physiopathology, CRC Les Cordeliers, Paris, 75006, INSERM UMRS872, Pierre et Marie Curie University, Paris Descartes University, Paris, France
| |
Collapse
|
41
|
Matsushita T, Fujihara A, Royall L, Kagiwada S, Kosaka M, Araki M. Immediate differentiation of neuronal cells from stem/progenitor-like cells in the avian iris tissues. Exp Eye Res 2014; 123:16-26. [DOI: 10.1016/j.exer.2014.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 04/04/2014] [Accepted: 04/07/2014] [Indexed: 01/19/2023]
|
42
|
Shu B, Xie JL, Xu YB, Yu JX, Shi Y, Liu J, Wang P, Liu XS, Qi SH. Directed differentiation of skin-derived precursors into fibroblast-like cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:1478-1486. [PMID: 24817943 PMCID: PMC4014227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 02/25/2014] [Indexed: 06/03/2023]
Abstract
Skin-derived precursors (SKPs), which are located at skin's dermis, display multi-lineage potential and can produce both neural and mesodermal progeny in vitro. SKPs are considered to take part in dermal reconstruction and may be an important source of fibroblast during wound repairing. To explore the possibility of differentiation of SKPs into fibroblasts, the 3(rd) passage SKPs were treated with 0, 20, 40, 100, or 500 ng/ml human recombinant connective tissue growth factor (CTGF) for 48 h or treated with 100 ng/ml CTGF for 0, 24, 48, 72, or 96 h. Subsequently, a series of methods were to be used to observe cells immunocytochemistry changes under fluorescence microscope, to validate the mRNA expression change of collagen I, collagen III, fibroblast-specific protein 1 (FSP-1) and alpha smooth muscle actin (α-SMA) by quantitative real-time reverse transcriptase polymerase chain reaction (QRT-PCR), to analyze the expression of collagen I and collagen III protein by Enzyme-linked immunosorbent assay (ELISA), to semiquantitatively measure the expression of FSP-1 and α-SMA by western-blot. After differentiation, cells showed that positively staining for collagen I, collagen III, α-SMA, and FSP-1, which are markers for fibroblasts, but negative expression for neural precursors. The effects of CTGF on collagen I, collagen III, FSP-1 and α-SMA in SKPs were detected both on the transcriptional and posttranscriptional levels. These findings indicate that SKPs can be induced to differentiate into fibroblast-like cells with CTGF treatment that may be a key source of fibroblast in wound healing.
Collapse
Affiliation(s)
- Bin Shu
- Department of Burns, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, Guangdong, China
| | - Ju-Lin Xie
- Department of Burns, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, Guangdong, China
| | - Ying-Bin Xu
- Department of Burns, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, Guangdong, China
| | - Jian-Xing Yu
- Center of Laser Cosmetic, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, Guangdong, China
| | - Yan Shi
- Department of Burns, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, Guangdong, China
| | - Jian Liu
- Department of Burns, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, Guangdong, China
| | - Peng Wang
- Department of Burns, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, Guangdong, China
| | - Xu-Sheng Liu
- Department of Burns, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, Guangdong, China
| | - Shao-Hai Qi
- Department of Burns, The First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, Guangdong, China
| |
Collapse
|
43
|
Kinsler VA, Anderson G, Latimer B, Natarajan D, Healy E, Moore GE, Sebire NJ. Immunohistochemical and ultrastructural features of congenital melanocytic naevus cells support a stem-cell phenotype. Br J Dermatol 2014; 169:374-83. [PMID: 23517330 PMCID: PMC3838625 DOI: 10.1111/bjd.12323] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2013] [Indexed: 01/23/2023]
Abstract
Background Multiple congenital melanocytic naevi (CMN) in one individual are caused by somatic mosaicism for NRAS mutations; however, the lineage of the mutated cells remains uncertain. Objectives To test the hypothesis that CMN may be derived from cutaneous stem cells. Methods Sixty-six CMN samples from 44 patients were stained for immunohistochemical (IHC) markers of melanocytic differentiation (TYR, TRP1, TRP2, LEF1, MITF, cKit), pluripotency (nestin, fascin, CD133, CD20, CD34), monocyte/macrophage lineage (CD68, CD163, CD14), proliferation (Ki67) and MTOR/Wnt-signalling pathway activation (pS6, β-catenin). Semiquantitative scoring compared samples with naevus cell nesting (group 1) with those with only diffuse dermal infiltration (group 2). Transmission electron microscopy (TEM) was performed on 10 samples. Results A normal melanocyte population was seen overlying many dermal CMN. Group 1 samples were significantly more likely to express melanocytic differentiation markers than group 2, and expression decreased significantly with depth. Expression of these markers was correlated with each other, and with nestin and fascin. CD20 staining was positive in a substantial proportion and was stronger superficially. Expression of β-catenin and pS6 was almost universal. Some samples expressed monocyte/macrophage markers. TEM revealed variable naevus cell morphology, striking macromelanosomes, double cilia and microvilli. Conclusions Congenital melanocytic naevi development frequently coexists with normal overlying melanocyte development, leading us to hypothesize that in these cases CMN are likely to develop from a cell present in the skin independent of, or remaining after, normal melanocytic migration. IHC and TEM findings are compatible with CMN cells being of cutaneous stem-cell origin, capable of some degree of melanocytic differentiation superficially.
Collapse
Affiliation(s)
- V A Kinsler
- Paediatric Dermatology, Great Ormond Street Hospital for Children, London, WC1N 3JH, UK.
| | | | | | | | | | | | | |
Collapse
|
44
|
Kaucká M, Adameyko I. Non-canonical functions of the peripheral nerve. Exp Cell Res 2014; 321:17-24. [DOI: 10.1016/j.yexcr.2013.10.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 10/01/2013] [Accepted: 10/05/2013] [Indexed: 12/24/2022]
|
45
|
Adipose stromal cells contain phenotypically distinct adipogenic progenitors derived from neural crest. PLoS One 2013; 8:e84206. [PMID: 24391913 PMCID: PMC3877257 DOI: 10.1371/journal.pone.0084206] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 11/13/2013] [Indexed: 12/11/2022] Open
Abstract
Recent studies have shown that adipose-derived stromal/stem cells (ASCs) contain phenotypically and functionally heterogeneous subpopulations of cells, but their developmental origin and their relative differentiation potential remain elusive. In the present study, we aimed at investigating how and to what extent the neural crest contributes to ASCs using Cre-loxP-mediated fate mapping. ASCs harvested from subcutaneous fat depots of either adult P0-Cre/or Wnt1-Cre/Floxed-reporter mice contained a few neural crest-derived ASCs (NCDASCs). This subpopulation of cells was successfully expanded in vitro under standard culture conditions and their growth rate was comparable to non-neural crest derivatives. Although NCDASCs were positive for several mesenchymal stem cell markers as non-neural crest derivatives, they exhibited a unique bipolar or multipolar morphology with higher expression of markers for both neural crest progenitors (p75NTR, Nestin, and Sox2) and preadipocytes (CD24, CD34, S100, Pref-1, GATA2, and C/EBP-delta). NCDASCs were able to differentiate into adipocytes with high efficiency but their osteogenic and chondrogenic potential was markedly attenuated, indicating their commitment to adipogenesis. In vivo, a very small proportion of adipocytes were originated from the neural crest. In addition, p75NTR-positive neural crest-derived cells were identified along the vessels within the subcutaneous adipose tissue, but they were negative for mural and endothelial markers. These results demonstrate that ASCs contain neural crest-derived adipocyte-restricted progenitors whose phenotype is distinct from that of non-neural crest derivatives.
Collapse
|
46
|
Fournier BPJ, Larjava H, Häkkinen L. Gingiva as a source of stem cells with therapeutic potential. Stem Cells Dev 2013; 22:3157-77. [PMID: 23944935 DOI: 10.1089/scd.2013.0015] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Postnatal connective tissues contain phenotypically heterogeneous cells populations that include distinct fibroblast subpopulations, pericytes, myofibroblasts, fibrocytes, and tissue-specific mesenchymal stem cells (MSCs). These cells play key roles in tissue development, maintenance, and repair and contribute to various pathologies. Depending on the origin of tissue, connective tissue cells, including MSCs, have different phenotypes. Understanding the identity and specific functions of these distinct tissue-specific cell populations may allow researchers to develop better treatment modalities for tissue regeneration and find novel approaches to prevent pathological conditions. Interestingly, MSCs from adult oral mucosal gingiva possess distinct characteristics, including neural crest origin, multipotent differentiation capacity, fetal-like phenotype, and potent immunomodulatory properties. These characteristics and an easy, relatively noninvasive access to gingival tissue, and fast tissue regeneration after tissue biopsy make gingiva an attractive target for cell isolation for therapeutic purposes aiming to promote tissue regeneration and fast, scar-free wound healing. The purpose of this review is to discuss the identity, phenotypical heterogeneity, and function of gingival MSCs and summarize what is currently known about their properties, role in scar-free healing, and their future therapeutic potential.
Collapse
Affiliation(s)
- Benjamin P J Fournier
- 1 Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia , Vancouver, Canada
| | | | | |
Collapse
|
47
|
Tolg C, Ahsan A, Dworski S, Kirwan T, Yu J, Aitken K, Bägli DJ. Pathologic bladder microenvironment attenuates smooth muscle differentiation of skin derived precursor cells: implications for tissue regeneration. PLoS One 2013; 8:e59413. [PMID: 23560047 PMCID: PMC3613403 DOI: 10.1371/journal.pone.0059413] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 02/14/2013] [Indexed: 12/17/2022] Open
Abstract
Smooth muscle cell containing organs (bladder, heart, blood vessels) are damaged by a variety of pathological conditions necessitating surgery or organ replacement. Currently, regeneration of contractile tissues is hampered by lack of functional smooth muscle cells. Multipotent skin derived progenitor cells (SKPs) can easily be isolated from adult skin and can be differentiated in vitro into contractile smooth muscle cells by exposure to FBS. Here we demonstrate an inhibitory effect of a pathologic contractile organ microenvironment on smooth muscle cell differentiation of SKPs. In vivo, urinary bladder strain induces microenvironmental changes leading to de-differentiation of fully differentiated bladder smooth muscle cells. Co-culture of SKPs with organoids isolated from ex vivo stretched bladders or exposure of SKPs to diffusible factors released by stretched bladders (e.g. bFGF) suppresses expression of smooth muscle markers (alpha SMactin, calponin, myocardin, myosin heavy chain) as demonstrated by qPCR and immunofluorescent staining. Rapamycin, an inhibitor of mTOR signalling, previously observed to prevent bladder strain induced de-differentiation of fully differentiated smooth muscle cells in vitro, inhibits FBS-induced smooth muscle cell differentiation of undifferentiated SKPs. These results suggest that intended precursor cell differentiation may be paradoxically suppressed by the disease context for which regeneration may be required. Organ-specific microenvironment contexts, particularly prevailing disease, may play a significant role in modulating or attenuating an intended stem cell phenotypic fate, possibly explaining the variable and inefficient differentiation of stem cell constructs in in vivo settings. These observations must be considered in drafting any regeneration strategies.
Collapse
Affiliation(s)
- Cornelia Tolg
- Developmental and Stem Cell Biology, Research Institute, Toronto, Ontario, Canada
| | - Alya Ahsan
- Developmental and Stem Cell Biology, Research Institute, Toronto, Ontario, Canada
| | - Shaalee Dworski
- Developmental and Stem Cell Biology, Research Institute, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Tyler Kirwan
- Developmental and Stem Cell Biology, Research Institute, Toronto, Ontario, Canada
| | - Jeffery Yu
- Developmental and Stem Cell Biology, Research Institute, Toronto, Ontario, Canada
| | - Karen Aitken
- Developmental and Stem Cell Biology, Research Institute, Toronto, Ontario, Canada
- Division of Urology, Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
| | - Darius Jehan Bägli
- Developmental and Stem Cell Biology, Research Institute, Toronto, Ontario, Canada
- Division of Urology, Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Departments of Surgery & Physiology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
48
|
The characterisation of Pax3 expressant cells in adult peripheral nerve. PLoS One 2013; 8:e59184. [PMID: 23527126 PMCID: PMC3602598 DOI: 10.1371/journal.pone.0059184] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 02/12/2013] [Indexed: 12/25/2022] Open
Abstract
Pax3 has numerous integral functions in embryonic tissue morphogenesis and knowledge of its complex function in cells of adult tissue continues to unfold. Across a variety of adult tissue lineages, the role of Pax3 is principally linked to maintenance of the tissue’s resident stem/progenitor cell population. In adult peripheral nerves, Pax3 is reported to be expressed in nonmyelinating Schwann cells, however, little is known about the purpose of this expression. Based on the evidence of the role of Pax3 in other adult tissue stem and progenitor cells, it was hypothesised that the cells in adult peripheral nerve that express Pax3 may be peripheral glioblasts. Here, methods have been developed for identification and visualisation of Pax3 expressant cells in normal 60 day old mouse peripheral nerve that allowed morphological and phenotypic distinctions to be made between Pax3 expressing cells and other nonmyelinating Schwann cells. The distinctions described provide compelling support for a resident glioblast population in adult mouse peripheral nerve.
Collapse
|
49
|
Chen Z, Pradhan S, Liu C, Le LQ. Skin-derived precursors as a source of progenitors for cutaneous nerve regeneration. Stem Cells 2013; 30:2261-70. [PMID: 22851518 DOI: 10.1002/stem.1186] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Peripheral nerves have the potential to regenerate axons and reinnervate end organs. Chronic denervation and disturbed nerve regeneration are thought to contribute to peripheral neuropathy, pain, and pruritus in the skin. The capacity of denervated distal nerves to support axonal regeneration requires proliferation by Schwann cells, which guide regenerating axons to their denervated targets. However, adult peripheral nerve Schwann cells do not retain a growth-permissive phenotype, as is required to produce new glia. Therefore, it is believed that following injury, mature Schwann cells dedifferentiate to a progenitor/stem cell phenotype to promote axonal regrowth. In this study, we show that skin-derived precursors (SKPs), a recently identified neural crest-related stem cell population in the dermis of skin, are an alternative source of progenitors for cutaneous nerve regeneration. Using in vivo and in vitro three-dimensional cutaneous nerve regeneration models, we show that the SKPs are neurotropic toward injured nerves and that they have a full capacity to differentiate into Schwann cells and promote axon regeneration. The identification of SKPs as a physiologic source of progenitors for cutaneous nerve regeneration in the skin, where SKPs physiologically reside, has important implications for understanding early cellular events in peripheral nerve regeneration. It also provides fertile ground for the elucidation of intrinsic and extrinsic factors within the nerve microenvironment that likely play essential roles in cutaneous nerve homeostasis.
Collapse
Affiliation(s)
- Zhiguo Chen
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9069, USA
| | | | | | | |
Collapse
|
50
|
Machens HG, Günter CI, Bader A. Skin. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|