51
|
Pattison JM, Melo SP, Piekos SN, Torkelson JL, Bashkirova E, Mumbach MR, Rajasingh C, Zhen HH, Li L, Liaw E, Alber D, Rubin AJ, Shankar G, Bao X, Chang HY, Khavari PA, Oro AE. Retinoic acid and BMP4 cooperate with p63 to alter chromatin dynamics during surface epithelial commitment. Nat Genet 2018; 50:1658-1665. [PMID: 30397335 PMCID: PMC6265075 DOI: 10.1038/s41588-018-0263-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 09/21/2018] [Indexed: 01/14/2023]
Abstract
Human embryonic stem cell (hESC) differentiation promises advances in regenerative medicine1–3, yet conversion into transplantable tissues remains poorly understood. Using our keratinocyte differentiation system, we employ a multi-dimensional genomics approach to interrogate the contributions of inductive morphogens retinoic acid (RA) and bone morphogenetic protein 4 (BMP4) and the epidermal master regulator p634,5 during surface ectoderm commitment. In contrast to other master regulators6–9, p63 effects major transcriptional changes only after morphogens alter chromatin accessibility, establishing an epigenetic landscape for p63 to modify. p63 distally closes chromatin accessibility and promotes accumulation of H3K27me3 modifications. Cohesin HiChIP10 visualizations of chromosome conformation reveal that p63 and the morphogens contribute to dynamic long-range chromatin interactions, as illustrated with TFAP2C regulation11. Our study demonstrates the unexpected dependency of p63 on morphogenetic signaling and provides novel insights into how a master regulator can specify diverse transcriptional programs based on the chromatin landscape induced by specific morphogen exposure.
Collapse
Affiliation(s)
- Jillian M Pattison
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sandra P Melo
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.,Agilent Technologies, Santa Clara, CA, USA
| | - Samantha N Piekos
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jessica L Torkelson
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Elizaveta Bashkirova
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.,Columbia Stem Cell Initiative, Columbia University Medical Center, New York, NY, USA
| | - Maxwell R Mumbach
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.,Center for Personal Dynamic Regulomes, Stanford, CA, USA
| | - Charlotte Rajasingh
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hanson Hui Zhen
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lingjie Li
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Eric Liaw
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.,David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Daniel Alber
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Adam J Rubin
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Gautam Shankar
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaomin Bao
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.,Departments of Molecular Sciences and Dermatology, Northwestern University, Evanston, IL, USA
| | - Howard Y Chang
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.,Center for Personal Dynamic Regulomes, Stanford, CA, USA.,Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Paul A Khavari
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anthony E Oro
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
52
|
|
53
|
Kim Y, Park N, Rim YA, Nam Y, Jung H, Lee K, Ju JH. Establishment of a complex skin structure via layered co-culture of keratinocytes and fibroblasts derived from induced pluripotent stem cells. Stem Cell Res Ther 2018; 9:217. [PMID: 30103800 PMCID: PMC6090613 DOI: 10.1186/s13287-018-0958-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022] Open
Abstract
Background Skin is an organ that plays an important role as a physical barrier and has many other complex functions. Skin mimetics may be useful for studying the pathophysiology of diseases in vitro and for repairing lesions in vivo. Cord blood mononuclear cells (CBMCs) have emerged as a potential cell source for regenerative medicine. Human induced pluripotent stem cells (iPSCs) derived from CBMCs have great potential for allogenic regenerative medicine. Further study is needed on skin differentiation using CBMC-iPSCs. Methods Human iPSCs were generated from CBMCs by Sendai virus. CBMC-iPSCs were differentiated to fibroblasts and keratinocytes using embryonic body formation. To generate CBMC-iPSC-derived 3D skin organoid, CBMC-iPSC-derived fibroblasts were added into the insert of a Transwell plate and CBMC-iPSC-derived keratinocytes were seeded onto the fibroblast layer. Transplantation of 3D skin organoid was performed by the tie-over dressing method. Results Epidermal and dermal layers were developed using keratinocytes and fibroblasts differentiated from cord blood-derived human iPSCs, respectively. A complex 3D skin organoid was generated by overlaying the epidermal layer onto the dermal layer. A humanized skin model was generated by transplanting this human skin organoid into SCID mice and effectively healed skin lesions. Conclusions This study reveals that a human skin organoid generated using CBMC iPSCs is a novel tool for in-vitro and in-vivo dermatologic research. Electronic supplementary material The online version of this article (10.1186/s13287-018-0958-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yena Kim
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea
| | - Narae Park
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea
| | - Yeri Alice Rim
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea
| | - Yoojun Nam
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea
| | - Hyerin Jung
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea
| | - Kijun Lee
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea
| | - Ji Hyeon Ju
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea. .,Division of Rheumatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea.
| |
Collapse
|
54
|
Fard M, Akhavan-Tavakoli M, Khanjani S, Zare S, Edalatkhah H, Arasteh S, Mehrabani D, Zarnani AH, Kazemnejad S, Shirazi R. Bilayer Amniotic Membrane/Nano-fibrous Fibroin Scaffold Promotes Differentiation Capability of Menstrual Blood Stem Cells into Keratinocyte-Like Cells. Mol Biotechnol 2018; 60:100-110. [PMID: 29247317 DOI: 10.1007/s12033-017-0049-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The skin provides a dynamic barrier separating and protecting human body from the exterior world, and then immediate repair and rebuilding of the epidermal barrier is crucial after wound and injury. Wound healing without scars and complete regeneration of skin tissue still remain as a clinical challenge. The demand to engineer scaffolds that actively promote regeneration of damaged areas of the skin has been increased. In this study, menstrual blood-derived stem cells (MenSCs) have been induced to differentiate into keratinocytes-like cells in the presence of human foreskin-derived keratinocytes on a bilayer scaffold based on amniotic membrane and silk fibroin. Based on the findings, newly differentiated keratinocytes from MenSCs successfully expressed the keratinocytes specific markers at both mRNA and protein levels judged by real-time PCR and immunostaining techniques, respectively. We could show that the differentiated cells over bilayer composite scaffolds express the keratinocytes specific markers at higher levels when compared with those cultured in conventional 2D culture system. Based on these findings, bilayer amniotic membrane/nano-fibrous fibroin scaffold represents an efficient natural construct with broad applicability to generate keratinocytes from MenSCs for stem cell-based skin wounds healing and regeneration.
Collapse
Affiliation(s)
- Maryam Fard
- Department of Anatomical Sciences, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Maryam Akhavan-Tavakoli
- Department of Anatomical Sciences, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sayeh Khanjani
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, P.O. Box: 1177-19615, Tehran, Iran
| | - Sona Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Haleh Edalatkhah
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, P.O. Box: 1177-19615, Tehran, Iran
| | - Shaghayegh Arasteh
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, P.O. Box: 1177-19615, Tehran, Iran
| | - Davood Mehrabani
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Amir-Hassan Zarnani
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Somaieh Kazemnejad
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, P.O. Box: 1177-19615, Tehran, Iran.
| | - Reza Shirazi
- Department of Anatomical Sciences, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran.
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences, P.O. Box: 1449614525, Tehran, Iran.
| |
Collapse
|
55
|
Oh JY, Suh HN, Choi GE, Lee HJ, Jung YH, Ko SH, Kim JS, Chae CW, Lee CK, Han HJ. Modulation of sonic hedgehog-induced mouse embryonic stem cell behaviours through E-cadherin expression and integrin β1-dependent F-actin formation. Br J Pharmacol 2018; 175:3548-3562. [PMID: 29933500 DOI: 10.1111/bph.14423] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 05/23/2018] [Accepted: 06/12/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE The sonic hedgehog pathway (Shh) plays a central role in maintaining stem cell function and behaviour in various processes related to self-renewal and tissue regeneration. However, the therapeutic effect of Shh on mouse embryonic stem cells (mESCs) has not yet been clearly elucidated. Thus, we investigated the effect of Shh on the regulation of mESC behaviour as well as the effect of Shh-pretreated mESCs in skin wound healing. EXPERIMENTAL APPROACH The underlying mechanisms of Shh signalling pathway in growth and motility of mESCs were investigated using Western blot analysis, a cell proliferation assay and cell migration assay. In addition, the effect of Shh-pretreated mESCs in skin wound healing was determined using a mouse excisional wound splinting model. KEY RESULTS Shh disrupted the adherens junction through proteolysis by activating MMPs. In addition, the release of β-catenin from adherens junctions mediated by Shh led to cell cycle-dependent mESC proliferation. Shh-mediated Gli1 expression led to integrin β1 up-regulation, followed by FAK and Src phosphorylation. Furthermore, among the Rho-GTPases, Rac1 and Cdc42 were activated in a Shh-dependent manner while F-actin expression was suppressed by Rac1 and Cdc42 siRNA transfection. Consistent with the in vitro results, the skin wound healing assay revealed that Shh-treated mESCs increased angiogenesis and skin wound repair compared to that in Shh-treated mESCs transfected with integrin β1 siRNA in vivo. CONCLUSIONS AND IMPLICATIONS Our results imply that Shh induces adherens junction disruption and integrin β1-dependent F-actin formation by a mechanism involving FAK/Src and Rac1/Cdc42 signalling pathways in mESCs.
Collapse
Affiliation(s)
- Ji Young Oh
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea.,Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - Han Na Suh
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea.,Minipig Model Group, Animal Model Center, Korea Institute of Toxicology, Jeongeup-si, Jeollabuk-do, Korea
| | - Gee Euhn Choi
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - So Hee Ko
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - Jun Sung Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - Chang-Kyu Lee
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea.,Institute of Green Bio Science and Technology, Seoul National University, Pyeong Chang, Kangwon do, Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| |
Collapse
|
56
|
Harhouri K, Navarro C, Depetris D, Mattei MG, Nissan X, Cau P, De Sandre-Giovannoli A, Lévy N. MG132-induced progerin clearance is mediated by autophagy activation and splicing regulation. EMBO Mol Med 2018; 9:1294-1313. [PMID: 28674081 PMCID: PMC5582415 DOI: 10.15252/emmm.201607315] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a lethal premature and accelerated aging disease caused by a de novo point mutation in LMNA encoding A‐type lamins. Progerin, a truncated and toxic prelamin A issued from aberrant splicing, accumulates in HGPS cells' nuclei and is a hallmark of the disease. Small amounts of progerin are also produced during normal aging. We show that progerin is sequestered into abnormally shaped promyelocytic nuclear bodies, identified as novel biomarkers in late passage HGPS cell lines. We found that the proteasome inhibitor MG132 induces progerin degradation through macroautophagy and strongly reduces progerin production through downregulation of SRSF‐1 and SRSF‐5 accumulation, controlling prelamin A mRNA aberrant splicing. MG132 treatment improves cellular HGPS phenotypes. MG132 injection in skeletal muscle of LmnaG609G/G609G mice locally reduces SRSF‐1 expression and progerin levels. Altogether, we demonstrate progerin reduction based on MG132 dual action and shed light on a promising class of molecules toward a potential therapy for children with HGPS.
Collapse
Affiliation(s)
- Karim Harhouri
- Aix Marseille Univ, INSERM, GMGF (Génétique Médicale et Génomique Fonctionnelle), Marseille, France
| | - Claire Navarro
- Aix Marseille Univ, INSERM, GMGF (Génétique Médicale et Génomique Fonctionnelle), Marseille, France
| | - Danielle Depetris
- Aix Marseille Univ, INSERM, GMGF (Génétique Médicale et Génomique Fonctionnelle), Marseille, France
| | - Marie-Geneviève Mattei
- Aix Marseille Univ, INSERM, GMGF (Génétique Médicale et Génomique Fonctionnelle), Marseille, France
| | - Xavier Nissan
- CECS, I-STEM, Institut des cellules Souches pour le Traitement et l'Etude des maladies Monogéniques, AFM, Evry, France
| | - Pierre Cau
- Aix Marseille Univ, INSERM, GMGF (Génétique Médicale et Génomique Fonctionnelle), Marseille, France.,AP-HM, Hôpital la Timone, Service de Biologie Cellulaire, Marseille, France
| | - Annachiara De Sandre-Giovannoli
- Aix Marseille Univ, INSERM, GMGF (Génétique Médicale et Génomique Fonctionnelle), Marseille, France.,AP-HM, Hôpital la Timone, Département de Génétique Médicale, Marseille, France
| | - Nicolas Lévy
- Aix Marseille Univ, INSERM, GMGF (Génétique Médicale et Génomique Fonctionnelle), Marseille, France .,AP-HM, Hôpital la Timone, Département de Génétique Médicale, Marseille, France
| |
Collapse
|
57
|
Li Z, Maitz P. Cell therapy for severe burn wound healing. BURNS & TRAUMA 2018; 6:13. [PMID: 29854856 PMCID: PMC5971426 DOI: 10.1186/s41038-018-0117-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/26/2018] [Indexed: 12/14/2022]
Abstract
Cell therapy has emerged as an important component of life-saving procedures in treating burns. Over past decades, advances in stem cells and regenerative medicine have offered exciting opportunities of developing cell-based alternatives and demonstrated the potential and feasibility of various stem cells for burn wound healing. However, there are still scientific and technical issues that should be resolved to facilitate the full potential of the cellular devices. More evidence from large, randomly controlled trials is also needed to understand the clinical impact of cell therapy in burns. This article aims to provide an up-to-date review of the research development and clinical applications of cell therapies in burn wound healing and skin regeneration.
Collapse
Affiliation(s)
- Zhe Li
- Burns Unit, Concord Hospital, Concord, New South Wales 2139 Australia
- Skin Laboratory, NSW Statewide Burns Service, Concord, New South Wales Australia
- Discipline of Surgery, University of Sydney Medical School, Camperdown, New South Wales Australia
| | - Peter Maitz
- Burns Unit, Concord Hospital, Concord, New South Wales 2139 Australia
- Skin Laboratory, NSW Statewide Burns Service, Concord, New South Wales Australia
- Discipline of Surgery, University of Sydney Medical School, Camperdown, New South Wales Australia
| |
Collapse
|
58
|
Xue M, Zhao R, Lin H, Jackson C. Delivery systems of current biologicals for the treatment of chronic cutaneous wounds and severe burns. Adv Drug Deliv Rev 2018; 129:219-241. [PMID: 29567398 DOI: 10.1016/j.addr.2018.03.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 02/08/2018] [Accepted: 03/13/2018] [Indexed: 12/15/2022]
Abstract
While wound therapy remains a clinical challenge in current medical practice, much effort has focused on developing biological therapeutic approaches. This paper presents a comprehensive review of delivery systems for current biologicals for the treatment of chronic wounds and severe burns. The biologicals discussed here include proteins such as growth factors and gene modifying molecules, which may be delivered to wounds free, encapsulated, or released from living systems (cells, skin grafts or skin equivalents) or biomaterials. Advances in biomaterial science and technologies have enabled the synthesis of delivery systems such as scaffolds, hydrogels and nanoparticles, designed to not only allow spatially and temporally controlled release of biologicals, but to also emulate the natural extracellular matrix microenvironment. These technologies represent an attractive field for regenerative wound therapy, by offering more personalised and effective treatments.
Collapse
|
59
|
Abstract
The normal wound healing process involves a well-organized cascade of biological pathways and any failure in this process leads to wounds becoming chronic. Non-healing wounds are a burden on healthcare systems and set to increase with aging population and growing incidences of obesity and diabetes. Stem cell-based therapies have the potential to heal chronic wounds but have so far seen little success in the clinic. Current research has been focused on using polymeric biomaterial systems that can act as a niche for these stem cells to improve their survival and paracrine activity that would eventually promote wound healing. Furthermore, different modification strategies have been developed to improve stem cell survival and differentiation, ultimately promoting regenerative wound healing. This review focuses on advanced polymeric scaffolds that have been used to deliver stem cells and have been tested for their efficiency in preclinical animal models of wounds.
Collapse
|
60
|
Kajiwara K, Tanemoto T, Wada S, Karibe J, Ihara N, Ikemoto Y, Kawasaki T, Oishi Y, Samura O, Okamura K, Takada S, Akutsu H, Sago H, Okamoto A, Umezawa A. Fetal Therapy Model of Myelomeningocele with Three-Dimensional Skin Using Amniotic Fluid Cell-Derived Induced Pluripotent Stem Cells. Stem Cell Reports 2018; 8:1701-1713. [PMID: 28591652 PMCID: PMC5470234 DOI: 10.1016/j.stemcr.2017.05.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 01/28/2023] Open
Abstract
Myelomeningocele (MMC) is a congenital disease without genetic abnormalities. Neurological symptoms are irreversibly impaired after birth, and no effective treatment has been reported to date. Only surgical repairs have been reported so far. In this study, we performed antenatal treatment of MMC with an artificial skin using induced pluripotent stem cells (iPSCs) generated from a patient with Down syndrome (AF-T21-iPSCs) and twin-twin transfusion syndrome (AF-TTTS-iPSCs) to a rat model. We manufactured three-dimensional skin with epidermis generated from keratinocytes derived from AF-T21-iPSCs and AF-TTTS-iPSCs and dermis of human fibroblasts and collagen type I. For generation of epidermis, we developed a protocol using Y-27632 and epidermal growth factor. The artificial skin was successfully covered over MMC defect sites during pregnancy, implying a possible antenatal surgical treatment with iPSC technology.
Collapse
Affiliation(s)
- Kazuhiro Kajiwara
- Department of Reproductive Biology, Center for Regenerative Medicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya, Tokyo 157-8535, Japan; Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| | - Tomohiro Tanemoto
- Department of Medical Science, Chiba University Graduate School of Medicine, Chiba 260-0856, Japan
| | - Seiji Wada
- Maternal-Fetal, Neonatal and Reproductive Medicine, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Jurii Karibe
- Department of Reproductive Biology, Center for Regenerative Medicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya, Tokyo 157-8535, Japan
| | - Norimasa Ihara
- Department of Reproductive Biology, Center for Regenerative Medicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya, Tokyo 157-8535, Japan
| | - Yu Ikemoto
- Department of Reproductive Biology, Center for Regenerative Medicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya, Tokyo 157-8535, Japan
| | - Tomoyuki Kawasaki
- Department of Reproductive Biology, Center for Regenerative Medicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya, Tokyo 157-8535, Japan
| | - Yoshie Oishi
- Department of Reproductive Biology, Center for Regenerative Medicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya, Tokyo 157-8535, Japan
| | - Osamu Samura
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| | - Kohji Okamura
- Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Shuji Takada
- Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Hidenori Akutsu
- Department of Reproductive Biology, Center for Regenerative Medicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya, Tokyo 157-8535, Japan
| | - Haruhiko Sago
- Maternal-Fetal, Neonatal and Reproductive Medicine, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| | - Akihiro Umezawa
- Department of Reproductive Biology, Center for Regenerative Medicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya, Tokyo 157-8535, Japan.
| |
Collapse
|
61
|
Lee J, Bӧscke R, Tang PC, Hartman BH, Heller S, Koehler KR. Hair Follicle Development in Mouse Pluripotent Stem Cell-Derived Skin Organoids. Cell Rep 2018; 22:242-254. [PMID: 29298425 PMCID: PMC5806130 DOI: 10.1016/j.celrep.2017.12.007] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/01/2017] [Accepted: 12/04/2017] [Indexed: 12/17/2022] Open
Abstract
The mammalian hair follicle arises during embryonic development from coordinated interactions between the epidermis and dermis. It is currently unclear how to recapitulate hair follicle induction in pluripotent stem cell cultures for use in basic research studies or in vitro drug testing. To date, generation of hair follicles in vitro has only been possible using primary cells isolated from embryonic skin, cultured alone or in a co-culture with stem cell-derived cells, combined with in vivo transplantation. Here, we describe the derivation of skin organoids, constituting epidermal and dermal layers, from a homogeneous population of mouse pluripotent stem cells in a 3D culture. We show that skin organoids spontaneously produce de novo hair follicles in a process that mimics normal embryonic hair folliculogenesis. This in vitro model of skin development will be useful for studying mechanisms of hair follicle induction, evaluating hair growth or inhibitory drugs, and modeling skin diseases.
Collapse
Affiliation(s)
- Jiyoon Lee
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Robert Bӧscke
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Pei-Ciao Tang
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Byron H Hartman
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stefan Heller
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Karl R Koehler
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
62
|
Garcia M, Morello E, Garnier J, Barrault C, Garnier M, Burucoa C, Lecron JC, Si-Tahar M, Bernard FX, Bodet C. Pseudomonas aeruginosa flagellum is critical for invasion, cutaneous persistence and induction of inflammatory response of skin epidermis. Virulence 2018; 9:1163-1175. [PMID: 30070169 PMCID: PMC6086312 DOI: 10.1080/21505594.2018.1480830] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 05/21/2018] [Indexed: 12/22/2022] Open
Abstract
Pseudomonas aeruginosa, an opportunistic pathogen involved in skin and lung diseases, possesses numerous virulence factors, including type 2 and 3 secretion systems (T2SS and T3SS) and its flagellum, whose functions remain poorly known during cutaneous infection. Using isogenic mutants deleted from genes encoding each or all of these three virulence factors, we investigated their role in induction of inflammatory response and in tissue invasiveness in human primary keratinocytes and reconstructed epidermis. Our results showed that flagellum, but not T2SS and T3SS, is involved in induction of a large panel of cytokine, chemokine, and antimicrobial peptide (AMP) mRNA in the infected keratinocytes. Chemokine secretion and AMP tissular production were also dependent on the presence of the bacterial flagellum. This pro-inflammatory effect was significantly reduced in keratinocytes infected in presence of anti-toll-like receptor 5 (TLR5) neutralizing antibody. Bacterial invasion of human epidermis and persistence in a mouse model of sub-cutaneous infection were dependent on the P. aeruginosa flagellum. We demonstrated that flagellum constitutes the main virulence factor of P. aeruginosa involved not only in early induction of the epidermis inflammatory response but also in bacterial invasion and cutaneous persistence. P. aeruginosa is mainly sensed by TLR5 during the early innate immune response of human primary keratinocytes.
Collapse
Affiliation(s)
- Magali Garcia
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
-
Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers
, Poitiers, France
| | - Eric Morello
-
Centre d'Etude des Pathologies Respiratoires, INSERM UMR 1100, Université de Tours
, Tours, France
| | | | | | - Martine Garnier
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
| | - Christophe Burucoa
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
-
Laboratoire de Bactériologie et Hygiène, CHU de Poitiers
, Poitiers, France
| | - Jean-Claude Lecron
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
-
Laboratoire d’Immunologie et Inflammation, CHU de Poitiers
, Poitiers, France
| | - Mustapha Si-Tahar
-
Centre d'Etude des Pathologies Respiratoires, INSERM UMR 1100, Université de Tours
, Tours, France
| | | | - Charles Bodet
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
| |
Collapse
|
63
|
Artificial Pigmented Human Skin Created by Muse Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1103:255-271. [PMID: 30484234 DOI: 10.1007/978-4-431-56847-6_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The skin composes physiological and chemical barrier and renews skin component cells throughout the human life. Melanocytes locate in the basal layer of the epidermis and produce melanin to protect the skin from ultraviolet. Melanin plays key roles in determining human skin and hair color. Melanocyte dysfunction observed in albinism and vitiligo not only causes cosmetic problems but also increases risk of skin cancer. As rejuvenate therapy, embryonic stem (ES) cells and induced pluripotent stem (iPS) cells have been reported to generate melanocytes. Other than ES and iPS cells, human skin tissues maintain pluripotent stem cells, named multilineage-differentiating stress-enduring (Muse) cells. We employ Muse cells isolated from human fibroblasts and adipose tissue to differentiate into melanocytes (Muse-MC). Muse-MC express melanocyte-related molecules, such as tyrosinase and DCT, and show tyrosinase activity. We also succeeded to differentiate Muse cells into fibroblasts and keratinocytes and created three-dimensional (3D) reconstituted skin with Muse cell-derived melanocytes, fibroblasts, and keratinocytes. The 3D reconstituted skin of Muse cell-derived cells coordinately showed epidermis layers and Muse-MC localized in the basal layer of the epidermis. Thus Muse cells in the human skin can be a source of rejuvenation medicine for the skin reconstruction.
Collapse
|
64
|
Kamarudin TA, Bojic S, Collin J, Yu M, Alharthi S, Buck H, Shortt A, Armstrong L, Figueiredo FC, Lako M. Differences in the Activity of Endogenous Bone Morphogenetic Protein Signaling Impact on the Ability of Induced Pluripotent Stem Cells to Differentiate to Corneal Epithelial-Like Cells. Stem Cells 2017; 36:337-348. [PMID: 29226476 PMCID: PMC5839253 DOI: 10.1002/stem.2750] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/27/2017] [Accepted: 11/15/2017] [Indexed: 12/13/2022]
Abstract
Cornea is a clear outermost layer of the eye which enables transmission of light onto the retina. The transparent corneal epithelium is regenerated by limbal stem cells (LSCs), whose loss/dysfunction results in LSCs deficiency (LSCD). Ex vivo expansion of autologous LSCs obtained from patient's healthy eye followed by transplantation onto the LSCs damaged/deficient eye, has provided a successful treatment for unilateral LSCD. However, this is not applicable to patient with total bilateral LSCD, where LSCs are lost/damaged from both eyes. We investigated the potential of human induced pluripotent stem cell (hiPSC) to differentiate into corneal epithelial‐like cells as a source of autologous stem cell treatment for patients with total bilateral LSCD. Our study showed that combined addition of bone morphogenetic protein 4 (BMP4), all trans‐retinoic acid and epidermal growth factor for the first 9 days of differentiation followed by cell‐replating on collagen‐IV‐coated surfaces with a corneal‐specific‐epithelial cell media for an additional 11 days, resulted in step wise differentiation of human embryonic stem cells (hESC) to corneal epithelial progenitors and mature corneal epithelial‐like cells. We observed differences in the ability of hiPSC lines to undergo differentiation to corneal epithelial‐like cells which were dependent on the level of endogenous BMP signaling and could be restored via the activation of this signaling pathway by a specific transforming growth factor β inhibitor (SB431542). Together our data reveal a differential ability of hiPSC lines to generate corneal epithelial cells which is underlined by the activity of endogenous BMP signaling pathway. Stem Cells2018;36:337–348
Collapse
Affiliation(s)
- Taty Anna Kamarudin
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Sanja Bojic
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Joseph Collin
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Min Yu
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Sameer Alharthi
- Princess Al Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Saudi Arabia
| | - Harley Buck
- UCL Institute of Immunology and Transplantation, Royal Free Campus, London, United Kingdom
| | - Alex Shortt
- UCL Institute of Immunology and Transplantation, Royal Free Campus, London, United Kingdom
| | - Lyle Armstrong
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Francisco C Figueiredo
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Central Parkway, Newcastle upon Tyne, United Kingdom.,Department of Ophthalmology, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne, United Kingdom
| | - Majlinda Lako
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Central Parkway, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
65
|
Domingues S, Masson Y, Marteyn A, Allouche J, Perrier AL, Peschanski M, Martinat C, Baldeschi C, Lemaître G. Differentiation of nonhuman primate pluripotent stem cells into functional keratinocytes. Stem Cell Res Ther 2017; 8:285. [PMID: 29258610 PMCID: PMC5738144 DOI: 10.1186/s13287-017-0741-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/16/2017] [Accepted: 12/01/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Epidermal grafting using cells derived from pluripotent stem cells will change the face of this side of regenerative cutaneous medicine. To date, the safety of the graft would be the major unmet deal in order to implement long-term skin grafting. In this context, experiments on large animals appear unavoidable to assess this question and possible rejection. Cellular tools for large animal models should be constructed. METHODS In this study, we generated monkey pluripotent stem cell-derived keratinocytes and evaluated their capacities to reconstruct an epidermis, in vitro as well as in vivo. RESULTS Monkey pluripotent stem cells were differentiated efficiently into keratinocytes able to reconstruct fully epidermis presenting a low level of major histocompatibility complex class-I antigens, opening the way for autologous or allogeneic epidermal long-term grafting. CONCLUSIONS Functional keratinocytes generated from nonhuman primate embryonic stem cells and induced pluripotent stem cells reproduce an in-vitro and in-vivo stratified epidermis. These monkey skin grafts will be considered to model autologous or allogeneic epidermal grafting using either embryonic stem cells or induced pluripotent stem cells. This graft model will allow us to further investigate the safety, efficacy and immunogenicity of nonhuman primate PSC-derived epidermis in the perspective of human skin cell therapy.
Collapse
Affiliation(s)
- Sophie Domingues
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France.,Centre d'Etude des Cellules Souches (CECS), I-Stem, AFM, 91100, Corbeil Essonnes, France
| | - Yolande Masson
- Centre d'Etude des Cellules Souches (CECS), I-Stem, AFM, 91100, Corbeil Essonnes, France
| | - Aurore Marteyn
- Centre d'Etude des Cellules Souches (CECS), I-Stem, AFM, 91100, Corbeil Essonnes, France
| | - Jennifer Allouche
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France.,Université Evry Val d'Essonne (UEVE), Paris Saclay, U861, I-Stem, AFM, 91030, Evry Cedex, France
| | - Anselme L Perrier
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France
| | - Marc Peschanski
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France.,Université Evry Val d'Essonne (UEVE), Paris Saclay, U861, I-Stem, AFM, 91030, Evry Cedex, France
| | - Cecile Martinat
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France.,Université Evry Val d'Essonne (UEVE), Paris Saclay, U861, I-Stem, AFM, 91030, Evry Cedex, France
| | - Christine Baldeschi
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France.,Université Evry Val d'Essonne (UEVE), Paris Saclay, U861, I-Stem, AFM, 91030, Evry Cedex, France
| | - Gilles Lemaître
- INSERM U-861, Institut des cellules Souches pour le Traitement et l'Etude des Maladies monogéniques (I-Stem), Association Française contre les Myopathies (AFM), 91100, Corbeil Essonnes, France. .,Université Evry Val d'Essonne (UEVE), Paris Saclay, U861, I-Stem, AFM, 91030, Evry Cedex, France.
| |
Collapse
|
66
|
Yamauchi T, Yamasaki K, Tsuchiyama K, Koike S, Aiba S. The Potential of Muse Cells for Regenerative Medicine of Skin: Procedures to Reconstitute Skin with Muse Cell-Derived Keratinocytes, Fibroblasts, and Melanocytes. J Invest Dermatol 2017; 137:2639-2642. [DOI: 10.1016/j.jid.2017.06.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/26/2017] [Accepted: 06/27/2017] [Indexed: 11/30/2022]
|
67
|
New Biological Activities of Lythrum salicaria L.: Effects on Keratinocytes, Reconstructed Epidermis and Reconstructed Skins, Applications in Dermo-Cosmetic Sciences. COSMETICS 2017. [DOI: 10.3390/cosmetics4040052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
68
|
Larribère L, Galach M, Novak D, Arévalo K, Volz HC, Stark HJ, Boukamp P, Boutros M, Utikal J. An RNAi Screen Reveals an Essential Role for HIPK4 in Human Skin Epithelial Differentiation from iPSCs. Stem Cell Reports 2017; 9:1234-1245. [PMID: 28966120 PMCID: PMC5639458 DOI: 10.1016/j.stemcr.2017.08.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 12/31/2022] Open
Abstract
Molecular mechanisms responsible for the development of human skin epithelial cells are incompletely understood. As a consequence, the efficiency to establish a pure skin epithelial cell population from human induced pluripotent stem cells (hiPSCs) remains poor. Using an approach including RNAi and high-throughput imaging of early epithelial cells, we identified candidate kinases involved in their differentiation from hiPSCs. Among these, we found HIPK4 to be an important inhibitor of this process. Indeed, its silencing increased the amount of generated skin epithelial precursors at an early time point, increased the amount of generated keratinocytes at a later time point, and improved growth and differentiation of organotypic cultures, allowing for the formation of a denser basal layer and stratification with the expression of several keratins. Our data bring substantial input regarding regulation of human skin epithelial differentiation and for improving differentiation protocols from pluripotent stem cells. High-throughput RNAi screen setup during human skin epithelial differentiation Identification of HIPK4 as a crucial blocker of human skin epithelial differentiation Improvement of human organotypic epithelial cultures after HIPK4 silencing
Collapse
Affiliation(s)
- Lionel Larribère
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69121 Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany.
| | - Marta Galach
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69121 Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Daniel Novak
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69121 Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Karla Arévalo
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69121 Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Hans Christian Volz
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Department of Cell and Molecular Biology, Heidelberg University, 69120 Heidelberg, Germany; Department of Cardiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Hans-Jürgen Stark
- Genetics of Skin Carcinogenesis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Petra Boukamp
- Genetics of Skin Carcinogenesis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; IUF-Leibniz Research Institute for Environmental Medicine, 40021 Düsseldorf, Germany
| | - Michael Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Department of Cell and Molecular Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Jochen Utikal
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69121 Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany.
| |
Collapse
|
69
|
Desmet E, Ramadhas A, Lambert J, Van Gele M. In vitro psoriasis models with focus on reconstructed skin models as promising tools in psoriasis research. Exp Biol Med (Maywood) 2017; 242:1158-1169. [PMID: 28585891 DOI: 10.1177/1535370217710637] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Psoriasis is a complex chronic immune-mediated inflammatory cutaneous disease associated with the development of inflammatory plaques on the skin. Studies proved that the disease results from a deregulated interplay between skin keratinocytes, immune cells and the environment leading to a persisting inflammatory process modulated by pro-inflammatory cytokines and activation of T cells. However, a major hindrance to study the pathogenesis of psoriasis more in depth and subsequent development of novel therapies is the lack of suitable pre-clinical models mimicking the complex phenotype of this skin disorder. Recent advances in and optimization of three-dimensional skin equivalent models have made them attractive and promising alternatives to the simplistic monolayer cultures, immunological different in vivo models and scarce ex vivo skin explants. Moreover, human skin equivalents are increasing in complexity level to match human biology as closely as possible. Here, we critically review the different types of three-dimensional skin models of psoriasis with relevance to their application potential and advantages over other models. This will guide researchers in choosing the most suitable psoriasis skin model for therapeutic drug testing (including gene therapy via siRNA molecules), or to examine biological features contributing to the pathology of psoriasis. However, the addition of T cells (as recently applied to a de-epidermized dermis-based psoriatic skin model) or other immune cells would make them even more attractive models and broaden their application potential. Eventually, the ultimate goal would be to substitute animal models by three-dimensional psoriatic skin models in the pre-clinical phases of anti-psoriasis candidate drugs. Impact statement The continuous development of novel in vitro models mimicking the psoriasis phenotype is important in the field of psoriasis research, as currently no model exists that completely matches the in vivo psoriasis skin or the disease pathology. This work provides a complete overview of the different available in vitro psoriasis models and suggests improvements for future models. Moreover, a focus was given to psoriatic skin equivalent models, as they offer several advantages over the other models, including commercial availability and validity. The potential and reported applicability of these models in psoriasis pre-clinical research is extensively discussed. As such, this work offers a guide to researchers in their choice of pre-clinical psoriasis model depending on their type of research question.
Collapse
Affiliation(s)
- Eline Desmet
- Department of Dermatology, Ghent University Hospital, Ghent 9000, Belgium
| | - Anesh Ramadhas
- Department of Dermatology, Ghent University Hospital, Ghent 9000, Belgium
| | - Jo Lambert
- Department of Dermatology, Ghent University Hospital, Ghent 9000, Belgium
| | - Mireille Van Gele
- Department of Dermatology, Ghent University Hospital, Ghent 9000, Belgium
| |
Collapse
|
70
|
Sun Q, Deng XM, Wang YL, Zhen YF, Li F, Chen RH, Liang HS, Zhang F, Qin MD, Zhang XG. Serum is an indispensable factor in the maintenance of the biological characteristics of sweat gland cells. Mol Med Rep 2017; 16:2691-2699. [PMID: 28677764 PMCID: PMC5547951 DOI: 10.3892/mmr.2017.6909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 05/08/2017] [Indexed: 12/29/2022] Open
Abstract
The tolerance of sweat gland cells for in vitro amplification and subcultivation is low as they are somatic cells. The present study aimed to formulate an optimal medium for the culture of human eccrine sweat gland cells (HESGCs) and to establish a method for induction of HESGCs proliferation, whilst maintaining the characteristics of sweat gland cells. HESGCs cultured in sweat gland (SG):keratinocyte growth medium-2 (KGM-2) (1:1) medium had a higher proliferation rate and a stable morphology compared with cells cultured in SG and KGM-2 medium only. Reverse transcription-quantitative polymerase chain reaction indicated that cells cultured in the SG:KGM-2 (1:1) medium exhibited higher expression levels of α-smooth muscle actin, keratin (K)77, carcinoembryonic antigen, K8, K18, ectodysplasin A receptor, c-Myc, Kruppel-like factor 4 and octamer-binding transcription factor 4 compared with cells cultured in SG only or KGM-2 only medium. Three-dimensional culture analysis revealed that HESGCs cultured in SG:KGM-2 1:1 medium differentiated into sweat gland-like structures, whereas cells cultured in KGM-2 only medium underwent cornification. The present study also determined that the maintenance of the biological characteristics of HESGCs occurred due to the presence of fetal bovine serum (FBS). Cells cultured in medium without FBS differentiated into keratinocytes. Therefore, the SG:KGM-2 (1:1) medium may be a suitable culture medium for HESGCs. In conclusion, this mixed medium is a valuable compound and should be considered to be a potential supplemental medium for HESGCs.
Collapse
Affiliation(s)
- Qing Sun
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Xiao-Mei Deng
- The Stem Cell and Biomedical Material Key Laboratory of Jiangsu, Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Yun-Liang Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Yun-Fang Zhen
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215005, P.R. China
| | - Fang Li
- The Stem Cell and Biomedical Material Key Laboratory of Jiangsu, Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Rui-Hua Chen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Han-Si Liang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Feng Zhang
- The Stem Cell and Biomedical Material Key Laboratory of Jiangsu, Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Ming-De Qin
- The Stem Cell and Biomedical Material Key Laboratory of Jiangsu, Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Xue-Guang Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| |
Collapse
|
71
|
Pohin M, Veaute C, Garnier J, Barrault C, Cronier L, Huguier V, Favot L, Mcheik J, Bernard FX, Lecron JC, Morel F, Jégou JF. Development of a new model of reconstituted mouse epidermis and characterization of its response to proinflammatory cytokines. J Tissue Eng Regen Med 2017; 12:e1098-e1107. [PMID: 28477582 DOI: 10.1002/term.2442] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 04/07/2017] [Accepted: 05/03/2017] [Indexed: 12/30/2022]
Abstract
The development of three-dimensional models of reconstituted mouse epidermis (RME) has been hampered by the difficulty to maintain murine primary keratinocyte cultures and to achieve a complete epidermal stratification. In this study, a new protocol is proposed for the rapid and convenient generation of RME, which reproduces accurately the architecture of a normal mouse epidermis. During RME morphogenesis, the expression of differentiation markers such as keratins, loricrin, filaggrin, E-cadherin and connexins was followed, showing that RME structure at day 5 was similar to those of a normal mouse epidermis, with the acquisition of the natural barrier function. It was also demonstrated that RME responded to skin-relevant proinflammatory cytokines by increasing the expression of antimicrobial peptides and chemokines, and inhibiting epidermal differentiation markers, as in the human system. This new model of RME is therefore suitable to investigate mouse epidermis physiology further and opens new perspectives to generate reconstituted epidermis from transgenic mice.
Collapse
Affiliation(s)
- Mathilde Pohin
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines (LITEC), EA 4331, Université de Poitiers, France
| | - Carolina Veaute
- Laboratorio de Inmunología Básica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | | | | | - Laurent Cronier
- STIM, CNRS ERL 7368, Université de Poitiers, Poitiers, France
| | - Vincent Huguier
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines (LITEC), EA 4331, Université de Poitiers, France.,CHU de Poitiers, France
| | - Laure Favot
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines (LITEC), EA 4331, Université de Poitiers, France
| | - Jiad Mcheik
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines (LITEC), EA 4331, Université de Poitiers, France.,CHU de Poitiers, France
| | - François-Xavier Bernard
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines (LITEC), EA 4331, Université de Poitiers, France.,Bioalternatives, Gençay, France
| | - Jean-Claude Lecron
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines (LITEC), EA 4331, Université de Poitiers, France.,CHU de Poitiers, France
| | - Franck Morel
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines (LITEC), EA 4331, Université de Poitiers, France
| | - Jean-François Jégou
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines (LITEC), EA 4331, Université de Poitiers, France
| |
Collapse
|
72
|
Akhavan-Tavakoli M, Fard M, Khanjani S, Zare S, Edalatkhah H, Mehrabani D, Zarnani AH, Shirazi R, Kazemnejad S. In vitro differentiation of menstrual blood stem cells into keratinocytes: A potential approach for management of wound healing. Biologicals 2017; 48:66-73. [PMID: 28579353 DOI: 10.1016/j.biologicals.2017.05.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 05/10/2017] [Accepted: 05/22/2017] [Indexed: 01/30/2023] Open
Abstract
The skin wounds caused by insults should be treated immediately to restore the functions and integrity. Recent studies suggest that stem cells-based therapies may be applicable in wound healing. Newly defined menstrual blood-derived stem cells (MenSCs) show high rate of cell proliferation and trans-differentiation potency to various cell types. However, MenSCs potential to generate keratinocyte for future therapeutic use of skin lesions has been remained to investigate. We cultivated MenSCs in the presence of isolated foreskin derived-keratinocytes using an indirect co-culture system and evaluated efficiency of this protocol to generate keratinocytes using immunofluorescent staining and Real Time PCR technique. Our results showed that differentiated keratinocytes express epidermal/keratinocytes lineage specific markers such as K14, p63, and involucrin at both mRNA and protein levels. Immunofluorescent staining showed the expression of involucrin and K14 in differentiated cells in contrast to undifferentiated cells. Moreover, mRNA expression levels of K14 (11.1 folds, p = 0.001), p63 (10.23 folds, p = 0.001), and involucrin (2.94 folds, p = 0.001) were higher in differentiated MenSCs compared to non-cocultured cells. Therefore, we firstly presented evidence about differentiation capability of MenSCs into epidermal/keratinocytes lineage. Considering the advantages of MenSCs such as great accessibility, these stem cells are promising for stem cells-based therapies of skin defects.
Collapse
Affiliation(s)
- Maryam Akhavan-Tavakoli
- Department of Anatomical Sciences, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Maryam Fard
- Department of Anatomical Sciences, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Sayeh Khanjani
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
| | - Sona Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Haleh Edalatkhah
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
| | - Davood Mehrabani
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Science, Shiraz, Iran.
| | - Amir-Hassan Zarnani
- Nanobiotechnology Research Centre, Avicenna Research Institute, ACECR, Tehran, Iran.
| | - Reza Shirazi
- Department of Anatomical Sciences, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran; Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Somaieh Kazemnejad
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
| |
Collapse
|
73
|
Accelerating full thickness wound healing using collagen sponge of mrigal fish (Cirrhinus cirrhosus) scale origin. Int J Biol Macromol 2016; 93:1507-1518. [DOI: 10.1016/j.ijbiomac.2016.04.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/28/2016] [Accepted: 04/12/2016] [Indexed: 01/06/2023]
|
74
|
Ghosh D, Mehta N, Patil A, Sengupta J. Ethical issues in biomedical use of human embryonic stem cells (hESCs). ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.jrhm.2016.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
75
|
Rao Y, Cui J, Yin L, Liu W, Liu W, Sun M, Yan X, Wang L, Chen F. Preclinical study of mouse pluripotent parthenogenetic embryonic stem cell derivatives for the construction of tissue-engineered skin equivalent. Stem Cell Res Ther 2016; 7:156. [PMID: 27770834 PMCID: PMC5075200 DOI: 10.1186/s13287-016-0407-z] [Citation(s) in RCA: 5] [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: 04/28/2016] [Revised: 08/04/2016] [Accepted: 09/06/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Embryonic stem cell (ESC) derivatives hold great promise for the construction of tissue-engineered skin equivalents (TESE). However, harvesting of ESCs destroys viable embryos and may lead to political and ethical concerns over their application. In the current study, we directed mouse parthenogenetic embryonic stem cells (pESCs) to differentiate into fibroblasts, constructed TESE, and evaluated its function in vivo. METHODS The stemness marker expression and the pluripotent differentiation ability of pESCs were tested. After embryoid body (EB) formation and adherence culture, mesenchymal stem cells (MSCs) were enriched and directed to differentiate into fibroblastic lineage. Characteristics of derived fibroblasts were assessed by quantitative real-time PCR and ELISA. Functional ability of the constructed TESE was tested by a mouse skin defects repair model. RESULTS Mouse pESCs expressed stemness marker and could form teratoma containing three germ layers. MSCs could be enriched from outgrowths of EBs and directed to differentiate into fibroblastic lineage. These cells express a high level of growth factors including FGF, EGF, VEGF, TGF, PDGF, and IGF1, similar to those of ESC-derived fibroblasts and mouse fibroblasts. Seeded into collagen gels, the fibroblasts derived from pESCs could form TESE. Mouse skin defects could be successfully repaired 15 days after transplantation of TESE constructed by fibroblasts derived from pESCs. CONCLUSIONS pESCs could be induced to differentiate into fibroblastic lineage, which could be applied to the construction of TESE and skin defect repair. Particularly, pESC derivatives avoid the limitations of political and ethical concerns, and provide a promising source for regenerative medicine.
Collapse
Affiliation(s)
- Yang Rao
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Taibai North Rd 229, Xi’an, Shaanxi Province 710069 People’s Republic of China
| | - Jihong Cui
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Taibai North Rd 229, Xi’an, Shaanxi Province 710069 People’s Republic of China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Taibai North Rd 229, Xi’an, Shaanxi Province 710069 People’s Republic of China
| | - Lu Yin
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Taibai North Rd 229, Xi’an, Shaanxi Province 710069 People’s Republic of China
| | - Wei Liu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Taibai North Rd 229, Xi’an, Shaanxi Province 710069 People’s Republic of China
| | - Wenguang Liu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Taibai North Rd 229, Xi’an, Shaanxi Province 710069 People’s Republic of China
| | - Mei Sun
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Taibai North Rd 229, Xi’an, Shaanxi Province 710069 People’s Republic of China
| | - Xingrong Yan
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Taibai North Rd 229, Xi’an, Shaanxi Province 710069 People’s Republic of China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Taibai North Rd 229, Xi’an, Shaanxi Province 710069 People’s Republic of China
| | - Ling Wang
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Taibai North Rd 229, Xi’an, Shaanxi Province 710069 People’s Republic of China
| | - Fulin Chen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Taibai North Rd 229, Xi’an, Shaanxi Province 710069 People’s Republic of China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Taibai North Rd 229, Xi’an, Shaanxi Province 710069 People’s Republic of China
| |
Collapse
|
76
|
Mohammadi MH, Heidary Araghi B, Beydaghi V, Geraili A, Moradi F, Jafari P, Janmaleki M, Valente KP, Akbari M, Sanati-Nezhad A. Skin Diseases Modeling using Combined Tissue Engineering and Microfluidic Technologies. Adv Healthc Mater 2016; 5:2459-2480. [PMID: 27548388 DOI: 10.1002/adhm.201600439] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 05/30/2016] [Indexed: 12/19/2022]
Abstract
In recent years, both tissue engineering and microfluidics have significantly contributed in engineering of in vitro skin substitutes to test the penetration of chemicals or to replace damaged skins. Organ-on-chip platforms have been recently inspired by the integration of microfluidics and biomaterials in order to develop physiologically relevant disease models. However, the application of organ-on-chip on the development of skin disease models is still limited and needs to be further developed. The impact of tissue engineering, biomaterials and microfluidic platforms on the development of skin grafts and biomimetic in vitro skin models is reviewed. The integration of tissue engineering and microfluidics for the development of biomimetic skin-on-chip platforms is further discussed, not only to improve the performance of present skin models, but also for the development of novel skin disease platforms for drug screening processes.
Collapse
Affiliation(s)
- Mohammad Hossein Mohammadi
- Department of Chemical and Petroleum Engineering; Sharif University of Technology; Azadi Ave Tehran Iran
| | - Behnaz Heidary Araghi
- Department of Materials Science and Engineering; Sharif University of Technology; Azadi Ave Tehran Iran
| | - Vahid Beydaghi
- Department of Chemical and Petroleum Engineering; Sharif University of Technology; Azadi Ave Tehran Iran
| | - Armin Geraili
- Department of Chemical and Petroleum Engineering; Sharif University of Technology; Azadi Ave Tehran Iran
| | - Farshid Moradi
- Department of Chemical and Petroleum Engineering; Sharif University of Technology; Azadi Ave Tehran Iran
| | - Parya Jafari
- Department of Electrical Engineering; Sharif University of Technology; Azadi Ave Tehran Iran
| | - Mohsen Janmaleki
- Department of Mechanical and Manufacturing Engineering; Center for Bioengineering Research and Education; University of Calgary; 2500 University Drive NW Calgary AB Canada
| | - Karolina Papera Valente
- Department of Mechanical Engineering, and Center for Biomedical Research; University of Victoria; Victoria BC Canada
| | - Mohsen Akbari
- Department of Mechanical Engineering, and Center for Biomedical Research; University of Victoria; Victoria BC Canada
| | - Amir Sanati-Nezhad
- Department of Mechanical and Manufacturing Engineering; Center for Bioengineering Research and Education; University of Calgary; 2500 University Drive NW Calgary AB Canada
| |
Collapse
|
77
|
Qu Y, Zhou B, Yang W, Han B, Yu-Rice Y, Gao B, Johnson J, Svendsen CN, Freeman MR, Giuliano AE, Sareen D, Cui X. Transcriptome and proteome characterization of surface ectoderm cells differentiated from human iPSCs. Sci Rep 2016; 6:32007. [PMID: 27550649 PMCID: PMC4994084 DOI: 10.1038/srep32007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/01/2016] [Indexed: 12/11/2022] Open
Abstract
Surface ectoderm (SE) cells give rise to structures including the epidermis and ectodermal associated appendages such as hair, eye, and the mammary gland. In this study, we validate a protocol that utilizes BMP4 and the γ-secretase inhibitor DAPT to induce SE differentiation from human induced pluripotent stem cells (hiPSCs). hiPSC-differentiated SE cells expressed markers suggesting their commitment to the SE lineage. Computational analyses using integrated quantitative transcriptomic and proteomic profiling reveal that TGFβ superfamily signaling pathways are preferentially activated in SE cells compared with hiPSCs. SE differentiation can be enhanced by selectively blocking TGFβ-RI signaling. We also show that SE cells and neural ectoderm cells possess distinct gene expression patterns and signaling networks as indicated by functional Ingenuity Pathway Analysis. Our findings advance current understanding of early human SE cell development and pave the way for modeling of SE-derived tissue development, studying disease pathogenesis, and development of regenerative medicine approaches.
Collapse
Affiliation(s)
- Ying Qu
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, 90048, USA
| | - Bo Zhou
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, 90048, USA
| | - Wei Yang
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, 90048, USA
| | - Bingchen Han
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, 90048, USA
| | - Yi Yu-Rice
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, 90048, USA
| | - Bowen Gao
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, 90048, USA
| | - Jeffery Johnson
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, 90048, USA
| | - Clive N Svendsen
- Board of Governors-Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Michael R Freeman
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, 90048, USA
| | - Armando E Giuliano
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, 90048, USA
| | - Dhruv Sareen
- Board of Governors-Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,iPSC Core, The David and Janet Polak Foundation Stem Cell Core Laboratory, Los Angeles, CA, 90048, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Xiaojiang Cui
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, 90048, USA
| |
Collapse
|
78
|
Li H, Zhou H, Fu X, Xiao R. Directed differentiation of human embryonic stem cells into keratinocyte progenitors in vitro: an attempt with promise of clinical use. In Vitro Cell Dev Biol Anim 2016; 52:885-93. [DOI: 10.1007/s11626-016-0024-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 03/27/2016] [Indexed: 01/24/2023]
|
79
|
Liang H, Sun Q, Zhen Y, Li F, Xu Y, Liu Y, Zhang X, Qin M. The differentiation of amniotic fluid stem cells into sweat glandlike cells is enhanced by the presence of Sonic hedgehog in the conditioned medium. Exp Dermatol 2016; 25:714-20. [PMID: 27120089 DOI: 10.1111/exd.13062] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2016] [Indexed: 11/30/2022]
Abstract
After patients suffer severe full-thickness burn injuries, the current treatments cannot lead to the complete self-regeneration of the sweat gland structure and function. Therefore, it is important to identify new methods for acquiring sufficient functional sweat gland cells to restore skin function. In this study, we induced CD117+ human amniotic fluid stem (hAFS) cells to differentiate into sweat glandlike (hAFS-SG) cells based on the use of conditioned medium (CM) from the human sweat gland (hSG) cells. Real-time PCR and immunofluorescent staining were used to confirm the expression of the sweat gland-related genes Ectodysplasin-A (EDA), Ectodysplasin-A receptor (EDAR), keratin 8 (K8) and carcino-embryonic antigen (CEA). Transmission electron microscopy analysis shows that microvilli, the cellular structures that are typical for hSG cells, can also be observed on the membrane of the hAFS-SG cells. Our test for the calcium response to acetylcholine (Ach) proved that hAFS-SG cells have the potential to respond to Ach in a manner similar to normal sweat glands. A three-dimensional culture is an effective approach that stimulates the hAFS-SG cells to form tubular structures and drives hAFS-SG cells to mature into higher stage. We also found that epidermal growth factor enhances the efficiency of differentiation and that Sonic hedgehog is an important factor of the CM that influences sweat gland differentiation. Our study provides the basis for further investigations into novel methods of inducing stem cells to differentiate into sweat glandlike cells.
Collapse
Affiliation(s)
- Hansi Liang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.,The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (The State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, China
| | - Qing Sun
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.,The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (The State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, China
| | - Yunfang Zhen
- The Center of Diagnosis and Treatment for Children's Bone Diseases, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Fang Li
- The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (The State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, China.,Department of Human Anatomy, Histology and Embryology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, China
| | - YunYun Xu
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yao Liu
- The Center of Diagnosis and Treatment for Children's Bone Diseases, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.,The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (The State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, China
| | - Mingde Qin
- The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (The State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
80
|
García M, Quintana-Bustamante O, Segovia JC, Bueren J, Martinez-Santamaría L, Guerrero-Aspizua S, Escamez MJ, Del Rio M, Larcher F. Long-term skin regeneration in xenografts from iPSC teratoma-derived human keratinocytes. Exp Dermatol 2016; 25:736-8. [DOI: 10.1111/exd.13049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Marta García
- Epithelial Biomedicine Division; CIEMAT-CIBERER (U714); Madrid Spain
- Department of Bioengineering; Universidad Carlos III de Madrid; Leganés Madrid Spain
- Cátedra Fundación Jiménez Díaz (IIS-FJD) de Medicina Regenerativa y Bioingeniería Tisular; Madrid Spain
| | - Oscar Quintana-Bustamante
- Hematopoietic Innovative Therapies Division; CIEMAT-CIBERER (U710); Madrid Spain
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - José C. Segovia
- Hematopoietic Innovative Therapies Division; CIEMAT-CIBERER (U710); Madrid Spain
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Juan Bueren
- Hematopoietic Innovative Therapies Division; CIEMAT-CIBERER (U710); Madrid Spain
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Lucia Martinez-Santamaría
- Epithelial Biomedicine Division; CIEMAT-CIBERER (U714); Madrid Spain
- Department of Bioengineering; Universidad Carlos III de Madrid; Leganés Madrid Spain
- Cátedra Fundación Jiménez Díaz (IIS-FJD) de Medicina Regenerativa y Bioingeniería Tisular; Madrid Spain
| | - Sara Guerrero-Aspizua
- Epithelial Biomedicine Division; CIEMAT-CIBERER (U714); Madrid Spain
- Department of Bioengineering; Universidad Carlos III de Madrid; Leganés Madrid Spain
- Cátedra Fundación Jiménez Díaz (IIS-FJD) de Medicina Regenerativa y Bioingeniería Tisular; Madrid Spain
| | - María José Escamez
- Epithelial Biomedicine Division; CIEMAT-CIBERER (U714); Madrid Spain
- Department of Bioengineering; Universidad Carlos III de Madrid; Leganés Madrid Spain
- Cátedra Fundación Jiménez Díaz (IIS-FJD) de Medicina Regenerativa y Bioingeniería Tisular; Madrid Spain
| | - Marcela Del Rio
- Epithelial Biomedicine Division; CIEMAT-CIBERER (U714); Madrid Spain
- Department of Bioengineering; Universidad Carlos III de Madrid; Leganés Madrid Spain
- Cátedra Fundación Jiménez Díaz (IIS-FJD) de Medicina Regenerativa y Bioingeniería Tisular; Madrid Spain
| | - Fernando Larcher
- Epithelial Biomedicine Division; CIEMAT-CIBERER (U714); Madrid Spain
- Department of Bioengineering; Universidad Carlos III de Madrid; Leganés Madrid Spain
- Cátedra Fundación Jiménez Díaz (IIS-FJD) de Medicina Regenerativa y Bioingeniería Tisular; Madrid Spain
| |
Collapse
|
81
|
Huang Z, Zhen Y, Yin W, Ma Z, Zhang L. Shh promotes sweat gland cell maturation in three-dimensional culture. Cell Tissue Bank 2016; 17:317-25. [PMID: 26908022 PMCID: PMC4882370 DOI: 10.1007/s10561-016-9548-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 02/16/2016] [Indexed: 02/01/2023]
Abstract
Sweat gland (SG) cells forming SG tubule-like structures in 3D culture, this is one of the most important methods to identify the biological function of SG cells and stem cells-derived SG-like cells, but also the important way on research of SG regeneration in vitro. In this study, we seeded human fibroblasts and SG cells in gels and used immunohistochemistry to confirm whether SG tubule-like structures formed. Fibroblasts are necessary factor in the process of SG cells maturation and forming SG’s secretory region in 3D culture. Further experimentation revealed that Sonic hedgehog (Shh) was secreted by fibroblasts within the 3D culture. By adding Shh protein to 3D culture, there had more SG tubule-like structures formed. These results suggest that Shh is an important factor during the process of forming SG tubule-like structures in 3D cultures, and adding Shh recommbinant protein could promote SG cell maturation and enhance the efficiency of structure formation.
Collapse
Affiliation(s)
- Zhijian Huang
- Department of Burn and Plastic Surgery, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Yunfang Zhen
- The Center of Diagnosis and Treatment for Children's Bone Diseases, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Wei Yin
- Department of Burn and Plastic Surgery, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Zhourui Ma
- Department of Burn and Plastic Surgery, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Liya Zhang
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China.
| |
Collapse
|
82
|
Ensuring the Quality of Stem Cell-Derived In Vitro Models for Toxicity Testing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 856:259-297. [DOI: 10.1007/978-3-319-33826-2_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
83
|
Movahednia MM, Kidwai FK, Jokhun DS, Squier CA, Toh WS, Cao T. Potential applications of keratinocytes derived from human embryonic stem cells. Biotechnol J 2015; 11:58-70. [PMID: 26663861 DOI: 10.1002/biot.201500099] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/13/2015] [Accepted: 10/19/2015] [Indexed: 01/03/2023]
Abstract
Although skin grafting is one of the most advanced cell therapy technique, wide application of skin substitutes is hampered by the difficulty in securing sufficient amount of epidermal substitute. Additionally, in understanding the progression of skin aging and disease, and in screening the cosmetic and pharmaceutical products, there is lack of a satisfactory human skin-specific in vitro model. Recently, human embryonic stem cells (hESCs) have been proposed as an unlimited and reliable cell source to obtain almost all cell types present in the human body. This review focuses on the potential off-the-shelf use of hESC-derived keratinocytes for future clinical applications as well as a powerful in vitro skin model to study skin function and integrity, host-pathogen interactions and disease pathogenesis. Furthermore, we discuss the industrial applications of hESC-derived keratinized multi-layer epithelium which provides a human-like test platform for understanding disease pathogenesis, evaluation of new therapeutic modalities and assessment of the safety and efficacy of skin cosmetics and therapeutics. Overall, we conclude that the hESC-derived keratinocytes have great potential for clinical, research and industrial applications.
Collapse
Affiliation(s)
| | - Fahad K Kidwai
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Doorgesh S Jokhun
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Christopher A Squier
- Department of Oral Pathology, Radiology & Medicine, and Dows, College of Dentistry, The University of Iowa, Iowa City, Iowa, USA
| | - Wei Seong Toh
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore.,Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Tong Cao
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore. .,Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore. .,National University of Singapore Graduate School for Integrative Sciences and Engineering (NGS), Singapore, Singapore.
| |
Collapse
|
84
|
Yang R, Zheng Y, Burrows M, Liu S, Wei Z, Nace A, Guo W, Kumar S, Cotsarelis G, Xu X. Generation of folliculogenic human epithelial stem cells from induced pluripotent stem cells. Nat Commun 2015; 5:3071. [PMID: 24468981 PMCID: PMC4049184 DOI: 10.1038/ncomms4071] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 12/05/2013] [Indexed: 12/15/2022] Open
Abstract
Epithelial stem cells (EpSCs) in the hair follicle bulge are required for hair follicle growth and cycling. The isolation and propagation of human EpSCs for tissue engineering purposes remains a challenge. Here we develop a strategy to differentiate human iPSCs (hiPSCs) into CD200+/ITGA6+ EpSCs that can reconstitute the epithelial components of the hair follicle and interfollicular epidermis. The hiPSC-derived CD200+/ITGA6+ cells show a similar gene expression signature as EpSCs directly isolated from human hair follicles. Human iPSC-derived CD200+/ITGA6+ cells are capable of generating all hair follicle lineages including the hair shaft, and the inner and outer root sheaths in skin reconstitution assays. The regenerated hair follicles possess a KRT15+ stem cell population and produce hair shafts expressing hair specific keratins. These results suggest an approach for generating large numbers of human EpSCs for tissue engineering and new treatments for hair loss, wound healing and other degenerative skin disorders.
Collapse
Affiliation(s)
- Ruifeng Yang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Ying Zheng
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Michelle Burrows
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Shujing Liu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, New Jersey 07102, USA
| | - Arben Nace
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Suresh Kumar
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - George Cotsarelis
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
85
|
Wang H, Luo X, Leighton J. Extracellular Matrix and Integrins in Embryonic Stem Cell Differentiation. BIOCHEMISTRY INSIGHTS 2015; 8:15-21. [PMID: 26462244 PMCID: PMC4589090 DOI: 10.4137/bci.s30377] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/02/2015] [Accepted: 09/04/2015] [Indexed: 12/17/2022]
Abstract
Embryonic stem cells (ESCs) are pluripotent cells with great therapeutic potentials. The in vitro differentiation of ESC was designed by recapitulating embryogenesis. Significant progress has been made to improve the in vitro differentiation protocols by toning soluble maintenance factors. However, more robust methods for lineage-specific differentiation and maturation are still under development. Considering the complexity of in vivo embryogenesis environment, extracellular matrix (ECM) cues should be considered besides growth factor cues. ECM proteins bind to cells and act as ligands of integrin receptors on cell surfaces. Here, we summarize the role of the ECM and integrins in the formation of three germ layer progenies. Various ECM–integrin interactions were found, facilitating differentiation toward definitive endoderm, hepatocyte-like cells, pancreatic beta cells, early mesodermal progenitors, cardiomyocytes, neuroectoderm lineages, and epidermal cells, such as keratinocytes and melanocytes. In the future, ECM combinations for the optimal ESC differentiation environment will require substantial study.
Collapse
Affiliation(s)
- Han Wang
- Department of Cellular and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xie Luo
- Department of Cellular and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jake Leighton
- Department of Cellular and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
86
|
Fibroblast heterogeneity and its implications for engineering organotypic skin models in vitro. Eur J Cell Biol 2015; 94:483-512. [PMID: 26344860 DOI: 10.1016/j.ejcb.2015.08.001] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 08/11/2015] [Accepted: 08/11/2015] [Indexed: 12/19/2022] Open
Abstract
Advances in cell culture methods, multidisciplinary research, clinical need to replace lost skin tissues and regulatory need to replace animal models with alternative test methods has led to development of three dimensional models of human skin. In general, these in vitro models of skin consist of keratinocytes cultured over fibroblast-populated dermal matrices. Accumulating evidences indicate that mesenchyme-derived signals are essential for epidermal morphogenesis, homeostasis and differentiation. Various studies show that fibroblasts isolated from different tissues in the body are dynamic in nature and are morphologically and functionally heterogeneous subpopulations. Further, these differences seem to be dictated by the local biological and physical microenvironment the fibroblasts reside resulting in "positional identity or memory". Furthermore, the heterogeneity among the fibroblasts play a critical role in scarless wound healing and complete restoration of native tissue architecture in fetus and oral mucosa; and excessive scar formation in diseased states like keloids and hypertrophic scars. In this review, we summarize current concepts about the heterogeneity among fibroblasts and their role in various wound healing environments. Further, we contemplate how the insights on fibroblast heterogeneity could be applied for the development of next generation organotypic skin models.
Collapse
|
87
|
Sebastiano V, Zhen HH, Haddad B, Derafshi BH, Bashkirova E, Melo SP, Wang P, Leung TL, Siprashvili Z, Tichy A, Li J, Ameen M, Hawkins J, Lee S, Li L, Schwertschkow A, Bauer G, Lisowski L, Kay MA, Kim SK, Lane AT, Wernig M, Oro AE. Human COL7A1-corrected induced pluripotent stem cells for the treatment of recessive dystrophic epidermolysis bullosa. Sci Transl Med 2015; 6:264ra163. [PMID: 25429056 DOI: 10.1126/scitranslmed.3009540] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Patients with recessive dystrophic epidermolysis bullosa (RDEB) lack functional type VII collagen owing to mutations in the gene COL7A1 and suffer severe blistering and chronic wounds that ultimately lead to infection and development of lethal squamous cell carcinoma. The discovery of induced pluripotent stem cells (iPSCs) and the ability to edit the genome bring the possibility to provide definitive genetic therapy through corrected autologous tissues. We generated patient-derived COL7A1-corrected epithelial keratinocyte sheets for autologous grafting. We demonstrate the utility of sequential reprogramming and adenovirus-associated viral genome editing to generate corrected iPSC banks. iPSC-derived keratinocytes were produced with minimal heterogeneity, and these cells secreted wild-type type VII collagen, resulting in stratified epidermis in vitro in organotypic cultures and in vivo in mice. Sequencing of corrected cell lines before tissue formation revealed heterogeneity of cancer-predisposing mutations, allowing us to select COL7A1-corrected banks with minimal mutational burden for downstream epidermis production. Our results provide a clinical platform to use iPSCs in the treatment of debilitating genodermatoses, such as RDEB.
Collapse
Affiliation(s)
- Vittorio Sebastiano
- Institute for Stem Cell Biology and Regenerative Medicine, and Department of Pathology, Stanford University, Stanford, CA 94305, USA. Department of Obstetrics and Gynecology, Stanford University, Stanford, CA 94305, USA
| | - Hanson Hui Zhen
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | | | - Bahareh Haddad Derafshi
- Institute for Stem Cell Biology and Regenerative Medicine, and Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Elizaveta Bashkirova
- Institute for Stem Cell Biology and Regenerative Medicine, and Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Sandra P Melo
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Pei Wang
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Thomas L Leung
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Zurab Siprashvili
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Andrea Tichy
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Jiang Li
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Mohammed Ameen
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - John Hawkins
- Institute for Stem Cell Biology and Regenerative Medicine, and Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Susie Lee
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Lingjie Li
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Aaron Schwertschkow
- Institute for Regenerative Cures, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Gerhard Bauer
- Institute for Regenerative Cures, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Leszek Lisowski
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Mark A Kay
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Alfred T Lane
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, and Department of Pathology, Stanford University, Stanford, CA 94305, USA.
| | - Anthony E Oro
- Program in Epithelial Biology, Department of Dermatology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
88
|
Ali N, Hosseini M, Vainio S, Taïeb A, Cario‐André M, Rezvani H. Skin equivalents: skin from reconstructions as models to study skin development and diseases. Br J Dermatol 2015; 173:391-403. [DOI: 10.1111/bjd.13886] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2015] [Indexed: 12/17/2022]
Affiliation(s)
- N. Ali
- Laboratory of Developmental Biology Faculty of Biochemistry and Molecular Medicine University of Oulu and Biocenter Oulu Aapistie 5A 90220 Oulu Finland
- Inserm U 1035 33076 Bordeaux France
- Université de Bordeaux 146 rue Léo Saignat 33076 Bordeaux France
| | - M. Hosseini
- Inserm U 1035 33076 Bordeaux France
- Université de Bordeaux 146 rue Léo Saignat 33076 Bordeaux France
| | - S. Vainio
- Laboratory of Developmental Biology Faculty of Biochemistry and Molecular Medicine University of Oulu and Biocenter Oulu Aapistie 5A 90220 Oulu Finland
| | - A. Taïeb
- Inserm U 1035 33076 Bordeaux France
- Université de Bordeaux 146 rue Léo Saignat 33076 Bordeaux France
- Centre de Référence pour les Maladies Rares de la Peau Bordeaux France
- Département de Dermatologie & Dermatologie Pédiatrique CHU de Bordeaux Bordeaux France
| | - M. Cario‐André
- Inserm U 1035 33076 Bordeaux France
- Université de Bordeaux 146 rue Léo Saignat 33076 Bordeaux France
- Centre de Référence pour les Maladies Rares de la Peau Bordeaux France
| | - H.R. Rezvani
- Inserm U 1035 33076 Bordeaux France
- Université de Bordeaux 146 rue Léo Saignat 33076 Bordeaux France
- Centre de Référence pour les Maladies Rares de la Peau Bordeaux France
| |
Collapse
|
89
|
Sun Q, Li F, Li H, Chen RH, Gu YZ, Chen Y, Liang HS, You XR, Ding SS, Gao L, Wang YL, Qin MD, Zhang XG. Amniotic fluid stem cells provide considerable advantages in epidermal regeneration: B7H4 creates a moderate inflammation microenvironment to promote wound repair. Sci Rep 2015; 5:11560. [PMID: 26101181 PMCID: PMC4477371 DOI: 10.1038/srep11560] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 05/18/2015] [Indexed: 11/09/2022] Open
Abstract
The current treatments for severe skin injury all involve skin grafting. However, there is a worldwide shortage of donor skin tissue. In this study, we examined the advantages of using human amniotic fluid stem (hAFS) cells in skin wound healing. In vitro, hAFS cells differentiate into keratinocytes (termed hAFS-K). Like keratinocytes, hAFS-K cells express the markers K5, K14, K10 and involucrin; display typical cellular structure, including a tonofibril-rich cytoplasm; and construct a completely pluristratified epithelium in 3D culture. In vivo, in a mouse excisional wound model, GFP-positive hAFS cells participate in wound repair. Co-localization of GFP/K14 and GFP/K10 in the repaired epidermis demonstrated that hAFS cells can differentiate into keratinocytes. Real-time PCR results confirmed that hAFS cells can initiate and promote early-stage repair of skin damage. During wound repair, hAFS cells did not directly secrete repair-related factors, such as bFGF, VEGF, CXCL12, TGF-β1 and KGF, and provided a moderate inflammation reaction with lower expression of IL-1β, IL-6, TNF-α, Cox2 and Mac3. In hAFS cells, the negative co-stimulatory molecule B7H4 regulates low immunogenicity, which can provide a modest inflammatory reaction microenvironment for wound repair. Furthermore, with their uniquely high proliferation rate, hAFS cells offer a promising alternative for epidermal regeneration.
Collapse
Affiliation(s)
- Qing Sun
- 1] The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (the State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, P.R. China [2] Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Fang Li
- 1] The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (the State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, P.R. China [2] Department of Human Anatomy, Histology and Embryology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Hong Li
- Center for Reproduction and Genetics, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu Province,P.R. China
| | - Rui-Hua Chen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Yan-Zheng Gu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Ying Chen
- Center for Reproduction and Genetics, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu Province,P.R. China
| | - Han-Si Liang
- 1] The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (the State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, P.R. China [2] Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Xin-Ran You
- 1] The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (the State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, P.R. China [2] Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Si-Si Ding
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Ling Gao
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Yun-Liang Wang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Ming-De Qin
- 1] The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (the State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, P.R. China [2] Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Xue-Guang Zhang
- 1] The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (the State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, P.R. China [2] Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| |
Collapse
|
90
|
Larcher F, Del Río M. Innovative Therapeutic Strategies for Recessive Dystrophic Epidermolysis Bullosa. ACTAS DERMO-SIFILIOGRAFICAS 2015. [DOI: 10.1016/j.adengl.2015.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
91
|
Duscher D, Barrera J, Wong VW, Maan ZN, Whittam AJ, Januszyk M, Gurtner GC. Stem Cells in Wound Healing: The Future of Regenerative Medicine? A Mini-Review. Gerontology 2015; 62:216-25. [PMID: 26045256 DOI: 10.1159/000381877] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/25/2015] [Indexed: 01/13/2023] Open
Abstract
The increased risk of disease and decreased capacity to respond to tissue insult in the setting of aging results from complex changes in homeostatic mechanisms, including the regulation of oxidative stress and cellular heterogeneity. In aged skin, the healing capacity is markedly diminished resulting in a high risk for chronic wounds. Stem cell-based therapies have the potential to enhance cutaneous regeneration, largely through trophic and paracrine activity. Candidate cell populations for therapeutic application include adult mesenchymal stem cells, embryonic stem cells and induced pluripotent stem cells. Autologous cell-based approaches are ideal to minimize immune rejection but may be limited by the declining cellular function associated with aging. One strategy to overcome age-related impairments in various stem cell populations is to identify and enrich with functionally superior stem cell subsets via single cell transcriptomics. Another approach is to optimize cell delivery to the harsh environment of aged wounds via scaffold-based cell applications to enhance engraftment and paracrine activity of therapeutic stem cells. In this review, we shed light on challenges and recent advances surrounding stem cell therapies for wound healing and discuss limitations for their clinical adoption.
Collapse
Affiliation(s)
- Dominik Duscher
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, Calif., USA
| | | | | | | | | | | | | |
Collapse
|
92
|
Sridhar S, Venugopal JR, Ramakrishna S. Improved regeneration potential of fibroblasts using ascorbic acid-blended nanofibrous scaffolds. J Biomed Mater Res A 2015; 103:3431-40. [DOI: 10.1002/jbm.a.35486] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/22/2015] [Accepted: 04/14/2015] [Indexed: 01/09/2023]
Affiliation(s)
- Sreepathy Sridhar
- Center for Nanofibers and Nanotechnology, NUSNNI, Faculty of Engineering; National University of Singapore; Singapore
- Department of Genetic Engineering; SRM University, Kattankulathur; Chennai Tamilnadu India
| | - Jayarama Reddy Venugopal
- Center for Nanofibers and Nanotechnology, NUSNNI, Faculty of Engineering; National University of Singapore; Singapore
| | - Seeram Ramakrishna
- Center for Nanofibers and Nanotechnology, NUSNNI, Faculty of Engineering; National University of Singapore; Singapore
| |
Collapse
|
93
|
Has C, Kiritsi D. Therapies for inherited skin fragility disorders. Exp Dermatol 2015; 24:325-31. [DOI: 10.1111/exd.12666] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Cristina Has
- Department of Dermatology; Medical Center - University of Freiburg; Freiburg Germany
| | - Dimitra Kiritsi
- Department of Dermatology; Medical Center - University of Freiburg; Freiburg Germany
| |
Collapse
|
94
|
Nanba D, Toki F, Tate S, Imai M, Matsushita N, Shiraishi K, Sayama K, Toki H, Higashiyama S, Barrandon Y. Cell motion predicts human epidermal stemness. ACTA ACUST UNITED AC 2015; 209:305-15. [PMID: 25897083 PMCID: PMC4411274 DOI: 10.1083/jcb.201409024] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 03/17/2015] [Indexed: 12/22/2022]
Abstract
Keratinocyte stem cell colonies can be identified by analyzing cell motion, an emergent stem cell property. Image-based identification of cultured stem cells and noninvasive evaluation of their proliferative capacity advance cell therapy and stem cell research. Here we demonstrate that human keratinocyte stem cells can be identified in situ by analyzing cell motion during their cultivation. Modeling experiments suggested that the clonal type of cultured human clonogenic keratinocytes can be efficiently determined by analysis of early cell movement. Image analysis experiments demonstrated that keratinocyte stem cells indeed display a unique rotational movement that can be identified as early as the two-cell stage colony. We also demonstrate that α6 integrin is required for both rotational and collective cell motion. Our experiments provide, for the first time, strong evidence that cell motion and epidermal stemness are linked. We conclude that early identification of human keratinocyte stem cells by image analysis of cell movement is a valid parameter for quality control of cultured keratinocytes for transplantation.
Collapse
Affiliation(s)
- Daisuke Nanba
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center; Department of Biochemistry and Molecular Genetics and Department of Dermatology, Graduate School of Medicine; and Translational Research Center, Ehime University Hospital, Ehime University, Toon, Ehime 791-0295, Japan Division of Cell Growth and Tumor Regulation, Proteo-Science Center; Department of Biochemistry and Molecular Genetics and Department of Dermatology, Graduate School of Medicine; and Translational Research Center, Ehime University Hospital, Ehime University, Toon, Ehime 791-0295, Japan
| | - Fujio Toki
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center; Department of Biochemistry and Molecular Genetics and Department of Dermatology, Graduate School of Medicine; and Translational Research Center, Ehime University Hospital, Ehime University, Toon, Ehime 791-0295, Japan
| | - Sota Tate
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center; Department of Biochemistry and Molecular Genetics and Department of Dermatology, Graduate School of Medicine; and Translational Research Center, Ehime University Hospital, Ehime University, Toon, Ehime 791-0295, Japan
| | - Matome Imai
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center; Department of Biochemistry and Molecular Genetics and Department of Dermatology, Graduate School of Medicine; and Translational Research Center, Ehime University Hospital, Ehime University, Toon, Ehime 791-0295, Japan
| | - Natsuki Matsushita
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center; Department of Biochemistry and Molecular Genetics and Department of Dermatology, Graduate School of Medicine; and Translational Research Center, Ehime University Hospital, Ehime University, Toon, Ehime 791-0295, Japan
| | - Ken Shiraishi
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center; Department of Biochemistry and Molecular Genetics and Department of Dermatology, Graduate School of Medicine; and Translational Research Center, Ehime University Hospital, Ehime University, Toon, Ehime 791-0295, Japan
| | - Koji Sayama
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center; Department of Biochemistry and Molecular Genetics and Department of Dermatology, Graduate School of Medicine; and Translational Research Center, Ehime University Hospital, Ehime University, Toon, Ehime 791-0295, Japan
| | - Hiroshi Toki
- Research Center for Nuclear Physics, Osaka University, Ibaraki, Osaka 567-0047, Japan
| | - Shigeki Higashiyama
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center; Department of Biochemistry and Molecular Genetics and Department of Dermatology, Graduate School of Medicine; and Translational Research Center, Ehime University Hospital, Ehime University, Toon, Ehime 791-0295, Japan Division of Cell Growth and Tumor Regulation, Proteo-Science Center; Department of Biochemistry and Molecular Genetics and Department of Dermatology, Graduate School of Medicine; and Translational Research Center, Ehime University Hospital, Ehime University, Toon, Ehime 791-0295, Japan
| | - Yann Barrandon
- Laboratory of Stem Cell Dynamics, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland Department of Experimental Surgery, Centre Hospitalier Universitaire Vaudois, CH-1011 Lausanne, Switzerland
| |
Collapse
|
95
|
Tadeu AMB, Lin S, Hou L, Chung L, Zhong M, Zhao H, Horsley V. Transcriptional profiling of ectoderm specification to keratinocyte fate in human embryonic stem cells. PLoS One 2015; 10:e0122493. [PMID: 25849374 PMCID: PMC4388500 DOI: 10.1371/journal.pone.0122493] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 02/22/2015] [Indexed: 11/18/2022] Open
Abstract
In recent years, several studies have shed light into the processes that regulate epidermal specification and homeostasis. We previously showed that a broad-spectrum γ-secretase inhibitor DAPT promoted early keratinocyte specification in human embryonic stem cells triggered to undergo ectoderm specification. Here, we show that DAPT accelerates human embryonic stem cell differentiation and induces expression of the ectoderm protein AP2. Furthermore, we utilize RNA sequencing to identify several candidate regulators of ectoderm specification including those involved in epithelial and epidermal development in human embryonic stem cells. Genes associated with transcriptional regulation and growth factor activity are significantly enriched upon DAPT treatment during specification of human embryonic stem cells to the ectoderm lineage. The human ectoderm cell signature identified in this study contains several genes expressed in ectodermal and epithelial tissues. Importantly, these genes are also associated with skin disorders and ectodermal defects, providing a platform for understanding the biology of human epidermal keratinocyte development under diseased and homeostatic conditions.
Collapse
Affiliation(s)
- Ana Mafalda Baptista Tadeu
- Yale University, Department of Molecular, Cell and Developmental Biology, New Haven, CT, 06511, United States of America
| | - Samantha Lin
- Yale University, Department of Molecular, Cell and Developmental Biology, New Haven, CT, 06511, United States of America
| | - Lin Hou
- Yale University, Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06520, United States of America
| | - Lisa Chung
- Yale University, Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06520, United States of America
| | - Mei Zhong
- Yale University, Yale Stem Cell Center, Genomics Facility, New Haven, CT, 06520, United States of America
| | - Hongyu Zhao
- Yale University, Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06520, United States of America
| | - Valerie Horsley
- Yale University, Department of Molecular, Cell and Developmental Biology, New Haven, CT, 06511, United States of America
- * E-mail:
| |
Collapse
|
96
|
Movahednia MM, Kidwai FK, Zou Y, Tong HJ, Liu X, Islam I, Toh WS, Raghunath M, Cao T. Differential Effects of the Extracellular Microenvironment on Human Embryonic Stem Cell Differentiation into Keratinocytes and Their Subsequent Replicative Life Span. Tissue Eng Part A 2015; 21:1432-43. [DOI: 10.1089/ten.tea.2014.0551] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Fahad Karim Kidwai
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Yu Zou
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Huei Jinn Tong
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Xiaochen Liu
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
- School and Hospital of Stomatology, Zhejiang University, Hangzhou, People's Republic of China
| | - Intekhab Islam
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Wei Seong Toh
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
- National University of Singapore Tissue Engineering Program (NUSTEP), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Michael Raghunath
- National University of Singapore Tissue Engineering Program (NUSTEP), Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tong Cao
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
- National University of Singapore Tissue Engineering Program (NUSTEP), Life Sciences Institute, National University of Singapore, Singapore, Singapore
- National University of Singapore Graduate School for Integrative Sciences and Engineering (NGS), Singapore, Singapore
| |
Collapse
|
97
|
Larcher F, Del Río M. Innovative therapeutic strategies for recessive dystrophic epidermolysis bullosa. ACTAS DERMO-SIFILIOGRAFICAS 2015; 106:376-82. [PMID: 25796272 DOI: 10.1016/j.ad.2015.01.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/12/2015] [Indexed: 02/07/2023] Open
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) is among the most serious rare skin diseases. It is also the rare skin disease for which most effort has been expended in developing advanced therapeutic interventions. RDEB is caused by collagen VII deficiency resulting from COL7A1 mutations. Therapeutic approaches seek to replenish collagen VII and thus restore dermal-epidermal adhesion. Therapeutic options under development include protein therapy and different cell-based and gene-based therapies. In addition to treating skin defects, some of these therapies may also target internal mucosa. In the coming years, these novel therapeutic approaches should substantially improve the quality of life of patients with RDEB.
Collapse
Affiliation(s)
- F Larcher
- División de Biomedicina Epitelial, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Madrid, España; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Madrid, España; Instituto de Investigaciones Sanitarias de la Fundación Jimenez Díaz (IIS-FJD), Madrid, España.
| | - M Del Río
- División de Biomedicina Epitelial, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Madrid, España; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Madrid, España; Instituto de Investigaciones Sanitarias de la Fundación Jimenez Díaz (IIS-FJD), Madrid, España; Departamento de Bioingeniería, Universidad Carlos III de Madrid (UC3M), Madrid, España
| |
Collapse
|
98
|
Chen D, Hao H, Tong C, Liu J, Dong L, Ti D, Hou Q, Liu H, Han W, Fu X. Transdifferentiation of Umbilical Cord–Derived Mesenchymal Stem Cells Into Epidermal-Like Cells by the Mimicking Skin Microenvironment. INT J LOW EXTR WOUND 2015; 14:136-45. [PMID: 25700709 DOI: 10.1177/1534734615569913] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human umbilical cord–derived mesenchymal stem cells (UC-MSCs) are multipotent, primitive, and have been widely used for skin tissue engineering. Their transdifferentiation is determined by the local microenvironment. In this study, we investigated the potential epidermal differentiation of UC-MSCs and the formation of epidermis substitutes in a 3-dimensional (3D) microenvironment, which was fabricated by UC-MSCs embedded into collagen–chitosan scaffolds (CCSs) combined with an air–liquid interface (ALI) culture system. Using fluorescence microscope, we observed that UC-MSCs were spindle-shaped and evenly distributed in the scaffold. Methyl thiazolyl blue tetrazolium bromide assay and Live/Dead assay indicated that the CCSs have good biocompatibility with UC-MSCs. Immunohistochemistry and western blotting assay showed that UC-MSCs on the surface of the CCSs were positive for the epidermal markers cytokeratin 19 and involucrin at 14 days. In addition, hematoxylin–eosin staining indicated that multilayered epidermis substitutes were established. The constructed epidermis substitutes were applied to treat full-thickness wounds in rats and proved to promote wound healing. In conclusion, manipulating the 3D microenvironment is a novel method for inducing the epidermal differentiation of MSCs to engineer epidermal substitutes, which provides an alternative strategy for skin tissue engineering.
Collapse
Affiliation(s)
- Deyun Chen
- Chinese PLA General Hospital, Beijing, China
| | - Haojie Hao
- Chinese PLA General Hospital, Beijing, China
| | - Chuan Tong
- Chinese PLA General Hospital, Beijing, China
| | - Jiejie Liu
- Chinese PLA General Hospital, Beijing, China
| | - Liang Dong
- Chinese PLA General Hospital, Beijing, China
| | - Dongdong Ti
- Chinese PLA General Hospital, Beijing, China
| | - Qian Hou
- Chinese PLA General Hospital, Beijing, China
| | - Huiling Liu
- Chinese PLA General Hospital, Beijing, China
| | - Weidong Han
- Chinese PLA General Hospital, Beijing, China
| | - Xiaobing Fu
- Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
99
|
Shimamoto A, Yokote K, Tahara H. Werner Syndrome-specific induced pluripotent stem cells: recovery of telomere function by reprogramming. Front Genet 2015; 6:10. [PMID: 25688260 PMCID: PMC4310323 DOI: 10.3389/fgene.2015.00010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/10/2015] [Indexed: 01/10/2023] Open
Abstract
Werner syndrome (WS) is a rare human autosomal recessive premature aging disorder characterized by early onset of aging-associated diseases, chromosomal instability, and cancer predisposition. The function of the DNA helicase encoded by WRN, the gene responsible for WS, has been studied extensively. WRN helicase is involved in the maintenance of chromosome integrity through DNA replication, repair, and recombination by interacting with a variety of proteins associated with DNA repair and telomere maintenance. The accelerated aging associated with WS is reportedly caused by telomere dysfunction, and the underlying mechanism of the disease is yet to be elucidated. Although it was reported that the life expectancy for patients with WS has improved over the last two decades, definitive therapy for these patients has not seen much development. Severe symptoms of the disease, such as leg ulcers, cause a significant decline in the quality of life in patients with WS. Therefore, the establishment of new therapeutic strategies for the disease is of utmost importance. Induced pluripotent stem cells (iPSCs) can be established by the introduction of several pluripotency genes, including Oct3/4, Sox2, Klf4, and c-myc into differentiated cells. iPSCs have the potential to differentiate into a variety of cell types that constitute the human body, and possess infinite proliferative capacity. Recent studies have reported the generation of iPSCs from the cells of patients with WS, and they have concluded that reprogramming represses premature senescence phenotypes in these cells. In this review, we summarize the findings of WS patient-specific iPSCs (WS iPSCs) and focus on the roles of telomere and telomerase in the maintenance of these cells. Finally, we discuss the potential use of WS iPSCs for clinical applications.
Collapse
Affiliation(s)
- Akira Shimamoto
- Department of Cellular and Molecular Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hiroshima, Japan
| | - Koutaro Yokote
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University Chiba, Japan
| | - Hidetoshi Tahara
- Department of Cellular and Molecular Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hiroshima, Japan
| |
Collapse
|
100
|
Couteaudier M, Trapp-Fragnet L, Auger N, Courvoisier K, Pain B, Denesvre C, Vautherot JF. Derivation of keratinocytes from chicken embryonic stem cells: establishment and characterization of differentiated proliferative cell populations. Stem Cell Res 2015; 14:224-37. [PMID: 25702531 DOI: 10.1016/j.scr.2015.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 12/16/2014] [Accepted: 01/12/2015] [Indexed: 11/18/2022] Open
Abstract
A common challenge in avian cell biology is the generation of differentiated cell-lines, especially in the keratinocyte lineage. Only a few avian cell-lines are available and very few of them show an interesting differentiation profile. During the last decade, mammalian embryonic stem cell-lines were shown to differentiate into almost all lineages, including keratinocytes. Although chicken embryonic stem cells had been obtained in the 1990s, few differentiation studies toward the ectodermal lineage were reported. Consequently, we explored the differentiation of chicken embryonic stem cells toward the keratinocyte lineage by using a combination of stromal induction, ascorbic acid, BMP4 and chicken serum. During the induction period, we observed a downregulation of pluripotency markers and an upregulation of epidermal markers. Three homogenous cell populations were derived, which were morphologically similar to chicken primary keratinocytes, displaying intracellular lipid droplets in almost every pavimentous cell. These cells could be serially passaged without alteration of their morphology and showed gene and protein expression profiles of epidermal markers similar to chicken primary keratinocytes. These cells represent an alternative to the isolation of chicken primary keratinocytes, being less cumbersome to handle and reducing the number of experimental animals used for the preparation of primary cells.
Collapse
Affiliation(s)
- Mathilde Couteaudier
- INRA, UMR 1282, Infectious Diseases and Public Health, ISP, Biova Team, Centre INRA de Tours, F-37380 Nouzilly, France.
| | - Laëtitia Trapp-Fragnet
- INRA, UMR 1282, Infectious Diseases and Public Health, ISP, Biova Team, Centre INRA de Tours, F-37380 Nouzilly, France.
| | - Nicolas Auger
- INRA, UMR 1282, Infectious Diseases and Public Health, ISP, Biova Team, Centre INRA de Tours, F-37380 Nouzilly, France
| | - Katia Courvoisier
- INRA, UMR 1282, Infectious Diseases and Public Health, ISP, Biova Team, Centre INRA de Tours, F-37380 Nouzilly, France
| | - Bertrand Pain
- INRA, USC 1361, INSERM U846, Université Lyon 1, U846 S, Institut Cellules Souches et Cerveau, F-69500 Bron, France.
| | - Caroline Denesvre
- INRA, UMR 1282, Infectious Diseases and Public Health, ISP, Biova Team, Centre INRA de Tours, F-37380 Nouzilly, France.
| | - Jean-François Vautherot
- INRA, UMR 1282, Infectious Diseases and Public Health, ISP, Biova Team, Centre INRA de Tours, F-37380 Nouzilly, France.
| |
Collapse
|