1
|
Choudhary V, Choudhary M, Bollag WB. Exploring Skin Wound Healing Models and the Impact of Natural Lipids on the Healing Process. Int J Mol Sci 2024; 25:3790. [PMID: 38612601 PMCID: PMC11011291 DOI: 10.3390/ijms25073790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/14/2024] Open
Abstract
Cutaneous wound healing is a complex biological process involving a series of well-coordinated events aimed at restoring skin integrity and function. Various experimental models have been developed to study the mechanisms underlying skin wound repair and to evaluate potential therapeutic interventions. This review explores the diverse array of skin wound healing models utilized in research, ranging from rodent excisional wounds to advanced tissue engineering constructs and microfluidic platforms. More importantly, the influence of lipids on the wound healing process is examined, emphasizing their role in enhancing barrier function restoration, modulating inflammation, promoting cell proliferation, and promoting remodeling. Lipids, such as phospholipids, sphingolipids, and ceramides, play crucial roles in membrane structure, cell signaling, and tissue repair. Understanding the interplay between lipids and the wound microenvironment provides valuable insights into the development of novel therapeutic strategies for promoting efficient wound healing and tissue regeneration. This review highlights the significance of investigating skin wound healing models and elucidating the intricate involvement of lipids in the healing process, offering potential avenues for improving clinical outcomes in wound management.
Collapse
Affiliation(s)
- Vivek Choudhary
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (V.C.)
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Mrunal Choudhary
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (V.C.)
| | - Wendy B. Bollag
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (V.C.)
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
- Department of Dermatology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
2
|
Cheng W, Fan C, Song Q, Chen P, Peng H, Lin L, Liu C, Wang B, Zhou Z. Induced pluripotent stem cell-based therapies for organ fibrosis. Front Bioeng Biotechnol 2023; 11:1119606. [PMID: 37274156 PMCID: PMC10232908 DOI: 10.3389/fbioe.2023.1119606] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/09/2023] [Indexed: 06/06/2023] Open
Abstract
Fibrotic diseases result in organ remodelling and dysfunctional failure and account for one-third of all deaths worldwide. There are no ideal treatments that can halt or reverse progressive organ fibrosis, moreover, organ transplantation is complicated by problems with a limited supply of donor organs and graft rejection. The development of new approaches, especially induced pluripotent stem cell (iPSC)-based therapy, is becoming a hot topic due to their ability to self-renew and differentiate into different cell types that may replace the fibrotic organs. In the past decade, studies have differentiated iPSCs into fibrosis-relevant cell types which were demonstrated to have anti-fibrotic effects that may have the potential to inform new effective precision treatments for organ-specific fibrosis. In this review, we summarize the potential of iPSC-based cellular approaches as therapeutic avenues for treating organ fibrosis, the advantages and disadvantages of iPSCs compared with other types of stem cell-based therapies, as well as the challenges and future outlook in this field.
Collapse
Affiliation(s)
- Wei Cheng
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Qing Song
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| | - Ping Chen
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| | - Hong Peng
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| | - Ling Lin
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| | - Cong Liu
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| | - Bin Wang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Zijing Zhou
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| |
Collapse
|
3
|
Oh JH, Cho S, Choi JA. Clinical Signs of Kawasaki Disease from the Perspective of Epithelial-to-Mesenchymal Transition Recruiting Erythrocytes: A Literature Review. Rev Cardiovasc Med 2023; 24:109. [PMID: 39076265 PMCID: PMC11273048 DOI: 10.31083/j.rcm2404109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/05/2023] [Accepted: 03/14/2023] [Indexed: 07/31/2024] Open
Abstract
Kawasaki disease (KD) is a systemic vasculitis affecting children younger than 5 years of age. Early period in life is marked by rapid somatic growth with cell proliferation and immaturity of the immunity with dominant innate immune system. Coronary complications in KD are the most common acquired heart disease in children, yet the diagnosis of KD still depends on the clinical diagnostic criteria. Glossy red lips and conjunctival injection are characteristic signs enabling pediatricians to make the initial diagnosis of KD; however, little is known why these are so characteristic. The diagnostic criteria of KD seem to be scattered in seemingly irrelevant body systems such as the eyes, lips, skin, and heart. KD is classified as a connective tissue disease. Recently, red blood cells (RBCs) have emerged as important modulators in innate immune response. RBCs are reported to participate in extracellular matrix remodeling and upregulating matrix metalloproteinase (MMP) expression in dermal fibroblasts. Also, fibroblast growth factors and microRNAs associated with fibrosis are drawing attention in KD. The cardinal signs of KD appear at the border of muco-cutaneous junction. Head and neck regions are abundant in tissues undergoing epithelial-to-mesenchymal transition (EMT). Interstitial carditis and valve insufficiency as well as coronary arterial lesions may complicate KD, and these lesions present in tissues that originated from epicardial progenitor cells by EMT. Having reviewed the recent research on KD, we presume that the signs of KD present at borders between keratinized and non-keratinized stratified squamous epithelium where the EMT is still ongoing for the rapid somatic growth where RBCs are recruited as an innate immune response and to prevent excessive fibrosis in mucosa. KD presents scarcely in adults with somatic growth and immune maturation completed. In this review, we attempted to explain the reasons for the clinical manifestations of KD and to search for a link among the diagnostic clues in the perspective of EMT during the somatic growth and immune system maturation in children with KD.
Collapse
Affiliation(s)
- Jin-Hee Oh
- Department of Pediatrics, St.Vincent's Hospital, College of Medicine, The Catholic University of Korea, 16247 Seoul, Republic of Korea
| | - Soyun Cho
- Department of Dermatology, Boramae Medical Center, College of Medicine, Seoul National University, 07061 Seoul, Republic of Korea
| | - Jin A Choi
- Department of Ophthalmology & Laboratory of Visual Science, St.Vincent’s Hospital, College of Medicine, The Catholic University of Korea, 16247 Seoul, Republic of Korea
| |
Collapse
|
4
|
Santarella F, do Amaral RJFC, Lemoine M, Kelly D, Cavanagh B, Marinkovic M, Smith A, Garlick J, O'Brien FJ, Kearney CJ. Personalized Scaffolds for Diabetic Foot Ulcer Healing Using Extracellular Matrix from Induced Pluripotent Stem-Reprogrammed Patient Cells. ADVANCED NANOBIOMED RESEARCH 2022; 2:2200052. [PMID: 36532145 PMCID: PMC9757804 DOI: 10.1002/anbr.202200052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023] Open
Abstract
Diabetic foot ulcers (DFU) are chronic wounds sustained by pathological fibroblasts and aberrant extracellular matrix (ECM). Porous collagen-based scaffolds (CS) have shown clinical promise for treating DFUs but may benefit from functional enhancements. Our previous work showed fibroblasts differentiated from induced pluripotent stem cells are an effective source of new ECM mimicking fetal matrix, which notably promotes scar-free healing. Likewise, functionalizing CS with this rejuvenated ECM showed potential for DFU healing. Here, we demonstrate for the first time an approach to DFU healing using biopsied cells from DFU patients, reprogramming those cells, and functionalizing CS with patient-specific ECM as a personalized acellular tissue engineered scaffold. We took a two-pronged approach: 1) direct ECM blending into scaffold fabrication; and 2) seeding scaffolds with reprogrammed fibroblasts for ECM deposition followed by decellularization. The decellularization approach reduced cell number requirements and maintained naturally deposited ECM proteins. Both approaches showed enhanced ECM deposition from DFU fibroblasts. Decellularized scaffolds additionally enhanced glycosaminoglycan deposition and subsequent vascularization. Finally, reprogrammed ECM scaffolds from patient-matched DFU fibroblasts outperformed those from healthy fibroblasts in several metrics, suggesting ECM is in fact able to redirect resident pathological fibroblasts in DFUs towards healing, and a patient-specific ECM signature may be beneficial.
Collapse
Affiliation(s)
- Francesco Santarella
- 123 Stephens Green, Kearney Lab/Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Ronaldo Jose Farias Correa do Amaral
- 123 Stephens Green, Kearney Lab/Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Laboratório de Proliferação e Diferenciação Celular, Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil
| | - Mark Lemoine
- 123 Stephens Green, Kearney Lab/Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Domhnall Kelly
- 123 Stephens Green, Kearney Lab/Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Brenton Cavanagh
- 123 Stephens Green, Kearney Lab/Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Milica Marinkovic
- 123 Stephens Green, Kearney Lab/Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Avi Smith
- Department of Diagnostic Sciences, Tufts University School of Dental Medicine, Boston, MA 02111 USA
| | - Jonathan Garlick
- Department of Diagnostic Sciences, Tufts University School of Dental Medicine, Boston, MA 02111 USA
| | - Fergal J O'Brien
- 123 Stephens Green, Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland and Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Cathal J Kearney
- Department of Biomedical Engineering, University of Massachusetts Amherst, USA
- 123 Stephens Green, Kearney Lab/Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland and Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| |
Collapse
|
5
|
Álvarez-Vásquez JL, Castañeda-Alvarado CP. Dental pulp fibroblast: A star Cell. J Endod 2022; 48:1005-1019. [DOI: 10.1016/j.joen.2022.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 12/16/2022]
|
6
|
Clinical Grade Human Pluripotent Stem Cell-Derived Engineered Skin Substitutes Promote Keratinocytes Wound Closure In Vitro. Cells 2022; 11:cells11071151. [PMID: 35406716 PMCID: PMC8998132 DOI: 10.3390/cells11071151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic wounds, such as leg ulcers associated with sickle cell disease, occur as a consequence of a prolonged inflammatory phase during the healing process. They are extremely hard to heal and persist as a significant health care problem due to the absence of effective treatment and the uprising number of patients. Indeed, there is a critical need to develop novel cell- and tissue-based therapies to treat these chronic wounds. Development in skin engineering leads to a small catalogue of available substitutes manufactured in Good Manufacturing Practices compliant (GMPc) conditions. Those substitutes are produced using primary cells that could limit their use due to restricted sourcing. Here, we propose GMPc protocols to produce functional populations of keratinocytes and fibroblasts derived from pluripotent stem cells to reconstruct the associated dermo-epidermal substitute with plasma-based fibrin matrix. In addition, this manufactured composite skin is biologically active and enhances in vitro wounding of keratinocytes. The proposed composite skin opens new perspectives for skin replacement using allogeneic substitute.
Collapse
|
7
|
|
8
|
Glycocalyx disruption enhances motility, proliferation and collagen synthesis in diabetic fibroblasts. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:118955. [PMID: 33421533 DOI: 10.1016/j.bbamcr.2021.118955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 12/24/2020] [Accepted: 01/04/2021] [Indexed: 11/22/2022]
Abstract
Impaired wound healing represents one of the most debilitating side effects of Diabetes mellitus. Though the role of fibroblasts in wound healing is well-known, the extent to which their function is altered in the context of diabetes remains incompletely understood. Here, we address this question by comparing the phenotypes of healthy dermal fibroblasts (HDFs) and diabetic dermal fibroblasts (DDFs). We show that DDFs are more elongated but less motile and less contractile than HDFs. Reduced motility of DDFs is attributed to formation of larger focal adhesions stabilized by a bulky glycocalyx, associated with increased expression of the cell surface glycoprotein mucin 16 (MUC 16). Disruption of the glycocalyx not only restored DDF motility to levels comparable to that of HDFs, but also led to increased proliferation and collagen synthesis. Collectively, our results illustrate the influence of glycocalyx disruption on mechanics of diabetic fibroblasts relevant to cell motility.
Collapse
|
9
|
Przekora A. A Concise Review on Tissue Engineered Artificial Skin Grafts for Chronic Wound Treatment: Can We Reconstruct Functional Skin Tissue In Vitro? Cells 2020; 9:cells9071622. [PMID: 32640572 PMCID: PMC7407512 DOI: 10.3390/cells9071622] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic wounds occur as a consequence of a prolonged inflammatory phase during the healing process, which precludes skin regeneration. Typical treatment for chronic wounds includes application of autografts, allografts collected from cadaver, and topical delivery of antioxidant, anti-inflammatory, and antibacterial agents. Nevertheless, the mentioned therapies are not sufficient for extensive or deep wounds. Moreover, application of allogeneic skin grafts carries high risk of rejection and treatment failure. Advanced therapies for chronic wounds involve application of bioengineered artificial skin substitutes to overcome graft rejection as well as topical delivery of mesenchymal stem cells to reduce inflammation and accelerate the healing process. This review focuses on the concept of skin tissue engineering, which is a modern approach to chronic wound treatment. The aim of the article is to summarize common therapies for chronic wounds and recent achievements in the development of bioengineered artificial skin constructs, including analysis of biomaterials and cells widely used for skin graft production. This review also presents attempts to reconstruct nerves, pigmentation, and skin appendages (hair follicles, sweat glands) using artificial skin grafts as well as recent trends in the engineering of biomaterials, aiming to produce nanocomposite skin substitutes (nanofilled polymer composites) with controlled antibacterial activity. Finally, the article describes the composition, advantages, and limitations of both newly developed and commercially available bioengineered skin substitutes.
Collapse
Affiliation(s)
- Agata Przekora
- Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland
| |
Collapse
|
10
|
Smith A, Huang M, Watkins T, Burguin F, Baskin J, Garlick JA. De novo production of human extracellular matrix supports increased throughput and cellular complexity in 3D skin equivalent model. J Tissue Eng Regen Med 2020; 14:1019-1027. [PMID: 32483913 DOI: 10.1002/term.3071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 04/14/2020] [Accepted: 05/11/2020] [Indexed: 12/17/2022]
Abstract
Three-dimensional (3D) tissue models of human skin are being developed to better understand disease phenotypes and to screen new drugs for potential therapies. Several factors will increase the value of these in vitro 3D skin tissues for these purposes. These include the need for human-derived extracellular matrix (ECM), higher throughput tissue formats, and greater cellular complexity. Here, we present an approach for the fabrication of 3D skin-like tissues as a platform that addresses these three considerations. We demonstrate that human adult and neonatal fibroblasts deposit an endogenous ECM de novo that serves as an effective stroma for full epithelial tissue development and differentiation. We have miniaturized these tissues to a 24-well format to adapt them for eventual higher throughput drug screening. We have shown that monocytes from the peripheral blood can be incorporated into this model as macrophages to increase tissue complexity. This humanized skin-like tissue decreases dependency on animal-derived ECM while increasing cellular complexity that can enable screening inflammatory responses in tissue models of human skin.
Collapse
Affiliation(s)
- Avi Smith
- Department of Diagnostic Science, Tufts University School of Dental Medicine, Boston, MA, USA
| | - Mengqi Huang
- Department of Diagnostic Science, Tufts University School of Dental Medicine, Boston, MA, USA.,Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Trishawna Watkins
- Department of Diagnostic Science, Tufts University School of Dental Medicine, Boston, MA, USA
| | - Fiona Burguin
- Department of Diagnostic Science, Tufts University School of Dental Medicine, Boston, MA, USA
| | - Jeremy Baskin
- Department of Diagnostic Science, Tufts University School of Dental Medicine, Boston, MA, USA
| | - Jonathan A Garlick
- Department of Diagnostic Science, Tufts University School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
11
|
Schreier S, Triampo W. The Blood Circulating Rare Cell Population. What is it and What is it Good For? Cells 2020; 9:cells9040790. [PMID: 32218149 PMCID: PMC7226460 DOI: 10.3390/cells9040790] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023] Open
Abstract
Blood contains a diverse cell population of low concentration hematopoietic as well as non-hematopoietic cells. The majority of such rare cells may be bone marrow-derived progenitor and stem cells. This paucity of circulating rare cells, in particular in the peripheral circulation, has led many to believe that bone marrow as well as other organ-related cell egress into the circulation is a response to pathological conditions. Little is known about this, though an increasing body of literature can be found suggesting commonness of certain rare cell types in the peripheral blood under physiological conditions. Thus, the isolation and detection of circulating rare cells appears to be merely a technological problem. Knowledge about rare cell types that may circulate the blood stream will help to advance the field of cell-based liquid biopsy by supporting inter-platform comparability, making use of biological correct cutoffs and “mining” new biomarkers and combinations thereof in clinical diagnosis and therapy. Therefore, this review intends to lay ground for a comprehensive analysis of the peripheral blood rare cell population given the necessity to target a broader range of cell types for improved biomarker performance in cell-based liquid biopsy.
Collapse
Affiliation(s)
- Stefan Schreier
- School of Bioinnovation and Bio-based Product Intelligence, Faculty of Science, Mahidol University, Rama VI Rd, Bangkok 10400, Thailand;
- Thailand Center of Excellence in Physics, Ministry of Higher Education, Science, Research and Innovation, 328 Si Ayutthaya Road, Bangkok 10400, Thailand
| | - Wannapong Triampo
- Thailand Center of Excellence in Physics, Ministry of Higher Education, Science, Research and Innovation, 328 Si Ayutthaya Road, Bangkok 10400, Thailand
- Department of Physics, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Correspondence:
| |
Collapse
|
12
|
Esteban-Vives R, Ziembicki J, Sun Choi M, Thompson RL, Schmelzer E, Gerlach JC. Isolation and Characterization of a Human Fetal Mesenchymal Stem Cell Population: Exploring the Potential for Cell Banking in Wound Healing Therapies. Cell Transplant 2019; 28:1404-1419. [PMID: 31407589 PMCID: PMC6802149 DOI: 10.1177/0963689718817524] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Various cell-based therapies are in development to address chronic and acute skin wound
healing, for example for burns and trauma patients. An off-the-shelf source of allogeneic
dermal cells could be beneficial for innovative therapies accelerating the healing in
extensive wounds where the availability of a patient’s own cells is limited. Human
fetal-derived dermal fibroblasts (hFDFs) show high in vitro division rates, exhibit low
immunological rejection properties, and present scarless wound healing in the fetus, and
previous studies on human fetal tissue-derived cell therapies have shown promising results
on tissue repair. However, little is known about cell lineage stability and cell
differentiation during the cell expansion process, required for any potential therapeutic
use. We describe an isolation method, characterize a population, and investigate its
potential for cell banking and thus suitability as a potential product for cell grafting
therapies. Our results show hFDFs and a bone marrow-derived mesenchymal stem cell (BM-MSC)
line shared identification markers and in vitro multilineage differentiation potential
into osteogenic, chondrogenic, and adipogenic lineages. The hFDF population exhibited
similar cell characteristics as BM-MSCs while producing lower pro-inflammatory cytokine
IL-6 levels and higher levels of the wound healing factor hepatocyte growth factor. We
demonstrate in vitro differentiation of hFDFs, which may be a problem in maintaining
long-term lineage stability, potentially limiting their use for cell banking and therapy
development.
Collapse
Affiliation(s)
- Roger Esteban-Vives
- Departments of Surgery and Bioengineering, McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA
| | - Jenny Ziembicki
- The University of Pittsburgh Medical Center, UPMC Mercy Hospital Trauma and Burn Centers, Pittsburgh, PA, USA
| | | | - R L Thompson
- Allegheny Reproductive Health Center, Pittsburgh, PA, USA
| | - Eva Schmelzer
- Departments of Surgery and Bioengineering, McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA
| | - Jörg C Gerlach
- Departments of Surgery and Bioengineering, McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA
| |
Collapse
|
13
|
Kaur A, Midha S, Giri S, Mohanty S. Functional Skin Grafts: Where Biomaterials Meet Stem Cells. Stem Cells Int 2019; 2019:1286054. [PMID: 31354835 PMCID: PMC6636521 DOI: 10.1155/2019/1286054] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/21/2019] [Indexed: 12/22/2022] Open
Abstract
Skin tissue engineering has attained several clinical milestones making remarkable progress over the past decades. Skin is inhabited by a plethora of cells spatiotemporally arranged in a 3-dimensional (3D) matrix, creating a complex microenvironment of cell-matrix interactions. This complexity makes it difficult to mimic the native skin structure using conventional tissue engineering approaches. With the advent of newer fabrication strategies, the field is evolving rapidly. However, there is still a long way before an artificial skin substitute can fully mimic the functions and anatomical hierarchy of native human skin. The current focus of skin tissue engineers is primarily to develop a 3D construct that maintains the functionality of cultured cells in a guided manner over a period of time. While several natural and synthetic biopolymers have been translated, only partial clinical success is attained so far. Key challenges include the hierarchical complexity of skin anatomy; compositional mismatch in terms of material properties (stiffness, roughness, wettability) and degradation rate; biological complications like varied cell numbers, cell types, matrix gradients in each layer, varied immune responses, and varied methods of fabrication. In addition, with newer biomaterials being adopted for fabricating patient-specific skin substitutes, issues related to escalating processing costs, scalability, and stability of the constructs under in vivo conditions have raised some concerns. This review provides an overview of the field of skin regenerative medicine, existing clinical therapies, and limitations of the current techniques. We have further elaborated on the upcoming tissue engineering strategies that may serve as promising alternatives for generating functional skin substitutes, the pros and cons associated with each technique, and scope of their translational potential in the treatment of chronic skin ailments.
Collapse
Affiliation(s)
- Amtoj Kaur
- Stem Cell Facility (DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| | - Swati Midha
- Stem Cell Facility (DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| | - Shibashish Giri
- Department of Cell Techniques and Applied Stem Cell Biology, Centre for Biotechnology and Biomedicine, University of Leipzig, Deutscher Platz 5, D-04103 Leipzig, Germany
- Department of Plastic Surgery and Hand Surgery, University Hospital Rechts der Isar, Technische Universität München, Munich, Germany
| | - Sujata Mohanty
- Stem Cell Facility (DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
14
|
Rousselle P, Braye F, Dayan G. Re-epithelialization of adult skin wounds: Cellular mechanisms and therapeutic strategies. Adv Drug Deliv Rev 2019; 146:344-365. [PMID: 29981800 DOI: 10.1016/j.addr.2018.06.019] [Citation(s) in RCA: 270] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/28/2018] [Accepted: 06/25/2018] [Indexed: 12/21/2022]
Abstract
Cutaneous wound healing in adult mammals is a complex multi-step process involving overlapping stages of blood clot formation, inflammation, re-epithelialization, granulation tissue formation, neovascularization, and remodelling. Re-epithelialization describes the resurfacing of a wound with new epithelium. The cellular and molecular processes involved in the initiation, maintenance, and completion of epithelialization are essential for successful wound closure. A variety of modulators are involved, including growth factors, cytokines, matrix metalloproteinases, cellular receptors, and extracellular matrix components. Here, we focus on cellular mechanisms underlying keratinocyte migration and proliferation during epidermal closure. Inability to re-epithelialize is a clear indicator of chronic non-healing wounds, which fail to proceed through the normal phases of wound healing in an orderly and timely manner. This review summarizes the current knowledge regarding the management and treatment of acute and chronic wounds, with a focus on re-epithelialization, offering some insights into novel future therapies.
Collapse
|
15
|
Lysyl oxidase enzymes mediate TGF-β1-induced fibrotic phenotypes in human skin-like tissues. J Transl Med 2019; 99:514-527. [PMID: 30568176 DOI: 10.1038/s41374-018-0159-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/17/2018] [Accepted: 10/29/2018] [Indexed: 01/09/2023] Open
Abstract
Cutaneous fibrosis is a common complication seen in mixed connective tissue diseases. It often occurs as a result of TGF-β-induced deposition of excessive amounts of collagen in the skin. Lysyl oxidases (LOXs), a family of extracellular matrix (ECM)-modifying enzymes responsible for collagen cross-linking, are known to be increased in dermal fibroblasts from patients with fibrotic diseases, denoting a possible role of LOXs in fibrosis. To directly study this, we have developed two bioengineered, in vitro skin-like models: human skin equivalents (hSEs), and self-assembled stromal tissues (SASs) that contain either normal or systemic sclerosis (SSc; scleroderma) patient-derived fibroblasts. These tissues provide an organ-level structure that could be combined with non-invasive, label-free, multiphoton microscopy (SHG/TPEF) to reveal alterations in the organization and cross-linking levels of collagen fibers during the development of cutaneous fibrosis, which demonstrated increased stromal rigidity and activation of dermal fibroblasts in response to TGF-β1. Specifically, inhibition of specific LOXs isoforms, LOX and LOXL4, in foreskin fibroblasts (HFFs) resulted in antagonistic effects on TGF-β1-induced fibrogenic hallmarks in both hSEs and SASs. In addition, a translational relevance of these models was seen as similar antifibrogenic phenotypes were achieved upon knocking down LOXL4 in tissues containing SSc patient-derived-dermal fibroblasts (SScDFs). These findings point to a pivotal role of LOXs in TGF-β1-induced cutaneous fibrosis through impaired ECM homeostasis in skin-like tissues, and show the value of these tissue platforms in accelerating the discovery of antifibrosis therapeutics.
Collapse
|
16
|
Insights into autophagy machinery in cells related to skin diseases and strategies for therapeutic modulation. Biomed Pharmacother 2019; 113:108775. [PMID: 30889485 DOI: 10.1016/j.biopha.2019.108775] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 03/11/2019] [Accepted: 03/13/2019] [Indexed: 02/05/2023] Open
Abstract
Autophagy, literally meaning "self-eating," is a highly conserved process that is part of the eukaryotic cell cycle. Morphologically, the double membrane contains vesicles with phagocytic components known as autophagosomes. Autophagy is often used as a cellular stress response and quality control mechanisms are used to maintain cell survival. Survival is facilitated by providing energy and metabolic precursors as well as removing damaged proteins or organelles. Moreover, autophagy refers to organelles fused together with part of the cell cytoplasm with a double or multi-membrane structure called phagosome. Research has demonstrated that autophagy is an important mediator of cell fate and has effects on inflammation, pathogen clearance, and antigen presentation. In recent years, studies discussing autophagy have increased in number. Nevertheless, only a small amount of research has considered the impact of autophagy on the pathogenesis of skin diseases. The skin is the largest organ of the body, with a surface area of around two square metre; it is the first line of defense against numerous environmental insults, including ultraviolet radiation, pathogens, mechanical stresses, and toxic chemicals. Autophagy is thought to be a vital modality for endogenous defenses against environmental derangements. This review provides an overview of autophagy machinery in keratinocytes, skin fibroblasts, melanocytes related to skin diseases as well as strategies for therapeutic modulation, for the future development of treatment for skin diseases.
Collapse
|
17
|
Peng J, Liu R, Peng L, Jia H. Calcium gluconate alleviates the toxic effect of hydrofluoric acid on human dermal fibroblasts through the Wnt/β-catenin pathway. Oncol Lett 2018; 16:2921-2928. [PMID: 30127880 PMCID: PMC6096138 DOI: 10.3892/ol.2018.8975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/15/2018] [Indexed: 12/22/2022] Open
Abstract
The present study was performed to determine the molecular mechanism of calcium gluconate (CG) in alleviating the toxic effect of hydrofluoric (HF) acid on human dermal fibroblasts (HDFs). HDF morphology was observed by optical microscopy and the vimentin immunofluorescence assay. Cell viability and apoptosis were evaluated by the Cell Counting Kit-8 and Annexin V/propidium iodide assays, respectively. The levels of apoptosis-associated factors, as well as Wnt2, Wnt3a and β-catenin were detected by reverse transcription-quantitative polymerase chain reaction and western blotting. Levels of matrix metalloproteinase (MMP)-1 and basic fibroblast growth factor (bFGF) were detected by ELISA and western blotting. Carboxyterminal propeptide of type I collagen (CICP) was detected by ELISA, while L-Hydroxyproline (L-HYP) was detected by colorimetry. First, the morphology of normal HDFs was observed. Cell viability was inhibited and apoptosis was increased in a dose- and time-dependent manner following treatment with HF acid [0, 2, 4, 6, 8, 10 and 20% (v/v)] for 0, 2, 4, 6, 8, 10 and 20 min. The effects were blocked by CG at different doses (50, 100 and 200 µmol/l) and time points (6, 12 and 24 h), following treatment with 8% (v/v) HF acid for 6 min. The levels of Caspase-3, B-cell lymphoma (Bcl)-2 associated X protein, Wnt2, Wnt3a and β-catenin were decreased, whereas Bcl-2 was increased by CG treatment dose-dependently, when compared with HF control. CG promoted the expression of MMP-1, bFGF and L-HYP, and inhibited CICP, when compared with HF control. Based on the present results, CG alleviated the toxic effect of HF acid on HDFs by regulating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Jinxia Peng
- Department of Burn and Plastic Surgery, Shiyan People's Hospital, People's Hospital Affiliated to Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Rui Liu
- Department of Oncology, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Ling Peng
- Department of Health Management, People's Hospital of Shiyan Economic Development Zone, Shiyan, Hubei 442000, P.R. China
| | - Hongtao Jia
- Department of Urological Surgery, Shiyan People's Hospital, People's Hospital Affiliated to Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
18
|
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
|
19
|
Stem Cells in Dentistry: Types of Intra- and Extraoral Tissue-Derived Stem Cells and Clinical Applications. Stem Cells Int 2018; 2018:4313610. [PMID: 30057624 PMCID: PMC6051054 DOI: 10.1155/2018/4313610] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/05/2018] [Accepted: 06/07/2018] [Indexed: 12/13/2022] Open
Abstract
Stem cells are undifferentiated cells, capable of renewing themselves, with the capacity to produce different cell types to regenerate missing tissues and treat diseases. Oral facial tissues have been identified as a source and therapeutic target for stem cells with clinical interest in dentistry. This narrative review report targets on the several extraoral- and intraoral-derived stem cells that can be applied in dentistry. In addition, stem cell origins are suggested in what concerns their ability to differentiate as well as their particular distinguishing quality of convenience and immunomodulatory for regenerative dentistry. The development of bioengineered teeth to replace the patient's missing teeth was also possible because of stem cell technologies. This review will also focus our attention on the clinical application of stem cells in dentistry. In recent years, a variety of articles reported the advantages of stem cell-based procedures in regenerative treatments. The regeneration of lost oral tissue is the target of stem cell research. Owing to the fact that bone imperfections that ensue after tooth loss can result in further bone loss which limit the success of dental implants and prosthodontic therapies, the rehabilitation of alveolar ridge height is prosthodontists' principal interest. The development of bioengineered teeth to replace the patient's missing teeth was also possible because of stem cell technologies. In addition, a “dental stem cell banking” is available for regenerative treatments in the future. The main features of stem cells in the future of dentistry should be understood by clinicians.
Collapse
|
20
|
Soundararajan M, Kannan S. Fibroblasts and mesenchymal stem cells: Two sides of the same coin? J Cell Physiol 2018; 233:9099-9109. [DOI: 10.1002/jcp.26860] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022]
Affiliation(s)
| | - Suresh Kannan
- Department of Biomedical Sciences Sri Ramachandra University Chennai Tamil Nadu India
| |
Collapse
|
21
|
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
|
22
|
Dixit S, Baganizi DR, Sahu R, Dosunmu E, Chaudhari A, Vig K, Pillai SR, Singh SR, Dennis VA. Immunological challenges associated with artificial skin grafts: available solutions and stem cells in future design of synthetic skin. J Biol Eng 2017; 11:49. [PMID: 29255480 PMCID: PMC5729423 DOI: 10.1186/s13036-017-0089-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 11/17/2017] [Indexed: 12/29/2022] Open
Abstract
The repair or replacement of damaged skins is still an important, challenging public health problem. Immune acceptance and long-term survival of skin grafts represent the major problem to overcome in grafting given that in most situations autografts cannot be used. The emergence of artificial skin substitutes provides alternative treatment with the capacity to reduce the dependency on the increasing demand of cadaver skin grafts. Over the years, considerable research efforts have focused on strategies for skin repair or permanent skin graft transplantations. Available skin substitutes include pre- or post-transplantation treatments of donor cells, stem cell-based therapies, and skin equivalents composed of bio-engineered acellular or cellular skin substitutes. However, skin substitutes are still prone to immunological rejection, and as such, there is currently no skin substitute available to overcome this phenomenon. This review focuses on the mechanisms of skin rejection and tolerance induction and outlines in detail current available strategies and alternatives that may allow achieving full-thickness skin replacement and repair.
Collapse
Affiliation(s)
- Saurabh Dixit
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA.,Immunity, Inflammation, and Disease Laboratory, NIH/NIEHS, Durham, 27709 NC USA
| | - Dieudonné R Baganizi
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Rajnish Sahu
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Ejowke Dosunmu
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Atul Chaudhari
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Komal Vig
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Shreekumar R Pillai
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Shree R Singh
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Vida A Dennis
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| |
Collapse
|
23
|
Higgins CA, Roger MF, Hill RP, Ali-Khan AS, Garlick JA, Christiano AM, Jahoda CAB. Multifaceted role of hair follicle dermal cells in bioengineered skins. Br J Dermatol 2017; 176:1259-1269. [PMID: 27679975 DOI: 10.1111/bjd.15087] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND The method of generating bioengineered skin constructs was pioneered several decades ago; nowadays these constructs are used regularly for the treatment of severe burns and nonhealing wounds. Commonly, these constructs are comprised of skin fibroblasts within a collagen scaffold, forming the skin dermis, and stratified keratinocytes overlying this, forming the skin epidermis. In the past decade there has been a surge of interest in bioengineered skins, with researchers seeking alternative cell sources, or scaffolds, from which constructs can be established, and for more biomimetic equivalents with skin appendages. OBJECTIVES To evaluate whether human hair follicle dermal cells can act as an alternative cell source for engineering the dermal component of engineered skin constructs. METHODS We established in vitro skin constructs by incorporating into the collagenous dermal compartment: (i) primary interfollicular dermal fibroblasts, (ii) hair follicle dermal papilla cells or (iii) hair follicle dermal sheath cells. In vivo skins were established by mixing dermal cells and keratinocytes in chambers on top of immunologically compromised mice. RESULTS All fibroblast subtypes were capable of supporting growth of overlying epithelial cells, both in vitro and in vivo. However, we found hair follicle dermal sheath cells to be superior to fibroblasts in their capacity to influence the establishment of a basal lamina. CONCLUSIONS Human hair follicle dermal cells can be readily interchanged with interfollicular fibroblasts and used as an alternative cell source for establishing the dermal component of engineered skin both in vitro and in vivo.
Collapse
Affiliation(s)
- C A Higgins
- Department of Dermatology, Columbia University, New York, NY, U.S.A.,Department of Bioengineering, Imperial College London, London, U.K
| | - M F Roger
- School of Biological and Biomedical Sciences, Durham University, Durham, U.K
| | - R P Hill
- School of Biological and Biomedical Sciences, Durham University, Durham, U.K
| | - A S Ali-Khan
- Department of Plastic Surgery, University Hospital of Durham, Durham, U.K
| | - J A Garlick
- Sackler Graduate School of Biomedical Sciences, Tufts University, Boston, MA, U.S.A
| | - A M Christiano
- Department of Dermatology, Columbia University, New York, NY, U.S.A.,Department of Genetics and Development, Columbia University, New York, NY, U.S.A
| | - C A B Jahoda
- School of Biological and Biomedical Sciences, Durham University, Durham, U.K
| |
Collapse
|
24
|
Pantazi E, Gemenetzidis E, Teh MT, Reddy SV, Warnes G, Evagora C, Trigiante G, Philpott MP. GLI2 Is a Regulator of β-Catenin and Is Associated with Loss of E-Cadherin, Cell Invasiveness, and Long-Term Epidermal Regeneration. J Invest Dermatol 2017; 137:1719-1730. [PMID: 28300597 DOI: 10.1016/j.jid.2016.11.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 10/31/2016] [Accepted: 11/26/2016] [Indexed: 12/31/2022]
Abstract
Uncontrolled hedgehog (HH)/glioma-associated oncogene (GLI) and WNT/β-catenin signaling are important events in the genesis of many cancers including skin cancer and are often implicated in tumor progression, invasion, and metastasis. However, because of the complexity and context dependency of both pathways, little is known about HH and WNT interactions in human carcinogenesis. In the current study, we provide evidence of HH/glioma-associated oncogene family zinc finger 2 (GLI2)-WNT/β-catenin signaling crosstalk in human keratinocytes. Overexpression of GLI2ΔN in human keratinocytes resulted in cytoplasmic accumulation and nuclear relocalization of β-catenin in vitro and in 3D organotypic cultures, accompanied by upregulation of WNT genes. Induction of GLI2ΔN enhanced the β-catenin-dependent transcriptional activation and the subsequent activation of β-catenin target genes including cyclin-D1. Additionally, GLI2 overexpression was associated with decreased E-cadherin protein levels; increased expression of SNAIL, matrix metalloproteinase 2, and integrin β1; and increased cell invasion in 3D organotypic cultures. Invasion was reduced by WNT inhibition, thus unveiling the direct role of GLI2/WNT crosstalk in cell invasion. We show that GLI2 overexpression supported long-term epidermal regeneration in 3D organotypic cultures, and resulted in the manifestation of an undifferentiated basal/stem cell-associated phenotype in human keratinocytes. Both these observations are consistent with the role of β-catenin and SNAIL in epidermal stem cell maintenance. This work suggests that GLI2 is a regulator of β-catenin and provides insights into its role in tumorigenesis.
Collapse
Affiliation(s)
- Eleni Pantazi
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Emilios Gemenetzidis
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Muy-Teck Teh
- Department of Diagnostic and Oral Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sreekanth Vootukuri Reddy
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gary Warnes
- Imaging and Flow Cytometry Core facilities, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Chris Evagora
- Pathology Core facilities, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Giuseppe Trigiante
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael P Philpott
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
25
|
Dermal-epidermal membrane systems by using human keratinocytes and mesenchymal stem cells isolated from dermis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 71:943-953. [PMID: 27987793 DOI: 10.1016/j.msec.2016.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/20/2016] [Accepted: 11/02/2016] [Indexed: 02/04/2023]
Abstract
Dermal-epidermal membrane systems were developed by co-culturing human keratinocytes with Skin derived Stem Cells (SSCs), which are Mesenchymal Stem Cells (MSCs) isolated from dermis, on biodegradable membranes of chitosan (CHT), polycaprolactone (PCL) and a polymeric blend of CHT and PCL. The membranes display physico-chemical, morphological, mechanical and biodegradation properties that could satisfy and fulfil specific requirements in skin tissue engineering. CHT membrane exhibits an optimal biodegradation rate for acute wounds; CHT-PCL for the chronic ones. On the other hand, PCL membrane in spite of its very slow biodegradation rate exhibits mechanical properties similar to in vivo dermis, a lower hydrophilic character, and a surface roughness, all properties that make it able to sustain cell adhesion and proliferation for in vitro skin models. Both CHT-PCL and PCL membranes guided epidermal and dermal differentiation of SSCs as pointed out by the expression of cytokeratins and the deposition of the ECM protein fibronectin, respectively. In the dermal-epidermal membrane systems, a more suitable microenvironment for the SSCs differentiation was promoted by the interactions and the mutual interplay with keratinocytes. Being skin tissue-biased stem cells committed to their specific final dermal and/or epidermal cell differentiation, SSCs are more suitable for skin tissue engineering than other adult MSCs with different origin. For this reason, they represent a useful autologous cell source for engineering skin substitutes for both in vivo and in vitro applications.
Collapse
|
26
|
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
|
27
|
Woodby B, Scott M, Bodily J. The Interaction Between Human Papillomaviruses and the Stromal Microenvironment. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:169-238. [PMID: 27865458 PMCID: PMC5727914 DOI: 10.1016/bs.pmbts.2016.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human papillomaviruses (HPVs) are small, double-stranded DNA viruses that replicate in stratified squamous epithelia and cause a variety of malignancies. Current efforts in HPV biology are focused on understanding the virus-host interactions that enable HPV to persist for years or decades in the tissue. The importance of interactions between tumor cells and the stromal microenvironment has become increasingly apparent in recent years, but how stromal interactions impact the normal, benign life cycle of HPVs, or progression of lesions to cancer is less understood. Furthermore, how productively replicating HPV impacts cells in the stromal environment is also unclear. Here we bring together some of the relevant literature on keratinocyte-stromal interactions and their impacts on HPV biology, focusing on stromal fibroblasts, immune cells, and endothelial cells. We discuss how HPV oncogenes in infected cells manipulate other cells in their environment, and, conversely, how neighboring cells may impact the efficiency or course of HPV infection.
Collapse
Affiliation(s)
- B Woodby
- Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - M Scott
- Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - J Bodily
- Louisiana State University Health Sciences Center, Shreveport, LA, United States.
| |
Collapse
|
28
|
Autologous adipose-derived adult stem cells injection versus platelet-rich plasma injection in the treatment of rolling postacne scars. JOURNAL OF THE EGYPTIAN WOMEN’S DERMATOLOGIC SOCIETY 2016. [DOI: 10.1097/01.ewx.0000489880.96422.b1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
29
|
Gerami-Naini B, Smith A, Maione AG, Kashpur O, Carpinito G, Veves A, Mooney DJ, Garlick JA. Generation of Induced Pluripotent Stem Cells from Diabetic Foot Ulcer Fibroblasts Using a Nonintegrative Sendai Virus. Cell Reprogram 2016; 18:214-23. [PMID: 27328415 DOI: 10.1089/cell.2015.0087] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Diabetic foot ulcers (DFUs) are nonhealing chronic wounds that are a serious complication of diabetes. Since induced pluripotent stem cells (iPSCs) may offer a potent source of autologous cells to heal these wounds, we studied if repair-deficient fibroblasts, derived from DFU patients and age- and site-matched control fibroblasts, could be reprogrammed to iPSCs. To establish this, we used Sendai virus to successfully reprogram six primary fibroblast cell lines derived from ulcerated skin of two DFU patients (DFU8, DFU25), nonulcerated foot skin from two diabetic patients (DFF24, DFF9), and healthy foot skin from two nondiabetic patients (NFF12, NFF14). We confirmed reprogramming to a pluripotent state through three independent criteria: immunofluorescent staining for SSEA-4 and TRA-1-81, formation of embryoid bodies with differentiation potential to all three embryonic germ layers in vitro, and formation of teratomas in vivo. All iPSC lines showed normal karyotypes and typical, nonmethylated CpG sites for OCT4 and NANOG. iPSCs derived from DFUs were similar to those derived from site-matched nonulcerated skin from both diabetic and nondiabetic patients. These results have established for the first time that multiple, DFU-derived fibroblast cell lines can be reprogrammed with efficiencies similar to control fibroblasts, thus demonstrating their utility for future regenerative therapy of DFUs.
Collapse
Affiliation(s)
- Behzad Gerami-Naini
- 1 Department of Diagnostic Sciences, School of Dental Medicine, Tufts University , Boston, Massachusetts
| | - Avi Smith
- 1 Department of Diagnostic Sciences, School of Dental Medicine, Tufts University , Boston, Massachusetts
| | - Anna G Maione
- 2 Department of Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University , Boston, Massachusetts
| | - Olga Kashpur
- 1 Department of Diagnostic Sciences, School of Dental Medicine, Tufts University , Boston, Massachusetts
| | - Gianpaolo Carpinito
- 1 Department of Diagnostic Sciences, School of Dental Medicine, Tufts University , Boston, Massachusetts
| | - Aristides Veves
- 3 Microcirculation Laboratory and Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center, Harvard University , Boston, Massachusetts
| | - David J Mooney
- 4 Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University , Cambridge, Massachusetts
| | - Jonathan A Garlick
- 1 Department of Diagnostic Sciences, School of Dental Medicine, Tufts University , Boston, Massachusetts
| |
Collapse
|
30
|
Tsai PC, Zhang Z, Florek C, Michniak-Kohn BB. Constructing Human Skin Equivalents on Porcine Acellular Peritoneum Extracellular Matrix forIn VitroIrritation Testing. Tissue Eng Part A 2016; 22:111-22. [DOI: 10.1089/ten.tea.2015.0209] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Pei-Chin Tsai
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers-The State University of New Jersey, Piscataway, New Jersey
| | - Zheng Zhang
- New Jersey Center for Biomaterials, Rutgers-The State University of New Jersey, Piscataway, New Jersey
| | | | - Bozena B. Michniak-Kohn
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers-The State University of New Jersey, Piscataway, New Jersey
- New Jersey Center for Biomaterials, Rutgers-The State University of New Jersey, Piscataway, New Jersey
| |
Collapse
|
31
|
Miller KJ, Brown DA, Ibrahim MM, Ramchal TD, Levinson H. MicroRNAs in skin tissue engineering. Adv Drug Deliv Rev 2015; 88:16-36. [PMID: 25953499 DOI: 10.1016/j.addr.2015.04.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 04/04/2015] [Accepted: 04/25/2015] [Indexed: 01/08/2023]
Abstract
35.2 million annual cases in the U.S. require clinical intervention for major skin loss. To meet this demand, the field of skin tissue engineering has grown rapidly over the past 40 years. Traditionally, skin tissue engineering relies on the "cell-scaffold-signal" approach, whereby isolated cells are formulated into a three-dimensional substrate matrix, or scaffold, and exposed to the proper molecular, physical, and/or electrical signals to encourage growth and differentiation. However, clinically available bioengineered skin equivalents (BSEs) suffer from a number of drawbacks, including time required to generate autologous BSEs, poor allogeneic BSE survival, and physical limitations such as mass transfer issues. Additionally, different types of skin wounds require different BSE designs. MicroRNA has recently emerged as a new and exciting field of RNA interference that can overcome the barriers of BSE design. MicroRNA can regulate cellular behavior, change the bioactive milieu of the skin, and be delivered to skin tissue in a number of ways. While it is still in its infancy, the use of microRNAs in skin tissue engineering offers the opportunity to both enhance and expand a field for which there is still a vast unmet clinical need. Here we give a review of skin tissue engineering, focusing on the important cellular processes, bioactive mediators, and scaffolds. We further discuss potential microRNA targets for each individual component, and we conclude with possible future applications.
Collapse
|
32
|
Brown JP, Galassi TV, Stoppato M, Schiele NR, Kuo CK. Comparative analysis of mesenchymal stem cell and embryonic tendon progenitor cell response to embryonic tendon biochemical and mechanical factors. Stem Cell Res Ther 2015; 6:89. [PMID: 25956970 PMCID: PMC4425922 DOI: 10.1186/s13287-015-0043-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 11/07/2014] [Accepted: 03/05/2015] [Indexed: 01/14/2023] Open
Abstract
Introduction Advances in tendon engineering with mesenchymal stem cells (MSCs) are hindered by a need for cues to direct tenogenesis, and markers to assess tenogenic state. We examined the effects of factors involved in embryonic tendon development on adult MSCs, and compared MSC responses to that of embryonic tendon progenitor cells (TPCs), a model system of tenogenically differentiating cells. Methods Murine MSCs and TPCs subjected to cyclic tensile loading, transforming growth factor-β2 (TGFβ2), and fibroblast growth factor-4 (FGF4) in vitro were assessed for proliferation and mRNA levels of scleraxis, TGFβ2, tenomodulin, collagen type I and elastin. Results Before treatment, scleraxis and elastin levels in MSCs were lower than in TPCs, while other tendon markers expressed at similar levels in MSCs as TPCs. TGFβ2 alone and combined with loading were tenogenic based on increased scleraxis levels in both MSCs and TPCs. Loading alone had minimal effect. FGF4 downregulated tendon marker levels in MSCs but not in TPCs. Select tendon markers were not consistently upregulated with scleraxis, demonstrating the importance of characterizing a profile of markers. Conclusions Similar responses as TPCs to specific treatments suggest MSCs have tenogenic potential. Potentially shared mechanisms of cell function between MSCs and TPCs should be investigated in longer term studies.
Collapse
Affiliation(s)
- Jeffrey P Brown
- Department of Biomedical Engineering Tufts University, Science and Technology Center, 4 Colby Street , Medford, MA, 02155, USA.
| | - Thomas V Galassi
- Department of Biomedical Engineering Tufts University, Science and Technology Center, 4 Colby Street , Medford, MA, 02155, USA.
| | - Matteo Stoppato
- Department of Biomedical Engineering Tufts University, Science and Technology Center, 4 Colby Street , Medford, MA, 02155, USA.
| | - Nathan R Schiele
- Department of Biomedical Engineering Tufts University, Science and Technology Center, 4 Colby Street , Medford, MA, 02155, USA.
| | - Catherine K Kuo
- Department of Biomedical Engineering Tufts University, Science and Technology Center, 4 Colby Street , Medford, MA, 02155, USA. .,Cell, Molecular & Developmental Biology Program Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 145 Harrison Avenue, Boston, MA, 02111, USA.
| |
Collapse
|
33
|
Hewitt KJ, Shamis Y, Gerami-Naini B, Garlick JA. Strategies for Oral Mucosal Repair by Engineering 3D Tissues with Pluripotent Stem Cells. Adv Wound Care (New Rochelle) 2014; 3:742-750. [PMID: 25493208 DOI: 10.1089/wound.2013.0480] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 09/03/2013] [Indexed: 11/12/2022] Open
Abstract
Significance: Human-induced pluripotent stem cells (iPSC) can be differentiated into patient-specific cells with a wide spectrum of cellular phenotypes and offer an alternative source of autologous cells for therapeutic use. Recent studies have shown that iPSC-derived fibroblasts display enhanced cellular functions suggesting that iPSC may eventually become an important source of stem cells for regenerative therapies. Recent Advances: The discovery of approaches to reprogram somatic cells into pluripotent cells opens exciting avenues for their use in personalized, regenerative therapies. The controlled differentiation of functional cell types from iPSC provides a replenishing source of fibroblasts. There is intriguing evidence that iPSC reprogramming and subsequent differentiation to fibroblast lineages may improve cellular functional properties. Augmenting the biological potency of iPSC-derived fibroblasts may enable the development of novel, personalized stem cell therapies to treat oral disease. Critical Issues: Numerous questions need to be addressed before iPSC-derived cells can be used as a practical oral therapy. This will include understanding why iPSC-derived cells are predisposed towards differentiation pathways along lineages related to their cell of origin, screening iPSC-derived cells to ensure their safety and phenotypic stability and developing engineered, three-dimensional tissue models to optimize their function and efficacy for future therapeutic transplantation. Future Directions: Future research will need to address how to develop efficient methods to deliver and integrate iPSC-derived fibroblasts into the oral mucosa. This will require an improved understanding of how to harness their biological potency for regenerative therapies that are specifically targeted to the oral mucosa.
Collapse
Affiliation(s)
- Kyle J. Hewitt
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Yulia Shamis
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Behzad Gerami-Naini
- Department of Oral and Maxillofacial Pathology, Tufts University, Boston, Massachusetts
| | - Jonathan A. Garlick
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
- Department of Oral and Maxillofacial Pathology, Tufts University, Boston, Massachusetts
| |
Collapse
|
34
|
Del Pino A, Ligero G, López MB, Navarro H, Carrillo JA, Pantoll SC, Díaz de la Guardia R. Morphology, cell viability, karyotype, expression of surface markers and plasticity of three human primary cell line cultures before and after the cryostorage in LN2 and GN2. Cryobiology 2014; 70:1-8. [PMID: 25445570 DOI: 10.1016/j.cryobiol.2014.10.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 09/23/2014] [Accepted: 10/29/2014] [Indexed: 01/09/2023]
Abstract
Primary cell line cultures from human skin biopsies, adipose tissue and tumor tissue are valuable samples for research and therapy. In this regard, their derivation, culture, storage, transport and thawing are important steps to be studied. Towards this end, we wanted to establish the derivation, and identify the culture characteristics and the loss of viability of three human primary cell line cultures (human adult dermal fibroblasts (hADFs), human adult mesenchymal stem cells (hMSCs), and primary culture of tumor cells from lung adenocarcinoma (PCTCLA)). Compared to fresh hADFs, hMSCs and PCTCLA, thawed cells stored in a cryogenic Dewar tanks with liquid nitrogen (LN2), displayed 98.20% ± 0.99, 95.40% ± 1.41 and 93.31% ± 3.83 of cell viability, respectively. Thawed cells stored in a Dry Vapor Shipper container with gas phase (GN2), for 20 days, in addition displayed 4.61% ± 2.78, 3.70% ± 4.09 and 9.13% ± 3.51 of average loss of cells viability, respectively, showing strong correlation between the loss of viability in hADFs and the number of post-freezing days in the Dry Vapor Shipper. No significant changes in morphological characteristics or in the expression of surface markers (being hADFs, hMSCs and PCTCLA characterized by positive markers CD73+; CD90+; CD105+; and negative markers CD14-; CD20-; CD34-; and CD45-; n=2) were found. Chromosome abnormalities in the karyotype were not found. In addition, under the right conditions hMSCs were differentiated into adipogenic, osteogenic and chondrogenic lineages in vitro. In this paper, we have shown the characteristics of three human primary cell line cultures when they are stored in LN2 and GN2.
Collapse
Affiliation(s)
- Alberto Del Pino
- Biobanco del Sistema Sanitario Público de Andalucía (BBSSPA), Centro de Investigaciones Biomédicas, Consejería de Salud - Universidad de Granada, Granada, Spain
| | - Gertrudis Ligero
- Biobanco del Sistema Sanitario Público de Andalucía (BBSSPA), Centro de Investigaciones Biomédicas, Consejería de Salud - Universidad de Granada, Granada, Spain
| | - María B López
- Department of Physiology, Institute of Nutrition and Food Technology, University of Granada, Granada, Spain
| | - Héctor Navarro
- Biobanco del Sistema Sanitario Público de Andalucía (BBSSPA), Centro de Investigaciones Biomédicas, Consejería de Salud - Universidad de Granada, Granada, Spain
| | - Jose A Carrillo
- Biobanco del Sistema Sanitario Público de Andalucía (BBSSPA), Centro de Investigaciones Biomédicas, Consejería de Salud - Universidad de Granada, Granada, Spain
| | - Siobhan C Pantoll
- Biobanco del Sistema Sanitario Público de Andalucía (BBSSPA), Centro de Investigaciones Biomédicas, Consejería de Salud - Universidad de Granada, Granada, Spain
| | - Rafael Díaz de la Guardia
- Biobanco del Sistema Sanitario Público de Andalucía (BBSSPA), Centro de Investigaciones Biomédicas, Consejería de Salud - Universidad de Granada, Granada, Spain.
| |
Collapse
|
35
|
Quan R, Zheng X, Xu S, Zhang L, Yang D. Gelatin-chondroitin-6-sulfate-hyaluronic acid scaffold seeded with vascular endothelial growth factor 165 modified hair follicle stem cells as a three-dimensional skin substitute. Stem Cell Res Ther 2014; 5:118. [PMID: 25331352 PMCID: PMC4535258 DOI: 10.1186/scrt508] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 10/10/2014] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION In the field of skin tissue engineering, gelatin-chondroitin-6-sulfate-hyaluronic acid (Gel-C6S-HA) stents are a suitable bio skin substitute. The purpose was to investigate the effect of genetically-modified hair follicle stem cells (HFSCs), combined with Gel-C6S-HA scaffolds, on the vascularization of tissue-engineered skin. METHODS Three-dimensional (3D) Gel-C6S-HA scaffolds were prepared by freeze-drying. Vascular endothelial growth factor (VEGF) 165 gene-modified rat HFSCs (rHFSCs) were inoculated into the scaffolds and cultured for 7 days. Two bilateral full-thickness skin defects were created on the back of 18 Sprague-Dawley rats. Rats were randomly divided into four groups: Group A, HFSCs transduced with VEGF165 seeded onto Gel-C6S-HA scaffolds; Group B, HFSCs transduced with empty vector seeded onto Gel-C6S-HA scaffolds; Group C, Gel-C6S-HA scaffold only; Group D, Vaseline gauze dressing. These compositions were implanted onto the defects and harvested at 7, 14 and 21 days. Wound healing was assessed and compared among groups according to hematoxylin-eosin staining, CD31 expression, alpha smooth muscle actin (α-SMA) and major histocompatibility complex class I (MHC-I) immunohistochemistry, and microvessel density (MVD) count, to evaluate the new blood vessels. RESULTS SEM revealed the Gel-C6S-HA scaffold was spongy and 3D, with an average pore diameter of 133.23 ± 43.36 μm. Cells seeded on scaffolds showed good adherent growth after 7 days culture. No significant difference in rHFSC morphology, adherence and proliferative capacity was found before and after transfection (P >0.05). After 14 and 21 days, the highest rate of wound healing was observed in Group A (P <0.05). Histological and immunological examination showed that after 21 days, MVD also reached a maximum in Group A (P <0.05). Therefore, the number of new blood vessels formed within the skin substitutes was greatest in Group A, followed by Group B. In Group C, only trace amounts of mature subcutaneous blood vessels were observed, and few subcutaneous tissue cells migrated into the scaffolds. CONCLUSIONS Tissue-engineered skin constructs, using 3D Gel-C6S-HA scaffolds seeded with VEGF165-modified rHFSCs, resulted in promotion of angiogenesis during wound healing and facilitation of vascularization in skin substitutes. This may be a novel approach for tissue-engineered skin substitutes.
Collapse
|
36
|
Otsu K, Kumakami-Sakano M, Fujiwara N, Kikuchi K, Keller L, Lesot H, Harada H. Stem cell sources for tooth regeneration: current status and future prospects. Front Physiol 2014; 5:36. [PMID: 24550845 PMCID: PMC3912331 DOI: 10.3389/fphys.2014.00036] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 01/17/2014] [Indexed: 12/13/2022] Open
Abstract
Stem cells are capable of renewing themselves through cell division and have the remarkable ability to differentiate into many different types of cells. They therefore have the potential to become a central tool in regenerative medicine. During the last decade, advances in tissue engineering and stem cell-based tooth regeneration have provided realistic and attractive means of replacing lost or damaged teeth. Investigation of embryonic and adult (tissue) stem cells as potential cell sources for tooth regeneration has led to many promising results. However, technical and ethical issues have hindered the availability of these cells for clinical application. The recent discovery of induced pluripotent stem (iPS) cells has provided the possibility to revolutionize the field of regenerative medicine (dentistry) by offering the option of autologous transplantation. In this article, we review the current progress in the field of stem cell-based tooth regeneration and discuss the possibility of using iPS cells for this purpose.
Collapse
Affiliation(s)
- Keishi Otsu
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University Yahaba, Japan
| | - Mika Kumakami-Sakano
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University Yahaba, Japan
| | - Naoki Fujiwara
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University Yahaba, Japan
| | - Kazuko Kikuchi
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University Yahaba, Japan ; Division of Special Care Dentistry, Department of Developmental Oral Health Science, Iwate Medical University Morioka, Japan
| | - Laetitia Keller
- INSERM UMR 1109, team "Osteoarticular and Dental Regenerative NanoMedicine", Faculté de Médecine, Université de Strasbourg Strasbourg, France
| | - Hervé Lesot
- INSERM UMR 1109, team "Osteoarticular and Dental Regenerative NanoMedicine", Faculté de Médecine, Université de Strasbourg Strasbourg, France ; Faculté de Chirurgie dentaire, Université de Strasbourg Strasbourg, France
| | - Hidemitsu Harada
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University Yahaba, Japan
| |
Collapse
|
37
|
Shamis Y, Silva EA, Hewitt KJ, Brudno Y, Levenberg S, Mooney DJ, Garlick JA. Fibroblasts derived from human pluripotent stem cells activate angiogenic responses in vitro and in vivo. PLoS One 2013; 8:e83755. [PMID: 24386271 PMCID: PMC3875480 DOI: 10.1371/journal.pone.0083755] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 11/07/2013] [Indexed: 01/08/2023] Open
Abstract
Human embryonic and induced pluripotent stem cells (hESC/hiPSC) are promising cell sources for the derivation of large numbers of specific cell types for tissue engineering and cell therapy applications. We have describe a directed differentiation protocol that generates fibroblasts from both hESC and hiPSC (EDK/iPDK) that support the repair and regeneration of epithelial tissue in engineered, 3D skin equivalents. In the current study, we analyzed the secretory profiles of EDK and iPDK cells to investigate the production of factors that activate and promote angiogenesis. Analysis of in vitro secretion profiles from EDK and iPDK cells demonstrated the elevated secretion of pro-angiogenic soluble mediators, including VEGF, HGF, IL-8, PDGF-AA, and Ang-1, that stimulated endothelial cell sprouting in a 3D model of angiogenesis in vitro. Phenotypic analysis of EDK and iPDK cells during the course of differentiation from hESCs and iPSCs revealed that both cell types progressively acquired pericyte lineage markers NG2, PDGFRβ, CD105, and CD73 and demonstrated transient induction of pericyte progenitor markers CD31, CD34, and Flk1/VEGFR2. Furthermore, when co-cultured with endothelial cells in 3D fibrin-based constructs, EDK and iPDK cells promoted self-assembly of vascular networks and vascular basement membrane deposition. Finally, transplantation of EDK cells into mice with hindlimb ischemia significantly reduced tissue necrosis and improved blood perfusion, demonstrating the potential of these cells to stimulate angiogenic responses in vivo. These findings demonstrate that stable populations of pericyte-like angiogenic cells can be generated with high efficiency from hESC and hiPSC using a directed differentiation approach. This provides new cell sources and opportunities for vascular tissue engineering and for the development of novel strategies in regenerative medicine.
Collapse
Affiliation(s)
- Yulia Shamis
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Eduardo A. Silva
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
- Wyss Institute For Biological Inspired Engineering, Harvard University, Boston, Massachusetts, United States of America
| | - Kyle J. Hewitt
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Yevgeny Brudno
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
- Wyss Institute For Biological Inspired Engineering, Harvard University, Boston, Massachusetts, United States of America
| | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - David J. Mooney
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Jonathan A. Garlick
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
38
|
Chen M, Su W, Lin X, Guo Z, Wang J, Zhang Q, Brand D, Ryffel B, Huang J, Liu Z, He X, Le AD, Zheng SG. Adoptive transfer of human gingiva-derived mesenchymal stem cells ameliorates collagen-induced arthritis via suppression of Th1 and Th17 cells and enhancement of regulatory T cell differentiation. ARTHRITIS AND RHEUMATISM 2013; 65:1181-93. [PMID: 23400582 PMCID: PMC4364405 DOI: 10.1002/art.37894] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 01/31/2013] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Current approaches offer no cures for rheumatoid arthritis (RA). Accumulating evidence has revealed that manipulation of bone marrow-derived mesenchymal stem cells (BM-MSCs) may have the potential to control or even prevent RA, but BM-MSC-based therapy faces many challenges, such as limited cell availability and reduced clinical feasibility. This study in mice with established collagen-induced arthritis (CIA) was undertaken to determine whether substitution of human gingiva-derived mesenchymal stem cells (G-MSCs) would significantly improve the therapeutic effects. METHODS CIA was induced in DBA/1J mice by immunization with type II collagen and Freund's complete adjuvant. G-MSCs were injected intravenously into the mice on day 14 after immunization. In some experiments, intraperitoneal injection of PC61 (anti-CD25 antibody) was used to deplete Treg cells in arthritic mice. RESULTS Infusion of G-MSCs in DBA/1J mice with CIA significantly reduced the severity of arthritis, decreased the histopathology scores, and down-regulated the production of inflammatory cytokines (interferon-γ and interleukin-17A). Infusion of G-MSCs also resulted in increased levels of CD4+CD39+FoxP3+ cells in arthritic mice. These increases were noted early after infusion in the spleens and lymph nodes, and later after infusion in the synovial fluid. The FoxP3+ Treg cells that were increased in frequency mainly consisted of Helios-negative cells. When Treg cells were depleted, infusion of G-MSCs partially interfered with the progression of CIA. Pretreatment of G-MSCs with a CD39 or CD73 inhibitor significantly reversed the protective effect of G-MSCs on CIA. CONCLUSION The role of G-MSCs in controlling the development and severity of CIA mostly depends on CD39/CD73 signals and partially depends on the induction of CD4+CD39+FoxP3+ Treg cells. G-MSCs provide a promising approach for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Maogen Chen
- Organ Transplant center, 1st affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P.R. China
- Division of Rheumatology and Immunology, Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA. 90033, USA
| | - Wenru Su
- Division of Surgical, Therapeutic and Bioengineering Sciences, Center for Craniofacial Molecular Biology, University of Southern California, Keck School of Medicine, Los Angeles, CA. 90033, USA
| | - Xiaohong Lin
- Division of Rheumatology and Immunology, Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA. 90033, USA
- Division of Surgery, First affiliated Hospital of Shantou University, Shantou, 515041 China
| | - Zhiyong Guo
- Organ Transplant center, 1st affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Julie Wang
- Division of Rheumatology and Immunology, Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA. 90033, USA
| | - Qunzhou Zhang
- Division of Surgical, Therapeutic and Bioengineering Sciences, Center for Craniofacial Molecular Biology, University of Southern California, Keck School of Medicine, Los Angeles, CA. 90033, USA
| | - David Brand
- Research Service, Veterans Affairs Medical Center, Memphis; TN. 38104, USA
| | - Bernhard Ryffel
- UMR6218, Molecular Immunology, University and CNRS, 3b rue de la Ferollerie, Orleans. 45071, France
| | - Jiefu Huang
- Organ Transplant center, 1st affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Zhongmin Liu
- Institute of Immunology, Shanghai East Hospital at Tongji University, Shanghai, 200120, China
| | - Xiaoshun He
- Organ Transplant center, 1st affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Anh D. Le
- Division of Surgical, Therapeutic and Bioengineering Sciences, Center for Craniofacial Molecular Biology, University of Southern California, Keck School of Medicine, Los Angeles, CA. 90033, USA
| | - Song Guo Zheng
- Division of Rheumatology and Immunology, Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA. 90033, USA
- Institute of Immunology, Shanghai East Hospital at Tongji University, Shanghai, 200120, China
| |
Collapse
|
39
|
Hewitt KJ, Garlick JA. Cellular reprogramming to reset epigenetic signatures. Mol Aspects Med 2012; 34:841-8. [PMID: 22982217 DOI: 10.1016/j.mam.2012.08.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 08/27/2012] [Indexed: 12/25/2022]
Abstract
The controlled differentiation of induced pluripotent stem cells (iPSC) towards clinically-relevant cell types has benefitted from epigenetic profiling of lineage-specific markers to confirm the phenotype of iPSC-derived cells. Mapping epigenetic marks throughout the genome has identified unique changes which occur in the DNA methylation profile of cells as they differentiate to specific cell types. Beyond characterizing the development of cells derived from pluripotent stem cells, the process of reprogramming cells to iPSC resets lineage-specific DNA methylation marks established during differentiation to specific somatic cell types. This property of reprogramming has potential utility in reverting aberrant epigenetic alterations in nuclear organization that are linked to disease progression. Since DNA methylation marks are reset following reprogramming, and contribute to restarting developmental programs, it is possible that DNA methylation marks associated with the disease state may also be erased in these cells. The subsequent differentiation of such cells could result in cell progeny that will function effectively as therapeutically-competent cell types for use in regenerative medicine. This suggests that through reprogramming it may be possible to directly modify the epigenetic memory of diseased cells and help to normalize their cellular phenotype, while also broadening our understanding of disease pathogenesis.
Collapse
Affiliation(s)
- Kyle J Hewitt
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, 1111 Highland Ave., Madison, WI 53705, USA
| | | |
Collapse
|
40
|
Adipose-derived stromal cells accelerate wound healing in an organotypic raft culture model. Ann Plast Surg 2012; 68:501-4. [PMID: 22510896 DOI: 10.1097/sap.0b013e31823b69fc] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Adipose tissue is a known reservoir of multipotent mesenchymal stem cells, which can be manipulated in culture to produce cells with different phenotypes. The goal of this study was to determine whether the addition of these multipotential cells to organotypic, human skin equivalent cultures would accelerate wound healing after laser injury. For our initial studies, we were able to obtain 3-dimensional raft cultures from adult skin explanted directly onto the dermal equivalent containing human fibroblasts with or without adipose-derived stromal cells (ADSCs). Two days after laser injury, the raft cultures of skin explants that contained ADSCs had a completely healed multilayered epidermis, whereas the control raft culture without the adipose-derived cells still had areas of injury. With this encouraging outcome, these experiments were then repeated in a raft culture system initiated from dissociated primary adult human keratinocytes on the humanized dermal equivalent. Again, the cultures containing ADSCs healed faster than the control cultures. In conclusion, these data provide support to our hypothesis that ADSCs are an excellent and readily available source of factors necessary for accelerated wound healing and tissue regeneration.
Collapse
|
41
|
Egusa H, Sonoyama W, Nishimura M, Atsuta I, Akiyama K. Stem cells in dentistry--part I: stem cell sources. J Prosthodont Res 2012; 56:151-65. [PMID: 22796367 DOI: 10.1016/j.jpor.2012.06.001] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 06/14/2012] [Indexed: 12/21/2022]
Abstract
Stem cells can self-renew and produce different cell types, thus providing new strategies to regenerate missing tissues and treat diseases. In the field of dentistry, adult mesenchymal stem/stromal cells (MSCs) have been identified in several oral and maxillofacial tissues, which suggests that the oral tissues are a rich source of stem cells, and oral stem and mucosal cells are expected to provide an ideal source for genetically reprogrammed cells such as induced pluripotent stem (iPS) cells. Furthermore, oral tissues are expected to be not only a source but also a therapeutic target for stem cells, as stem cell and tissue engineering therapies in dentistry continue to attract increasing clinical interest. Part I of this review outlines various types of intra- and extra-oral tissue-derived stem cells with regard to clinical availability and applications in dentistry. Additionally, appropriate sources of stem cells for regenerative dentistry are discussed with regard to differentiation capacity, accessibility and possible immunomodulatory properties.
Collapse
Affiliation(s)
- Hiroshi Egusa
- Department of Fixed Prosthodontics, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | |
Collapse
|
42
|
Justewicz DM, Shokes JE, Reavis B, Boyd SA, Burnette TB, Halberstadt CR, Spencer T, Ludlow JW, Bertram TA, Jain D. Characterization of the human smooth muscle cell secretome for regenerative medicine. Tissue Eng Part C Methods 2012; 18:797-816. [PMID: 22530582 DOI: 10.1089/ten.tec.2012.0054] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Smooth muscle cells (SMC) play a central role in maintaining the structural and functional integrity of muscle tissue. Little is known about the early in vitro events that guide the assembly of 'bioartificial tissue' (constructs) and recapitulate the key aspects of smooth muscle differentiation and development before surgical implantation. Biomimetic approaches have been proposed that enable the identification of in vitro processes which allow standardized manufacturing, thus improving both product quality and the consistency of patient outcomes. One essential element of this approach is the description of the SMC secretome, that is, the soluble and deposited factors produced within the three-dimensional (3D) extracellular matrix (ECM) microenvironment. In this study, we utilized autologous SMC from multiple tissue types that were expanded ex vivo and generated with a rigorous focus on operational phenotype and genetic stability. The objective of this study was to characterize the spatiotemporal dynamics of the first week of organoid maturation using a well-defined in vitro-like, 3D-engineered scale model of our validated manufacturing process. Functional proteomics was used to identify the topological properties of the networks of interacting proteins that were derived from the SMC secretome, revealing overlapping central nodes related to SMC differentiation and proliferation, actin cytoskeleton regulation, and balanced ECM accumulation. The critical functions defined by the Ingenuity Pathway Analysis included cell signaling, cellular movement and proliferation, and cellular and organismal development. The results confirm the phenotypic and functional similarity of the SMC generated by our platform technology at the molecular level. Furthermore, these data validate the biomimetic approaches that have been established to maintain manufacturing consistency.
Collapse
Affiliation(s)
- Dominic M Justewicz
- Department of Bioprocess Research & Development, Tengion, Inc., 3929 Westpoint Blvd., Suite G, Winston-Salem, NC 27103, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hewitt KJ, Shamis Y, Knight E, Smith A, Maione A, Alt-Holland A, Sheridan SD, Haggarty SJ, Garlick JA. PDGFRβ expression and function in fibroblasts derived from pluripotent cells is linked to DNA demethylation. J Cell Sci 2012; 125:2276-87. [PMID: 22344267 DOI: 10.1242/jcs.099192] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Platelet-derived growth factor receptor-beta (PDGFRβ) is required for the development of mesenchymal cell types, and plays a diverse role in the function of fibroblasts in tissue homeostasis and regeneration. In this study, we characterized the expression of PDGFRβ in fibroblasts derived from human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), and showed that this expression is important for cellular functions such as migration, extracellular matrix production and assembly in 3D self-assembled tissues. To determine potential regulatory regions predictive of expression of PDGFRβ following differentiation from ESCs and iPSCs, we analyzed the DNA methylation status of a region of the PDGFRB promoter that contains multiple CpG sites, before and after differentiation. We demonstrated that this promoter region is extensively demethylated following differentiation, and represents a developmentally regulated, differentially methylated region linked to PDGFRβ expression. Understanding the epigenetic regulation of genes such as PDGFRB, and identifying sites of active DNA demethylation, is essential for future applications of iPSC-derived fibroblasts for regenerative medicine.
Collapse
Affiliation(s)
- Kyle J Hewitt
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
iPSC-derived fibroblasts demonstrate augmented production and assembly of extracellular matrix proteins. In Vitro Cell Dev Biol Anim 2012; 48:112-22. [PMID: 22259014 DOI: 10.1007/s11626-011-9478-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 12/21/2011] [Indexed: 12/26/2022]
Abstract
Reprogramming of somatic cells to induced pluripotent stem cells (iPSC) provides an important cell source to derive patient-specific cells for potential therapeutic applications. However, it is not yet clear whether reprogramming through pluripotency allows the production of differentiated cells with improved functional properties that may be beneficial in regenerative therapies. To address this, we compared the production and assembly of extracellular matrix (ECM) by iPSC-derived fibroblasts to that of the parental, dermal fibroblasts (BJ), from which these iPSC were initially reprogrammed, and to fibroblasts differentiated from human embryonic stem cells (hESC). iPSC- and hESC-derived fibroblasts demonstrated stable expression of surface markers characteristic of stromal fibroblasts during prolonged culture and showed an elevated growth potential when compared to the parental BJ fibroblasts. We found that in the presence of L: -ascorbic acid-2-phosphate, iPSC- and hESC-derived fibroblasts increased their expression of collagen genes, secretion of soluble collagen, and extracellular deposition of type I collagen to a significantly greater degree than that seen in the parental BJ fibroblasts. Under culture conditions that enabled the self-assembly of a 3D stromal tissue, iPSC- and hESC-derived fibroblasts generated a well organized, ECM that was enriched in type III collagen. By characterizing the functional properties of iPSC-derived fibroblasts compared to their parental fibroblasts, we demonstrate that these cells represent a promising, alternative source of fibroblasts to advance future regenerative therapies.
Collapse
|