1
|
Kuenzel NA, Dobner J, Reichert D, Rossi A, Boukamp P, Esser C. Vδ1 T Cells Integrated in Full-Thickness Skin Equivalents: A Model for the Role of Human Skin-Resident γδT Cells. J Invest Dermatol 2024:S0022-202X(24)02173-0. [PMID: 39384018 DOI: 10.1016/j.jid.2024.08.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/25/2024] [Accepted: 08/12/2024] [Indexed: 10/11/2024]
Abstract
Vδ1 T cells are a subpopulation of γδT cells found in human dermis. Much less is known regarding their role and function in skin health and disease than regarding the roles of murine skin-resident γδT cells. In this study, we report the successful integration of Vδ1 T cells into long-term fibroblast-derived matrix skin equivalents. We isolated Vδ1 T cells from human blood, where they are rare, and established conditions for the integration and maintenance of the freshly isolated Vδ1 T cells in the skin equivalents. Plated on top of the dermal equivalents, almost all Vδ1 T cells migrated into the dermal matrix where they exerted their influence on both the dermal equivalents and the epithelium. Vδ1 T cells contributed to epidermal differentiation of HaCaT cells as indicated by histology, expression of epidermal differentiation markers, and RNA-sequencing expression profile. When complemented with the carcinoma-derived SCC13 cells instead of HaCaT, our data suggest a role for Vδ1 T cells in slowing growth of the tumor cells, as indicated by reduced stratification and changes in gene expression profiles. Together, we demonstrate the successful establishment of human Vδ1 T cell-competent skin equivalents and skin carcinoma equivalents and provide evidence for molecular and functional consequences of the Vδ1 T cells on their respective environment.
Collapse
Affiliation(s)
| | - Jochen Dobner
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Doreen Reichert
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Andrea Rossi
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Petra Boukamp
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany; German Cancer Research Centre, Heidelberg, Germany
| | - Charlotte Esser
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| |
Collapse
|
2
|
Yasti AÇ, Çolak B, Özcan F, Kismet K, Sürel AA, Akgün AE, Akin M. Oxygen transmission rates of skin substitutes and graft survival. Burns 2023; 49:1654-1662. [PMID: 37280139 DOI: 10.1016/j.burns.2023.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/17/2023] [Accepted: 05/18/2023] [Indexed: 06/08/2023]
Abstract
AIM Oxygen is required for cell migration into the scaffold and for the survival of the overlying graft in the use of a single-layer scaffold. In the absence of diffusion from the avascular wound base, such as in areas above the bone/tendon, oxygen delivery from the lateral edges of the scaffold is important. This study compared the oxygen permeability of skin scaffolds, currently commercially available in Turkey (Nevelia®, MatriDerm®, and Pelnac®), in the lateral plane. MATERIALS AND METHODS To measure oxygen permeability, an interconnected closed system was created. Oxygen permeability was evaluated based on the color change that occurred as a result of the reaction of iron with oxygen. After the dermal matrices placed in the closed system were exposed to oxygen, the color change on the surface of the dermal matrices was measured, and electron microscopic images were recorded to compare deformation before and after the procedure. RESULTS Two scaffolds did not show deformation after the procedure while Pelnac® had minimal deformation. The oxygen rates on the nitrogen side of the test apparatus were found to be 29%, 34%, and 27% for Nevelia®, MatriDerm®, and Pelnac®, respectively; and the oxygen transmission lengths (length of color change) of these scaffolds in the lateral plane were 1, 2, and 0.5 cm, respectively. CONCLUSION Although none of the scaffolds showed significant deformation, and all continued to exhibit their scaffold properties after the procedure, MatriDerm® was determined to be the most suitable scaffold for use in avascular areas, with a 2-cm oxygen transmission length in terms of lateral oxygenation.
Collapse
Affiliation(s)
- Ahmet Çınar Yasti
- Health Sciences University Medical School, Chair: Burn Treatment Center, City Hospital, Turkey
| | - Bayram Çolak
- Selçuk University Medical School, Department of General Surgery, Konya, Turkey
| | - Fatih Özcan
- Selçuk University Faculty of Science, Department of Chemistry, Konya, Turkey
| | - Kemal Kismet
- General Surgery, Selçuk University, Chair Faculty of Nursing, Department of Surgical Nursing, Konya, Turkey
| | - Aziz Ahmet Sürel
- Health Sciences University Medical School, Ankara City Hospital, Department of General Surgery, Ankara, Turkey
| | - Ali Emre Akgün
- Ankara City Hospital, General Surgery, Burn Treatment Center, Turkey
| | - Merve Akin
- Ankara City Hospital, General Surgery, Burn Treatment Center, Turkey.
| |
Collapse
|
3
|
Joshi A, Nuntapramote T, Brüggemann D. Self-Assembled Fibrinogen Scaffolds Support Cocultivation of Human Dermal Fibroblasts and HaCaT Keratinocytes. ACS OMEGA 2023; 8:8650-8663. [PMID: 36910955 PMCID: PMC9996769 DOI: 10.1021/acsomega.2c07896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 01/31/2023] [Indexed: 06/18/2023]
Abstract
Self-assembled fibrinogen scaffolds are highly attractive biomaterials to mimic native blood clots. To explore their potential for wound healing, we studied the interaction of cocultures of human dermal fibroblasts (HDFs) and HaCaT keratinocytes with nanofibrous, planar, and physisorbed fibrinogen. Cell viability analysis indicated that the growth of HDFs and HaCaTs was supported by all fibrinogen topographies until 14 days, either in mono- or coculture. Using scanning electron microscopy and cytoskeletal staining, we observed that the native morphology of both cell types was preserved on all topographies. Expression of the marker proteins vimentin and cytokeratin-14 showed that the native phenotype of fibroblasts and undifferentiated keratinocytes, respectively, was maintained. HDFs displayed their characteristic wound healing phenotype, characterized by expression of fibronectin. Finally, to mimic the multilayered microenvironment of skin, we established successive cocultures of both cells, for which we found consistently high metabolic activities. SEM analysis revealed that HaCaTs arranged into a confluent top layer after 14 days, while fluorescent labeling confirmed the presence of both cells in the layered structure after 6 days. In conclusion, all fibrinogen topographies successfully supported the cocultivation of fibroblasts and keratinocytes, with fibrinogen nanofibers being particularly attractive for skin regeneration due to their biomimetic porous architecture and the technical possibility to be detached from an underlying substrate.
Collapse
Affiliation(s)
- Arundhati Joshi
- Institute
for Biophysics, University of Bremen, Otto-Hahn-Allee 1, 28359 Bremen, Germany
| | - Titinun Nuntapramote
- Institute
for Biophysics, University of Bremen, Otto-Hahn-Allee 1, 28359 Bremen, Germany
| | - Dorothea Brüggemann
- Institute
for Biophysics, University of Bremen, Otto-Hahn-Allee 1, 28359 Bremen, Germany
- MAPEX
Center for Materials and Processes, University
of Bremen, 28359 Bremen, Germany
| |
Collapse
|
4
|
Tham M, Stark HJ, Jauch A, Harwood C, Pavez Lorie E, Boukamp P. Adverse Effects of Vemurafenib on Skin Integrity: Hyperkeratosis and Skin Cancer Initiation Due to Altered MEK/ERK-Signaling and MMP Activity. Front Oncol 2022; 12:827985. [PMID: 35174094 PMCID: PMC8842679 DOI: 10.3389/fonc.2022.827985] [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: 12/02/2021] [Accepted: 01/03/2022] [Indexed: 11/24/2022] Open
Abstract
The BRAF inhibitor vemurafenib, approved for treating patients with BRAF V600E-mutant and unresectable or metastatic melanomas, rapidly induces cutaneous adverse events, including hyperkeratotic skin lesions and cutaneous squamous cell carcinomas (cSCC). To determine, how vemurafenib would provoke these adverse events, we utilized long-term in vitro skin equivalents (SEs) comprising epidermal keratinocytes and dermal fibroblasts in their physiological environment. We inserted keratinocytes with different genetic background [normal keratinocytes: NHEK, HaCaT (p53/mut), and HrasA5 (p53/mut+Hras/mut)] to analyze effects depending on the stage of carcinogenesis. We now show that vemurafenib activates MEK-ERK signaling in both, keratinocytes, and fibroblasts in vitro and in the in vivo-like SEs. As a consequence, vemurafenib does not provide a growth advantage but leads to a differentiation phenotype, causing accelerated differentiation and hyperkeratosis in the NHEK and normalized stratification and cornification in the transformed keratinocytes. Although all keratinocytes responded very similarly to vemurafenib in their expression profile, particularly with a significant induction of MMP1 and MMP3, only the HrasA5 cells revealed a vemurafenib-dependent pathophysiological shift to tumor progression, i.e., the initiation of invasive growth. This was shown by increased proteolytic activity allowing for penetration of the basement membrane and invasion into the disrupted underlying matrix. Blocking MMP activity, by the addition of ilomastat, prevented invasion with all corresponding degradative activities, thus substantiating that the RAS-RAF-MEK-ERK/MMP axis is the most important molecular basis for the rapid switch towards tumorigenic conversion of the HrasA5 keratinocytes upon vemurafenib treatment. Finally, cotreatment with vemurafenib and the MEK inhibitor cobimetinib prevented MEK-ERK hyperactivation and with that abolished both, the epidermal differentiation and the tumor invasion phenotype. This suggests that both cutaneous adverse events are under direct control of vemurafenib-dependent MEK-ERK hyperactivation and confirms the dependence on preexisting genetic alterations of the skin keratinocytes that determine the basis towards induction of tumorigenic progression.
Collapse
Affiliation(s)
- Marius Tham
- Department of Genetics of Skin Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hans-Jürgen Stark
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Anna Jauch
- Institute of Human Genetics, University Heidelberg, Heidelberg, Germany
| | - Catherine Harwood
- Department of Dermatology, Royal London Hospital, Barts Health NHS Trust, London, United Kingdom.,Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Petra Boukamp
- Department of Genetics of Skin Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany.,IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| |
Collapse
|
5
|
Iriyama S, Ogura Y, Nishikawa S, Hosoi J, Amano S. Regeneration of collagen fibrils at the papillary dermis by reconstructing basement membrane at the dermal-epidermal junction. Sci Rep 2022; 12:795. [PMID: 35039587 PMCID: PMC8764085 DOI: 10.1038/s41598-022-04856-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/28/2021] [Indexed: 11/13/2022] Open
Abstract
The epidermal basement membrane deteriorates with aging. We previously reported that basement membrane reconstruction not only serves to maintain epidermal stem/progenitor cells in the epidermis, but also increases collagen fibrils in the papillary dermis. Here, we investigated the mechanism of the latter action. Collagen fibrils in the papillary dermis were increased in organotypic human skin culture treated with matrix metalloproteinase and heparinase inhibitors. The expression levels of COL5A1 and COL1A1 genes (encoding collagen type V α 1 chain and collagen type I α 1 chain, respectively) were increased in fibroblasts cultured with conditioned medium from a skin equivalent model cultured with the inhibitors and in keratinocytes cultured on laminin-511 E8 fragment-coated plates. We then examined cytokine expression, and found that the inhibitors increased the expression of PDGF-BB (platelet-derived growth factor consisting of two B subunits) in epidermis. Expression of COL5A1 and COL1A1 genes was increased in cultured fibroblasts stimulated with PDGF-BB. Further, the bifunctional inhibitor hydroxyethyl imidazolidinone (HEI) increased skin elasticity and the thickness of the papillary dermis in the skin equivalent. Taken together, our data suggests that reconstructing the basement membrane promotes secretion of PDGF-BB by epidermal keratinocytes, leading to increased collagen expression at the papillary dermis.
Collapse
Affiliation(s)
- Shunsuke Iriyama
- Shiseido Global Innovation Center, 1-2-11, Takashima, Nishi-ku, Yokohama, 220-0011, Japan.
| | - Yuki Ogura
- Shiseido Global Innovation Center, 1-2-11, Takashima, Nishi-ku, Yokohama, 220-0011, Japan
| | - Saori Nishikawa
- Shiseido Global Innovation Center, 1-2-11, Takashima, Nishi-ku, Yokohama, 220-0011, Japan
| | - Junichi Hosoi
- Shiseido Global Innovation Center, 1-2-11, Takashima, Nishi-ku, Yokohama, 220-0011, Japan
| | - Satoshi Amano
- Shiseido Global Innovation Center, 1-2-11, Takashima, Nishi-ku, Yokohama, 220-0011, Japan
| |
Collapse
|
6
|
Pavez Loriè E, Baatout S, Choukér A, Buchheim JI, Baselet B, Dello Russo C, Wotring V, Monici M, Morbidelli L, Gagliardi D, Stingl JC, Surdo L, Yip VLM. The Future of Personalized Medicine in Space: From Observations to Countermeasures. Front Bioeng Biotechnol 2021; 9:739747. [PMID: 34966726 PMCID: PMC8710508 DOI: 10.3389/fbioe.2021.739747] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 11/19/2021] [Indexed: 12/12/2022] Open
Abstract
The aim of personalized medicine is to detach from a “one-size fits all approach” and improve patient health by individualization to achieve the best outcomes in disease prevention, diagnosis and treatment. Technological advances in sequencing, improved knowledge of omics, integration with bioinformatics and new in vitro testing formats, have enabled personalized medicine to become a reality. Individual variation in response to environmental factors can affect susceptibility to disease and response to treatments. Space travel exposes humans to environmental stressors that lead to physiological adaptations, from altered cell behavior to abnormal tissue responses, including immune system impairment. In the context of human space flight research, human health studies have shown a significant inter-individual variability in response to space analogue conditions. A substantial degree of variability has been noticed in response to medications (from both an efficacy and toxicity perspective) as well as in susceptibility to damage from radiation exposure and in physiological changes such as loss of bone mineral density and muscle mass in response to deconditioning. At present, personalized medicine for astronauts is limited. With the advent of longer duration missions beyond low Earth orbit, it is imperative that space agencies adopt a personalized strategy for each astronaut, starting from pre-emptive personalized pre-clinical approaches through to individualized countermeasures to minimize harmful physiological changes and find targeted treatment for disease. Advances in space medicine can also be translated to terrestrial applications, and vice versa. This review places the astronaut at the center of personalized medicine, will appraise existing evidence and future preclinical tools as well as clinical, ethical and legal considerations for future space travel.
Collapse
Affiliation(s)
| | - Sarah Baatout
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium.,Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Alexander Choukér
- Laboratory of Translational Research "Stress and Immunity", Department of Anesthesiology, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Judith-Irina Buchheim
- Laboratory of Translational Research "Stress and Immunity", Department of Anesthesiology, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Bjorn Baselet
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Cinzia Dello Russo
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica Del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,MRC Centre for Drug Safety Science and Wolfson Centre for Personalized Medicine, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, United Kingdom
| | | | - Monica Monici
- ASA Campus Joint Laboratory, ASA Research Division, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | | | - Dimitri Gagliardi
- Manchester Institute of Innovation Research, Alliance Manchester Business School, The University of Manchester, Manchester, United Kingdom
| | - Julia Caroline Stingl
- Institute of Clinical Pharmacology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Leonardo Surdo
- Space Applications Services NV/SA for the European Space Agency, Noordwijk, Netherlands
| | - Vincent Lai Ming Yip
- MRC Centre for Drug Safety Science and Wolfson Centre for Personalized Medicine, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
7
|
Marinova IN, Wandall HH, Dabelsteen S. Protocol for CRISPR-Cas9 modification of glycosylation in 3D organotypic skin models. STAR Protoc 2021; 2:100668. [PMID: 34485933 PMCID: PMC8403582 DOI: 10.1016/j.xpro.2021.100668] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Glycosylation is one of the most common protein modifications in living organisms and has important regulatory roles in animal tissue development and homeostasis. Here, we present a protocol for generation of 3D organotypic skin models using CRISPR-Cas9 genetically engineered human keratinocytes (N/TERT-1) to study the role of glycans in epithelial tissue formation. This strategy is also applicable to other gene targets and organotypic tissue models. Careful handling of the cell cultures is critical for the successful formation of the organoids. For complete details on the use and execution of this protocol, please refer to Dabelsteen et al. (2020). CRISPR-Cas9 gene targeting to generate a library of 3D organotypic skin tissues Approach can be used to study the role of glycans in epithelial tissue formation Strategy applicable to other targets and organotypic tissue models
Collapse
Affiliation(s)
- Irina N Marinova
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Hans H Wandall
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Sally Dabelsteen
- Department of Oral Pathology, School of Dentistry, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Montero A, Quílez C, Valencia L, Girón P, Jorcano JL, Velasco D. Effect of Fibrin Concentration on the In Vitro Production of Dermo-Epidermal Equivalents. Int J Mol Sci 2021; 22:ijms22136746. [PMID: 34201667 PMCID: PMC8269027 DOI: 10.3390/ijms22136746] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/10/2021] [Accepted: 06/17/2021] [Indexed: 01/18/2023] Open
Abstract
Human plasma-derived bilayered skin substitutes were successfully used by our group to produce human-based in vitro skin models for toxicity, cosmetic, and pharmaceutical testing. However, mechanical weakness, which causes the plasma-derived fibrin matrices to contract significantly, led us to attempt to improve their stability. In this work, we studied whether an increase in fibrin concentration from 1.2 to 2.4 mg/mL (which is the useful fibrinogen concentration range that can be obtained from plasma) improves the matrix and, hence, the performance of the in vitro skin cultures. The results show that this increase in fibrin concentration indeed affected the mechanical properties by doubling the elastic moduli and the maximum load. A structural analysis indicated a decreased porosity for the 2.4 mg/mL hydrogels, which can help explain this mechanical behavior. The contraction was clearly reduced for the 2.4 mg/mL matrices, which also allowed for the growth and proliferation of primary fibroblasts and keratinocytes, although at a somewhat reduced rate compared to the 1.2 mg/mL gels. Finally, both concentrations of fibrin gave rise to organotypic skin cultures with a fully differentiated epidermis, although their lifespans were longer (25–35%) in cultures with more concentrated matrices, which improves their usefulness. These systems will allow the generation of much better in vitro skin models for the testing of drugs, cosmetics and chemicals, or even to “personalized” skin for the diagnosis or determination of the most effective treatment possible.
Collapse
Affiliation(s)
- Andrés Montero
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid (UC3M), 28903 Madrid, Spain; (A.M.); (C.Q.); (L.V.); (P.G.)
| | - Cristina Quílez
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid (UC3M), 28903 Madrid, Spain; (A.M.); (C.Q.); (L.V.); (P.G.)
| | - Leticia Valencia
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid (UC3M), 28903 Madrid, Spain; (A.M.); (C.Q.); (L.V.); (P.G.)
| | - Paula Girón
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid (UC3M), 28903 Madrid, Spain; (A.M.); (C.Q.); (L.V.); (P.G.)
| | - José Luis Jorcano
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid (UC3M), 28903 Madrid, Spain; (A.M.); (C.Q.); (L.V.); (P.G.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Correspondence: (J.L.J.); (D.V.)
| | - Diego Velasco
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid (UC3M), 28903 Madrid, Spain; (A.M.); (C.Q.); (L.V.); (P.G.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Correspondence: (J.L.J.); (D.V.)
| |
Collapse
|
9
|
Ren G, Zheng X, Sharma V, Letson J, Nestor-Kalinoski AL, Furuta S. Loss of Nitric Oxide Induces Fibrogenic Response in Organotypic 3D Co-Culture of Mammary Epithelia and Fibroblasts-An Indicator for Breast Carcinogenesis. Cancers (Basel) 2021; 13:cancers13112815. [PMID: 34198735 PMCID: PMC8201212 DOI: 10.3390/cancers13112815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Fibrosis, which is often caused by chronic diseases and environmental substances, is closely associated with cancer. Thus, the development of a robust method allowing for deep studies of the linkage between fibrosis and cancer is essential. Here, we tested whether our novel three-dimensional (3D) co-culture of breast epithelia and fibroblasts would be a suitable model for that purpose. We compared the phenotypic effects of L-NAME, an inhibitor of nitric oxide (NO) production, on 3D mono- and co-cultures. We previously reported that prolonged NO depletion with L-NAME caused fibrosis and tumorigenesis in mouse mammary glands. Such in vivo effects of L-NAME were well recapitulated in 3D co-cultures, but not in 3D mono-cultures of epithelia and fibroblasts. These results support not only the essential roles of the presence of the stroma in cancer development, but also the utility of this co-culture in studying the causal relationship between fibrosis and cancer. Abstract Excessive myofibroblast activation, which leads to dysregulated collagen deposition and the stiffening of the extracellular matrix (ECM), plays pivotal roles in cancer initiation and progression. Cumulative evidence attests to the cancer-causing effects of a number of fibrogenic factors found in the environment, diseases and drugs. While identifying such factors largely depends on epidemiological studies, it would be of great importance to develop a robust in vitro method to demonstrate the causal relationship between fibrosis and cancer. Here, we tested whether our recently developed organotypic three-dimensional (3D) co-culture would be suitable for that purpose. This co-culture system utilizes the discontinuous ECM to separately culture mammary epithelia and fibroblasts in the discrete matrices to model the complexity of the mammary gland. We observed that pharmaceutical deprivation of nitric oxide (NO) in 3D co-cultures induced myofibroblast differentiation of the stroma as well as the occurrence of epithelial–mesenchymal transition (EMT) of the parenchyma. Such in vitro response to NO deprivation was unique to co-cultures and closely mimicked the phenotype of NO-depleted mammary glands exhibiting stromal desmoplasia and precancerous lesions undergoing EMT. These results suggest that this novel 3D co-culture system could be utilized in the deep mechanistic studies of the linkage between fibrosis and cancer.
Collapse
Affiliation(s)
- Gang Ren
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
| | - Xunzhen Zheng
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
| | - Vandana Sharma
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
| | - Joshua Letson
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
| | - Andrea L. Nestor-Kalinoski
- Department of Surgery, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA;
| | - Saori Furuta
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
- Correspondence:
| |
Collapse
|
10
|
Yang R, Li G, Zhuang C, Yu P, Ye T, Zhang Y, Shang P, Huang J, Cai M, Wang L, Cui W, Deng L. Gradient bimetallic ion-based hydrogels for tissue microstructure reconstruction of tendon-to-bone insertion. SCIENCE ADVANCES 2021; 7:eabg3816. [PMID: 34162547 PMCID: PMC8221628 DOI: 10.1126/sciadv.abg3816] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 04/29/2021] [Indexed: 05/11/2023]
Abstract
Although gradients play an essential role in guiding the function of tissues, achieving synchronous regeneration of gradient tissue injuries remains a challenge. Here, a gradient bimetallic (Cu and Zn) ion-based hydrogel was first constructed via the one-step coordinative crosslinking of sulfhydryl groups with copper and zinc ions for the microstructure reconstruction of the tendon-to-bone insertion. In this bimetallic hydrogel system, zinc and copper ions could not only act as crosslinkers but also provide strong antibacterial effects and induce regenerative capacity in vitro. The capability of hydrogels in simultaneously promoting tenogenesis and osteogenesis was further verified in a rat rotator cuff tear model. It was found that the Cu/Zn gradient layer could induce considerable collagen and fibrocartilage arrangement and ingrowth at the tendon-to-bone interface. Overall, the gradient bimetallic ion-based hydrogel ensures accessibility and provides opportunities to regenerate inhomogeneous tissue with physiological complexity or interface tissue.
Collapse
Affiliation(s)
- Renhao Yang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Gen Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Chengyu Zhuang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Pei Yu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Tingjun Ye
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Yin Zhang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Peiyang Shang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Jingjing Huang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Ming Cai
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301 Middle Yanchang Road, Shanghai 200072, P. R. China
| | - Lei Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China.
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China.
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China.
| |
Collapse
|
11
|
Kurinna S, Seltmann K, Bachmann AL, Schwendimann A, Thiagarajan L, Hennig P, Beer HD, Mollo MR, Missero C, Werner S. Interaction of the NRF2 and p63 transcription factors promotes keratinocyte proliferation in the epidermis. Nucleic Acids Res 2021; 49:3748-3763. [PMID: 33764436 PMCID: PMC8053124 DOI: 10.1093/nar/gkab167] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/27/2021] [Accepted: 03/03/2021] [Indexed: 12/22/2022] Open
Abstract
Epigenetic regulation of cell and tissue function requires the coordinated action of transcription factors. However, their combinatorial activities during regeneration remain largely unexplored. Here, we discover an unexpected interaction between the cytoprotective transcription factor NRF2 and p63- a key player in epithelial morphogenesis. Chromatin immunoprecipitation combined with sequencing and reporter assays identifies enhancers and promoters that are simultaneously activated by NRF2 and p63 in human keratinocytes. Modeling of p63 and NRF2 binding to nucleosomal DNA suggests their chromatin-assisted interaction. Pharmacological and genetic activation of NRF2 increases NRF2–p63 binding to enhancers and promotes keratinocyte proliferation, which involves the common NRF2–p63 target cyclin-dependent kinase 12. These results unravel a collaborative function of NRF2 and p63 in the control of epidermal renewal and suggest their combined activation as a strategy to promote repair of human skin and other stratified epithelia.
Collapse
Affiliation(s)
- Svitlana Kurinna
- Division of Cell Matrix Biology and Regenerative Medicine, FBMH, University of Manchester, M13 9PT, United Kingdom
| | - Kristin Seltmann
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Andreas L Bachmann
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Andreas Schwendimann
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Lalitha Thiagarajan
- Division of Cell Matrix Biology and Regenerative Medicine, FBMH, University of Manchester, M13 9PT, United Kingdom
| | - Paulina Hennig
- Department of Dermatology, University Hospital Zurich, 8006 Zurich, Switzerland
| | - Hans-Dietmar Beer
- Department of Dermatology, University Hospital Zurich, 8006 Zurich, Switzerland
| | - Maria Rosaria Mollo
- CEINGE Biotecnologie Avanzate, Naples, Italy, University of Naples Federico II, 80131 Naples, Italy
| | - Caterina Missero
- CEINGE Biotecnologie Avanzate, Naples, Italy, University of Naples Federico II, 80131 Naples, Italy
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
12
|
Ramasamy S, Davoodi P, Vijayavenkataraman S, Teoh JH, Thamizhchelvan AM, Robinson KS, Wu B, Fuh JY, DiColandrea T, Zhao H, Lane EB, Wang CH. Optimized construction of a full thickness human skin equivalent using 3D bioprinting and a PCL/collagen dermal scaffold. ACTA ACUST UNITED AC 2021. [DOI: 10.1016/j.bprint.2020.e00123] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
13
|
Besnard M, Padonou F, Provin N, Giraud M, Guillonneau C. AIRE deficiency, from preclinical models to human APECED disease. Dis Model Mech 2021; 14:dmm046359. [PMID: 33729987 PMCID: PMC7875492 DOI: 10.1242/dmm.046359] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (APECED) is a rare life-threatening autoimmune disease that attacks multiple organs and has its onset in childhood. It is an inherited condition caused by a variety of mutations in the autoimmune regulator (AIRE) gene that encodes a protein whose function has been uncovered by the generation and study of Aire-KO mice. These provided invaluable insights into the link between AIRE expression in medullary thymic epithelial cells (mTECs), and the broad spectrum of self-antigens that these cells express and present to the developing thymocytes. However, these murine models poorly recapitulate all phenotypic aspects of human APECED. Unlike Aire-KO mice, the recently generated Aire-KO rat model presents visual features, organ lymphocytic infiltrations and production of autoantibodies that resemble those observed in APECED patients, making the rat model a main research asset. In addition, ex vivo models of AIRE-dependent self-antigen expression in primary mTECs have been successfully set up. Thymus organoids based on pluripotent stem cell-derived TECs from APECED patients are also emerging, and constitute a promising tool to engineer AIRE-corrected mTECs and restore the generation of regulatory T cells. Eventually, these new models will undoubtedly lead to main advances in the identification and assessment of specific and efficient new therapeutic strategies aiming to restore immunological tolerance in APECED patients.
Collapse
Affiliation(s)
- Marine Besnard
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| | - Francine Padonou
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| | - Nathan Provin
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| | - Matthieu Giraud
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| | - Carole Guillonneau
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| |
Collapse
|
14
|
Gronbach L, Jurmeister P, Schäfer-Korting M, Keilholz U, Tinhofer I, Zoschke C. Primary Extracellular Matrix Enables Long-Term Cultivation of Human Tumor Oral Mucosa Models. Front Bioeng Biotechnol 2020; 8:579896. [PMID: 33344431 PMCID: PMC7746540 DOI: 10.3389/fbioe.2020.579896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 11/10/2020] [Indexed: 11/18/2022] Open
Abstract
3D tumor models clearly outperform 2D cell cultures in recapitulating tissue architecture and drug response. However, their potential in understanding treatment efficacy and resistance development should be better exploited if also long-term effects of treatment could be assessed in vitro. The main disadvantages of the matrices commonly used for in vitro culture are their limited cultivation time and the low comparability with patient-specific matrix properties. Extended cultivation periods are feasible when primary human cells produce the extracellular matrix in situ. Herein, we adapted the hyalograft-3D approach from reconstructed human skin to normal and tumor oral mucosa models and compared the results to bovine collagen-based models. The hyalograft models showed similar morphology and cell proliferation after 7 weeks compared to collagen-based models after 2 weeks of cultivation. Tumor thickness and VEGF expression increased in hyalograft-based tumor models, whereas expression of laminin-332, tenascin C, and hypoxia-inducible factor 1α was lower than in collagen-based models. Taken together, the in situ produced extracellular matrix better confined tumor invasion in the first part of the cultivation period, with continuous tumor proliferation and increasing invasion later on. This proof-of-concept study showed the successful transfer of the hyalograft approach to tumor oral mucosa models and lays the foundation for the assessment of long-term drug treatment effects. Moreover, the use of an animal-derived extracellular matrix is avoided.
Collapse
Affiliation(s)
- Leonie Gronbach
- Institute of Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Berlin, Germany
| | - Philipp Jurmeister
- Institute of Pathology, Berlin Institute of Health, Humboldt-Universität zu Berlin, Corporate Member of Freie Universität Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Heidelberg and German Cancer Consortum Partner Site Berlin, German Cancer Research Center, Berlin, Germany
| | - Monika Schäfer-Korting
- Institute of Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Berlin, Germany
| | - Ulrich Keilholz
- Comprehensive Cancer Center, Berlin Institute of Health, Humboldt-Universität zu Berlin, Corporate Member of Freie Universität Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ingeborg Tinhofer
- Heidelberg and German Cancer Consortum Partner Site Berlin, German Cancer Research Center, Berlin, Germany.,Department of Radiooncology and Radiotherapy, Berlin Institute of Health, Humboldt-Universität zu Berlin, Corporate Member of Freie Universität Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Zoschke
- Institute of Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
15
|
Organotypic Co-Cultures as a Novel 3D Model for Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12082330. [PMID: 32824777 PMCID: PMC7463661 DOI: 10.3390/cancers12082330] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/09/2020] [Accepted: 08/14/2020] [Indexed: 01/09/2023] Open
Abstract
Background: Head and neck squamous cell carcinomas (HNSCC) are phenotypically and molecularly heterogeneous and frequently develop therapy resistance. Reliable patient-derived 3D tumor models are urgently needed to further study the complex pathogenesis of these tumors and to overcome treatment failure. Methods: We developed a three-dimensional organotypic co-culture (3D-OTC) model for HNSCC that maintains the architecture and cell composition of the individual tumor. A dermal equivalent (DE), composed of healthy human-derived fibroblasts and viscose fibers, served as a scaffold for the patient sample. DEs were co-cultivated with 13 vital HNSCC explants (non-human papillomavirus (HPV) driven, n = 7; HPV-driven, n = 6). Fractionated irradiation was applied to 5 samples (non-HPV-driven, n = 2; HPV-driven n = 3). To evaluate expression of ki-67, cleaved caspase-3, pan-cytokeratin, p16INK4a, CD45, ∝smooth muscle actin and vimentin over time, immunohistochemistry and immunofluorescence staining were performed Patient checkup data were collected for up to 32 months after first diagnosis. Results: All non-HPV-driven 3D-OTCs encompassed proliferative cancer cells during cultivation for up to 21 days. Proliferation indices of primaries and 3D-OTCs were comparable and consistent over time. Overall, tumor explants displayed heterogeneous growth patterns (i.e., invasive, expansive, silent). Cancer-associated fibroblasts and leukocytes could be detected for up to 21 days. HPV DNA was detectable in both primary and 3D-OTCs (day 14) of HPV-driven tumors. However, p16INK4a expression levels were varying. Morphological alterations and radioresistant tumor cells were detected in 3D-OTC after fractionated irradiation in HPV-driven and non-driven samples. Conclusions: Our 3D-OTC model for HNSCC supports cancer cell survival and proliferation in their original microenvironment. The model enables investigation of invasive cancer growth and might, in the future, serve as a platform to perform sensitivity testing upon treatment to predict therapy response.
Collapse
|
16
|
|
17
|
Ren G, Sharma V, Letson J, Walia Y, Fernando V, Furuta S. Reconstituting Breast Tissue with Organotypic Three-dimensional Co-culture of Epithelial and Stromal Cells in Discontinuous Extracellular Matrices. Bio Protoc 2019; 9:e3392. [PMID: 33654884 DOI: 10.21769/bioprotoc.3392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/29/2019] [Accepted: 09/11/2019] [Indexed: 11/02/2022] Open
Abstract
Co-culture systems utilizing reconstituted or synthetic extracellular matrix (ECM) and micropatterning techniques have enabled the reconstruction of surface epithelial tissues. This technique has been utilized in the regeneration, disease modeling and drug screening of the surface epithelia, such as the skin and esophagus. On the other hand, the reconstruction of glandular epithelia would require more intricate ECM organizations. Here we describe a protocol for a novel three-dimensional organotypic co-culture system for the reconstruction of mammary glands that utilizes the discontinuous ECM. In this technique, primary mammary fibroblasts first establish a layer of the connective tissue rich in collagen I. Then, mammary epithelial cells form acinar structures, the functional glandular units, within the laminin-rich basement membrane embedded in the connective tissue. This method allows for the regeneration of the in vivo-like architecture of mammary glands and could be utilized for monitoring the real-time response of mammary glands to drug treatment.
Collapse
Affiliation(s)
- Gang Ren
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
| | - Vandana Sharma
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
| | - Joshua Letson
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
| | - Yashna Walia
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
| | - Veani Fernando
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
| | - Saori Furuta
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
| |
Collapse
|
18
|
De Angelis B, D'Autilio MFLM, Orlandi F, Pepe G, Garcovich S, Scioli MG, Orlandi A, Cervelli V, Gentile P. Wound Healing: In Vitro and In Vivo Evaluation of a Bio-Functionalized Scaffold Based on Hyaluronic Acid and Platelet-Rich Plasma in Chronic Ulcers. J Clin Med 2019; 8:jcm8091486. [PMID: 31540446 PMCID: PMC6780765 DOI: 10.3390/jcm8091486] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 12/20/2022] Open
Abstract
Chronic ulcers are characterized by loss of substance without a normal tendency towards spontaneous healing. The Wound Bed Preparation Guideline advises that after diagnosis, the expert should correct the biological state of the ulcer micro-environment based on TIME principles (Tissue, Infection, Moisture balance, Epidermal). There are many ways to treat such ulcers, for example through use of advanced dressings, negative pressure, surgical toilets, dermal substitutes, autologous skin grafting, and free or local flaps. In vitro and in vivo pre-clinical models hold widely acknowledged potential yet complex limitations. Tissue bioengineering could be an ideal approach to foster innovative strategies in wound healing. Our observational study reports on an in vitro and in vivo evaluation of a bio-functionalized scaffold composed of platelet-rich plasma (PRP) and hyaluronic acid (HA) used in 182 patients affected by chronic ulcers (diabetic and vascular), comparing the results with a control group of 182 patients treated with traditional dressings (HA alone). After 30 days the patients who had undergone the combined treatment (PRP + HA), showed 96.8% ± 1.5% re-epithelialization, as compared to 78.4% ± 4.4% in the control group (HA only). Within 80 days, they had 98.4% ± 1.3% re-epithelialization as compared to 87.8% ± 4.1% in the control group (HA only; p < 0.05). No local recurrence was observed during the follow-up period. PRP + HA treatment showed stronger regenerative potential in terms of epidermal proliferation and dermal renewal compared with HA alone.
Collapse
Affiliation(s)
- Barbara De Angelis
- Department of Surgical Science, University of Rome Tor Vergata, Rome 00133, Italy.
| | | | - Fabrizio Orlandi
- Department of Surgical Science, University of Rome Tor Vergata, Rome 00133, Italy.
| | - Giampiero Pepe
- Department of Surgical Science, University of Rome Tor Vergata, Rome 00133, Italy.
| | - Simone Garcovich
- Institute of Dermatology, F. Policlinico Gemelli IRCSS, Università Cattolica del Sacro Cuore, Rome 00168, Italy.
| | - Maria Giovanna Scioli
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome 00133, Italy.
| | - Augusto Orlandi
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome 00133, Italy.
| | - Valerio Cervelli
- Department of Surgical Science, University of Rome Tor Vergata, Rome 00133, Italy.
| | - Pietro Gentile
- Department of Surgical Science, University of Rome Tor Vergata, Rome 00133, Italy.
| |
Collapse
|
19
|
Ultrastructural morphology is distinct among primary progenitor cell isolates from normal, inflamed, and cryopreserved equine hoof tissue and CD105 +K14 + progenitor cells. In Vitro Cell Dev Biol Anim 2019; 55:641-655. [PMID: 31297697 PMCID: PMC6717190 DOI: 10.1007/s11626-019-00380-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/17/2019] [Indexed: 12/21/2022]
Abstract
The equine hoof dermal-epidermal interface requires progenitor cells with distinct characteristics. This study was designed to provide accurate ultrastructural depictions of progenitor cells isolated from inflamed tissue and normal tissue before and after cryopreservation and following selection of cells expressing both keratin (K) 14 (ectodermal) and cluster of differentiation (CD) 105 (mesodermal). Passage 3 cell ultrastructure was assessed following 2D culture and after 3D culture on decellularized hoof tissue scaffolds. Outcome measures included light, transmission electron, and scanning electron microscopy, immunocytochemistry, and CD105+K14+ cell trilineage plasticity. Cells from normal tissue had typical progenitor cell characteristics. Those from inflamed tissue had organelles and morphology consistent with catabolic activities including lysosomes, irregular rough endoplasmic reticulum, and fewer vacuoles and early endosomes than those from normal tissue. Cryopreserved tissue cells appeared apoptotic with an irregular cell membrane covered by cytoplasmic protrusions closely associated with endocytic and exocytic vesicles, chromatin aggregated on the nuclear envelop, abundant, poorly organized rough endoplasmic reticulum, and plentiful lysosomes. Cells that were CD105+K14+ were distinguishable from heterogenous cells by infrequent microvilli on the cell surface, sparse endosomes and vesicles, and desmosomes between cells. Cells expressed ectodermal (K15) and mesodermal (CD105) proteins in 2D and 3D cultures. Inflamed and cryopreserved tissue isolates attached poorly to tissue scaffold while normal tissue cells attached well, but only CD105+K14+ cells produced extracellular matrix after 4 d. The CD105+K14+ cells exhibited osteoblastic, adipocytic, and neurocytic differentiation. Ultrastructural information provided by this study contributes to understanding of equine hoof progenitor cells to predict their potential contributions to tissue maintenance, healing, and damage as well post-implantation behavior.
Collapse
|
20
|
Suhail S, Sardashti N, Jaiswal D, Rudraiah S, Misra M, Kumbar SG. Engineered Skin Tissue Equivalents for Product Evaluation and Therapeutic Applications. Biotechnol J 2019; 14:e1900022. [PMID: 30977574 PMCID: PMC6615970 DOI: 10.1002/biot.201900022] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 03/20/2019] [Indexed: 12/12/2022]
Abstract
The current status of skin tissue equivalents that have emerged as relevant tools in commercial and therapeutic product development applications is reviewed. Due to the rise of animal welfare concerns, numerous companies have designed skin model alternatives to assess the efficacy of pharmaceutical, skincare, and cosmetic products in an in vitro setting, decreasing the dependency on such methods. Skin models have also made an impact in determining the root causes of skin diseases. When designing a skin model, there are various chemical and physical considerations that need to be considered to produce a biomimetic design. This includes designing a structure that mimics the structural characteristics and mechanical strength needed for tribological property measurement and toxicological testing. Recently, various commercial products have made significant progress towards achieving a native skin alternative. Further research involve the development of a functional bilayered model that mimics the constituent properties of the native epidermis and dermis. In this article, the skin models are divided into three categories: in vitro epidermal skin equivalents, in vitro full-thickness skin equivalents, and clinical skin equivalents. A description of skin model characteristics, testing methods, applications, and potential improvements is presented.
Collapse
Affiliation(s)
- Sana Suhail
- Department of Orthopaedic Surgery, University of Connecticut Health, 263 Farmington Ave., Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT 06269, USA
| | - Naseem Sardashti
- Department of Orthopaedic Surgery, University of Connecticut Health, 263 Farmington Ave., Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT 06269, USA
| | - Devina Jaiswal
- Department of Orthopaedic Surgery, University of Connecticut Health, 263 Farmington Ave., Farmington, CT 06030, USA
- Department of Biomedical Engineering, Western New England University, 1215 Wilbrahan Road, Springfield, MA 01119
| | - Swetha Rudraiah
- Department of Orthopaedic Surgery, University of Connecticut Health, 263 Farmington Ave., Farmington, CT 06030, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Saint Joseph, 229 Trumbull St., Hartford CT 06103, USA
| | - Manoj Misra
- Unilever R&D, 40 Merritt Blvd, Trumbull, CT 06611, USA
| | - Sangamesh G. Kumbar
- Department of Orthopaedic Surgery, University of Connecticut Health, 263 Farmington Ave., Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT 06269, USA
| |
Collapse
|
21
|
|
22
|
The renaissance of human skin organ culture: A critical reappraisal. Differentiation 2018; 104:22-35. [DOI: 10.1016/j.diff.2018.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 10/03/2018] [Accepted: 10/15/2018] [Indexed: 02/07/2023]
|
23
|
Yan WC, Davoodi P, Vijayavenkataraman S, Tian Y, Ng WC, Fuh JY, Robinson KS, Wang CH. 3D bioprinting of skin tissue: From pre-processing to final product evaluation. Adv Drug Deliv Rev 2018; 132:270-295. [PMID: 30055210 DOI: 10.1016/j.addr.2018.07.016] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 07/17/2018] [Accepted: 07/20/2018] [Indexed: 02/07/2023]
Abstract
Bioprinted skin tissue has the potential for aiding drug screening, formulation development, clinical transplantation, chemical and cosmetic testing, as well as basic research. Limitations of conventional skin tissue engineering approaches have driven the development of biomimetic skin equivalent via 3D bioprinting. A key hope for bioprinting skin is the improved tissue authenticity over conventional skin equivalent construction, enabling the precise localization of multiple cell types and appendages within a construct. The printing of skin faces challenges broadly associated with general 3D bioprinting, including the selection of cell types and biomaterials, and additionally requires in vitro culture formats that allow for growth at an air-liquid interface. This paper provides a thorough review of current 3D bioprinting technologies used to engineer human skin constructs and presents the overall pipelines of designing a biomimetic artificial skin via 3D bioprinting from the design phase (i.e. pre-processing phase) through the tissue maturation phase (i.e. post-processing) and into final product evaluation for drug screening, development, and drug delivery applications.
Collapse
|
24
|
Chermnykh E, Kalabusheva E, Vorotelyak E. Extracellular Matrix as a Regulator of Epidermal Stem Cell Fate. Int J Mol Sci 2018; 19:ijms19041003. [PMID: 29584689 PMCID: PMC5979429 DOI: 10.3390/ijms19041003] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/15/2018] [Accepted: 03/21/2018] [Indexed: 12/17/2022] Open
Abstract
Epidermal stem cells reside within the specific anatomic location, called niche, which is a microenvironment that interacts with stem cells to regulate their fate. Regulation of many important processes, including maintenance of stem cell quiescence, self-renewal, and homeostasis, as well as the regulation of division and differentiation, are common functions of the stem cell niche. As it was shown in multiple studies, extracellular matrix (ECM) contributes a lot to stem cell niches in various tissues, including that of skin. In epidermis, ECM is represented, primarily, by a highly specialized ECM structure, basement membrane (BM), which separates the epidermal and dermal compartments. Epidermal stem cells contact with BM, but when they lose the contact and migrate to the overlying layers, they undergo terminal differentiation. When considering all of these factors, ECM is of fundamental importance in regulating epidermal stem cells maintenance, proper mobilization, and differentiation. Here, we summarize the remarkable progress that has recently been made in the research of ECM role in regulating epidermal stem cell fate, paying special attention to the hair follicle stem cell niche. We show that the destruction of ECM components impairs epidermal stem cell morphogenesis and homeostasis. A deep understanding of ECM molecular structure as well as the development of in vitro system for stem cell maintaining by ECM proteins may bring us to developing new approaches for regenerative medicine.
Collapse
Affiliation(s)
- Elina Chermnykh
- Koltzov Institute of Developmental Biology Russian Academy of Sciences, Moscow 119334, Russia.
- Department of Regenerative Medicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia.
| | - Ekaterina Kalabusheva
- Koltzov Institute of Developmental Biology Russian Academy of Sciences, Moscow 119334, Russia.
- Department of Regenerative Medicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia.
| | - Ekaterina Vorotelyak
- Koltzov Institute of Developmental Biology Russian Academy of Sciences, Moscow 119334, Russia.
- Department of Regenerative Medicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia.
- Faculty of Biology, Lomonosov Moscow State University, Moscow 119991, Russia.
| |
Collapse
|
25
|
Mieremet A, Rietveld M, van Dijk R, Bouwstra JA, El Ghalbzouri A. Recapitulation of Native Dermal Tissue in a Full-Thickness Human Skin Model Using Human Collagens. Tissue Eng Part A 2017; 24:873-881. [PMID: 29130419 DOI: 10.1089/ten.tea.2017.0326] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Full-thickness skin models comprise a three-dimensional dermal equivalent based on an animal-derived collagen matrix that harbors fibroblasts and an epidermal equivalent formed by keratinocytes. The functionality of both equivalents is influenced by many factors, including extracellular matrix composition and resident cell type. Animal-derived collagens differ in amino acid composition and physicochemical properties from human collagens. This composition could alter the functionality of the dermal equivalent and epidermal morphogenesis with the barrier formation in full-thickness models (FTMs). By replacement of animal-derived collagen for human collagen, we generated and characterized the animal material-free human collagen full-thickness models (hC-FTMs) that better mimic native dermal tissue. MATERIALS AND METHODS An isolation procedure to obtain soluble collagen from human abdominal dermis was developed. Both FTMs and hC-FTMs were generated with primary human fibroblasts and keratinocytes. Immunohistochemical analyses with biomarkers for the dermal matrix composition, basement membrane (BM) formation, epidermal proliferation, differentiation, and activation were performed. The stratum corneum (SC) lipid composition was studied with liquid chromatography-mass spectrometry. Lipid lamellar organization was determined by small-angle X-ray diffraction. RESULTS The FTMs and hC-FTMs exhibit many similarities, including the dermal matrix structure, BM formation, epidermal basal layer proliferation, and execution of differentiation programs. The SC contains a similar number of corneocyte layers and the same level of lipids. The ceramide chain length distribution and ceramide subclass profile showed only minor differences. Subsequently, this led to an unaltered lamellar organization. CONCLUSION The animal material-free hC-FTM is generated successfully using collagens isolated from human abdominal dermis. Utilization of human collagens revealed that (epi-)dermal morphogenesis and lipid barrier formation resembled that of original FTMs. The hC-FTMs contain a dermal equivalent that mimics the native stromal tissue to a higher extent. Therefore these in vitro skin models can be used as promising tool for research purposes that contribute to animal-free experimentation.
Collapse
Affiliation(s)
- Arnout Mieremet
- 1 Department of Dermatology, Leiden University Medical Centre , Leiden, The Netherlands
| | - Marion Rietveld
- 1 Department of Dermatology, Leiden University Medical Centre , Leiden, The Netherlands
| | - Rianne van Dijk
- 2 Division of Drug Delivery Technology, LACDR, Leiden University , Leiden, The Netherlands
| | - Joke A Bouwstra
- 2 Division of Drug Delivery Technology, LACDR, Leiden University , Leiden, The Netherlands
| | | |
Collapse
|
26
|
Planz V, Seif S, Atchison JS, Vukosavljevic B, Sparenberg L, Kroner E, Windbergs M. Three-dimensional hierarchical cultivation of human skin cells on bio-adaptive hybrid fibers. Integr Biol (Camb) 2017; 8:775-84. [PMID: 27241237 DOI: 10.1039/c6ib00080k] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The human skin comprises a complex multi-scale layered structure with hierarchical organization of different cells within the extracellular matrix (ECM). This supportive fiber-reinforced structure provides a dynamically changing microenvironment with specific topographical, mechanical and biochemical cell recognition sites to facilitate cell attachment and proliferation. Current advances in developing artificial matrices for cultivation of human cells concentrate on surface functionalizing of biocompatible materials with different biomolecules like growth factors to enhance cell attachment. However, an often neglected aspect for efficient modulation of cell-matrix interactions is posed by the mechanical characteristics of such artificial matrices. To address this issue, we fabricated biocompatible hybrid fibers simulating the complex biomechanical characteristics of native ECM in human skin. Subsequently, we analyzed interactions of such fibers with human skin cells focusing on the identification of key fiber characteristics for optimized cell-matrix interactions. We successfully identified the mediating effect of bio-adaptive elasto-plastic stiffness paired with hydrophilic surface properties as key factors for cell attachment and proliferation, thus elucidating the synergistic role of these parameters to induce cellular responses. Co-cultivation of fibroblasts and keratinocytes on such fiber mats representing the specific cells in dermis and epidermis resulted in a hierarchical organization of dermal and epidermal tissue layers. In addition, terminal differentiation of keratinocytes at the air interface was observed. These findings provide valuable new insights into cell behaviour in three-dimensional structures and cell-material interactions which can be used for rational development of bio-inspired functional materials for advanced biomedical applications.
Collapse
Affiliation(s)
- Viktoria Planz
- Department of Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus Building A 4.1, 66123 Saarbrücken, Germany.
| | - Salem Seif
- Department of Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus Building A 4.1, 66123 Saarbrücken, Germany. and PharmBioTec GmbH, Science Park 1, 66123 Saarbrücken, Germany
| | - Jennifer S Atchison
- INM - Leibniz Institute for New Materials, Campus Building D 2.2, 66123 Saarbrücken, Germany
| | - Branko Vukosavljevic
- Helmholtz Centre for Infection Research (HZI) and Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Department of Drug Delivery (DDEL), Campus Building E 8.1, 66123 Saarbrücken, Germany
| | - Lisa Sparenberg
- Department of Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus Building A 4.1, 66123 Saarbrücken, Germany.
| | - Elmar Kroner
- INM - Leibniz Institute for New Materials, Campus Building D 2.2, 66123 Saarbrücken, Germany
| | - Maike Windbergs
- Department of Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus Building A 4.1, 66123 Saarbrücken, Germany. and PharmBioTec GmbH, Science Park 1, 66123 Saarbrücken, Germany and Helmholtz Centre for Infection Research (HZI) and Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Department of Drug Delivery (DDEL), Campus Building E 8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
27
|
Larribère L, Galach M, Novak D, Arévalo K, Volz HC, Stark HJ, Boukamp P, Boutros M, Utikal J. An RNAi Screen Reveals an Essential Role for HIPK4 in Human Skin Epithelial Differentiation from iPSCs. Stem Cell Reports 2017; 9:1234-1245. [PMID: 28966120 PMCID: PMC5639458 DOI: 10.1016/j.stemcr.2017.08.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 12/31/2022] Open
Abstract
Molecular mechanisms responsible for the development of human skin epithelial cells are incompletely understood. As a consequence, the efficiency to establish a pure skin epithelial cell population from human induced pluripotent stem cells (hiPSCs) remains poor. Using an approach including RNAi and high-throughput imaging of early epithelial cells, we identified candidate kinases involved in their differentiation from hiPSCs. Among these, we found HIPK4 to be an important inhibitor of this process. Indeed, its silencing increased the amount of generated skin epithelial precursors at an early time point, increased the amount of generated keratinocytes at a later time point, and improved growth and differentiation of organotypic cultures, allowing for the formation of a denser basal layer and stratification with the expression of several keratins. Our data bring substantial input regarding regulation of human skin epithelial differentiation and for improving differentiation protocols from pluripotent stem cells. High-throughput RNAi screen setup during human skin epithelial differentiation Identification of HIPK4 as a crucial blocker of human skin epithelial differentiation Improvement of human organotypic epithelial cultures after HIPK4 silencing
Collapse
Affiliation(s)
- Lionel Larribère
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69121 Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany.
| | - Marta Galach
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69121 Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Daniel Novak
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69121 Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Karla Arévalo
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69121 Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Hans Christian Volz
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Department of Cell and Molecular Biology, Heidelberg University, 69120 Heidelberg, Germany; Department of Cardiology, Heidelberg University, 69120 Heidelberg, Germany
| | - Hans-Jürgen Stark
- Genetics of Skin Carcinogenesis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Petra Boukamp
- Genetics of Skin Carcinogenesis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; IUF-Leibniz Research Institute for Environmental Medicine, 40021 Düsseldorf, Germany
| | - Michael Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Department of Cell and Molecular Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Jochen Utikal
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69121 Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany.
| |
Collapse
|
28
|
Desmet E, Ramadhas A, Lambert J, Van Gele M. In vitro psoriasis models with focus on reconstructed skin models as promising tools in psoriasis research. Exp Biol Med (Maywood) 2017; 242:1158-1169. [PMID: 28585891 DOI: 10.1177/1535370217710637] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Psoriasis is a complex chronic immune-mediated inflammatory cutaneous disease associated with the development of inflammatory plaques on the skin. Studies proved that the disease results from a deregulated interplay between skin keratinocytes, immune cells and the environment leading to a persisting inflammatory process modulated by pro-inflammatory cytokines and activation of T cells. However, a major hindrance to study the pathogenesis of psoriasis more in depth and subsequent development of novel therapies is the lack of suitable pre-clinical models mimicking the complex phenotype of this skin disorder. Recent advances in and optimization of three-dimensional skin equivalent models have made them attractive and promising alternatives to the simplistic monolayer cultures, immunological different in vivo models and scarce ex vivo skin explants. Moreover, human skin equivalents are increasing in complexity level to match human biology as closely as possible. Here, we critically review the different types of three-dimensional skin models of psoriasis with relevance to their application potential and advantages over other models. This will guide researchers in choosing the most suitable psoriasis skin model for therapeutic drug testing (including gene therapy via siRNA molecules), or to examine biological features contributing to the pathology of psoriasis. However, the addition of T cells (as recently applied to a de-epidermized dermis-based psoriatic skin model) or other immune cells would make them even more attractive models and broaden their application potential. Eventually, the ultimate goal would be to substitute animal models by three-dimensional psoriatic skin models in the pre-clinical phases of anti-psoriasis candidate drugs. Impact statement The continuous development of novel in vitro models mimicking the psoriasis phenotype is important in the field of psoriasis research, as currently no model exists that completely matches the in vivo psoriasis skin or the disease pathology. This work provides a complete overview of the different available in vitro psoriasis models and suggests improvements for future models. Moreover, a focus was given to psoriatic skin equivalent models, as they offer several advantages over the other models, including commercial availability and validity. The potential and reported applicability of these models in psoriasis pre-clinical research is extensively discussed. As such, this work offers a guide to researchers in their choice of pre-clinical psoriasis model depending on their type of research question.
Collapse
Affiliation(s)
- Eline Desmet
- Department of Dermatology, Ghent University Hospital, Ghent 9000, Belgium
| | - Anesh Ramadhas
- Department of Dermatology, Ghent University Hospital, Ghent 9000, Belgium
| | - Jo Lambert
- Department of Dermatology, Ghent University Hospital, Ghent 9000, Belgium
| | - Mireille Van Gele
- Department of Dermatology, Ghent University Hospital, Ghent 9000, Belgium
| |
Collapse
|
29
|
Lelièvre SA, Kwok T, Chittiboyina S. Architecture in 3D cell culture: An essential feature for in vitro toxicology. Toxicol In Vitro 2017; 45:287-295. [PMID: 28366709 DOI: 10.1016/j.tiv.2017.03.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/20/2017] [Accepted: 03/28/2017] [Indexed: 01/06/2023]
Abstract
Three-dimensional cell culture has the potential to revolutionize toxicology studies by allowing human-based reproduction of essential elements of organs. Beyond the study of toxicants on the most susceptible organs such as liver, kidney, skin, lung, gastrointestinal tract, testis, heart and brain, carcinogenesis research will also greatly benefit from 3D cell culture models representing any normal tissue. No tissue function can be suitably reproduced without the appropriate tissue architecture whether mimicking acini, ducts or tubes, sheets of cells or more complex cellular organizations like hepatic cords. In this review, we illustrate the fundamental characteristics of polarity that is an essential architectural feature of organs for which different 3D cell culture models are available for toxicology studies in vitro. The value of tissue polarity for the development of more accurate carcinogenesis studies is also exemplified, and the concept of using extracellular gradients of gaseous or chemical substances produced with microfluidics in 3D cell culture is discussed. Indeed such gradients-on-a-chip might bring unprecedented information to better determine permissible exposure levels. Finally, the impact of tissue architecture, established via cell-matrix interactions, on the cell nucleus is emphasized in light of the importance in toxicology of morphological and epigenetic alterations of this organelle.
Collapse
Affiliation(s)
- Sophie A Lelièvre
- Purdue University, Department of Basic Medical Sciences, 625 Harrison Street, West Lafayette, IN 47907, USA; 3D Cell Culture Core (3D3C) Facility, Birck Nanotechnology Center, Purdue University Discovery Park, 1205 West State Street, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, 201 S University Street, West Lafayette, IN 47907, USA.
| | - Tim Kwok
- 3D Cell Culture Core (3D3C) Facility, Birck Nanotechnology Center, Purdue University Discovery Park, 1205 West State Street, West Lafayette, IN 47907, USA
| | - Shirisha Chittiboyina
- Purdue University, Department of Basic Medical Sciences, 625 Harrison Street, West Lafayette, IN 47907, USA; 3D Cell Culture Core (3D3C) Facility, Birck Nanotechnology Center, Purdue University Discovery Park, 1205 West State Street, West Lafayette, IN 47907, USA
| |
Collapse
|
30
|
Planz V, Lehr CM, Windbergs M. In vitro models for evaluating safety and efficacy of novel technologies for skin drug delivery. J Control Release 2016; 242:89-104. [PMID: 27612408 DOI: 10.1016/j.jconrel.2016.09.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/22/2016] [Accepted: 09/05/2016] [Indexed: 12/14/2022]
Abstract
For preclinical testing of novel therapeutics, predictive in vitro models of the human skin are required to assess efficacy, absorption and safety. Simple as well as more sophisticated three-dimensional organotypic models of the human skin emerged as versatile and powerful tools simulating healthy as well as diseased skin states. Besides addressing the demands of research and industry, such models serve as valid alternative to animal testing. Recently, the acceptance of several models by regulatory authorities corroborates their role as important building block for preclinical development. However, valid assessment of readout parameters derived from these models requires suitable analytical techniques. Standard analytical methods are mostly destructive and limited regarding in-depth investigation on molecular level. The combination of adequate in vitro models with modern non-invasive analytical modalities bears a great potential to address important skin drug delivery related questions. Topics of interest are for instance the assessment of repeated dosing effects and xenobiotic biotransformation, which cannot be analyzed by destructive techniques. This review provides a comprehensive overview of current in vitro skin models differing in functional complexity and mimicking healthy as well as diseased skin states. Further, benefits and limitations regarding analytical evaluation of efficacy, absorption and safety of novel drug carrier systems applied to such models are discussed along with a prospective view of anticipated future directions. In addition, emerging non-invasive imaging modalities are introduced and their significance and potential to advance current knowledge in the field of skin drug delivery is explored.
Collapse
Affiliation(s)
- Viktoria Planz
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Department of Drug Delivery (DDEL), 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; PharmBioTec GmbH, 66123 Saarbrücken, Germany
| | - Claus-Michael Lehr
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Department of Drug Delivery (DDEL), 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; PharmBioTec GmbH, 66123 Saarbrücken, Germany
| | - Maike Windbergs
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Department of Drug Delivery (DDEL), 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; PharmBioTec GmbH, 66123 Saarbrücken, Germany.
| |
Collapse
|
31
|
Fino P, Spagnoli AM, Ruggieri M, Onesti MG. Caustic burn caused by intradermal self administration of muriatic acid for suicidal attempt: optimal wound healing and functional recovery with a non surgical treatment. G Chir 2015; 36:214-8. [PMID: 26712258 DOI: 10.11138/gchir/2015.36.5.214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Caustic burns are burns of third and fourth degree caused by strong acids or strong bases. Muriatic acid is often used for suicidal attempt by ingestion. We describe a case of a caustic skin lesion caused by intravenous failed attempt of suicide by injection of Muriatic acid in a woman affected with bipolar-syndrome. Generally, caustic burns are treated by cleansing, escarectomy and coverage with skin grafts. CASE REPORT We treated the patient with a non invasive technique with collagenase and hyaluronic acid sodium salt cream (Bionect start®), hyaluronic acid-based matrix (Hyalomatrix®) and Vacuum-Assisted Closure (VAC) Therapy®. RESULTS We obtained complete healing in 6 weeks. CONCLUSIONS Combined use of non invasive techniques seems to ensure only advantages for both the patients and the Health System. It reduces health care costs and risks for the patients such as nosocomial infections. Patient's compliance is high, as its quality of life. Complete healing of the wound is fast and recovery of function is full.
Collapse
|
32
|
Fibroblast heterogeneity and its implications for engineering organotypic skin models in vitro. Eur J Cell Biol 2015; 94:483-512. [PMID: 26344860 DOI: 10.1016/j.ejcb.2015.08.001] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 08/11/2015] [Accepted: 08/11/2015] [Indexed: 12/19/2022] Open
Abstract
Advances in cell culture methods, multidisciplinary research, clinical need to replace lost skin tissues and regulatory need to replace animal models with alternative test methods has led to development of three dimensional models of human skin. In general, these in vitro models of skin consist of keratinocytes cultured over fibroblast-populated dermal matrices. Accumulating evidences indicate that mesenchyme-derived signals are essential for epidermal morphogenesis, homeostasis and differentiation. Various studies show that fibroblasts isolated from different tissues in the body are dynamic in nature and are morphologically and functionally heterogeneous subpopulations. Further, these differences seem to be dictated by the local biological and physical microenvironment the fibroblasts reside resulting in "positional identity or memory". Furthermore, the heterogeneity among the fibroblasts play a critical role in scarless wound healing and complete restoration of native tissue architecture in fetus and oral mucosa; and excessive scar formation in diseased states like keloids and hypertrophic scars. In this review, we summarize current concepts about the heterogeneity among fibroblasts and their role in various wound healing environments. Further, we contemplate how the insights on fibroblast heterogeneity could be applied for the development of next generation organotypic skin models.
Collapse
|
33
|
Pinto S, Stark HJ, Martin I, Boukamp P, Kyewski B. 3D Organotypic Co-culture Model Supporting Medullary Thymic Epithelial Cell Proliferation, Differentiation and Promiscuous Gene Expression. J Vis Exp 2015:e52614. [PMID: 26275017 DOI: 10.3791/52614] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Intra-thymic T cell development requires an intricate three-dimensional meshwork composed of various stromal cells, i.e., non-T cells. Thymocytes traverse this scaffold in a highly coordinated temporal and spatial order while sequentially passing obligatory check points, i.e., T cell lineage commitment, followed by T cell receptor repertoire generation and selection prior to their export into the periphery. The two major resident cell types forming this scaffold are cortical (cTECs) and medullary thymic epithelial cells (mTECs). A key feature of mTECs is the so-called promiscuous expression of numerous tissue-restricted antigens. These tissue-restricted antigens are presented to immature thymocytes directly or indirectly by mTECs or thymic dendritic cells, respectively resulting in self-tolerance. Suitable in vitro models emulating the developmental pathways and functions of cTECs and mTECs are currently lacking. This lack of adequate experimental models has for instance hampered the analysis of promiscuous gene expression, which is still poorly understood at the cellular and molecular level. We adapted a 3D organotypic co-culture model to culture ex vivo isolated mTECs. This model was originally devised to cultivate keratinocytes in such a way as to generate a skin equivalent in vitro. The 3D model preserved key functional features of mTEC biology: (i) proliferation and terminal differentiation of CD80(lo), Aire-negative into CD80(hi), Aire-positive mTECs, (ii) responsiveness to RANKL, and (iii) sustained expression of FoxN1, Aire and tissue-restricted genes in CD80(hi) mTECs.
Collapse
Affiliation(s)
- Sheena Pinto
- Division of Developmental Immunology, German Cancer Research Center (DKFZ);
| | - Hans-Jürgen Stark
- Genetics of Skin Carcinogenesis, German Cancer Research Center (DKFZ);
| | - Iris Martin
- Genetics of Skin Carcinogenesis, German Cancer Research Center (DKFZ)
| | - Petra Boukamp
- Genetics of Skin Carcinogenesis, German Cancer Research Center (DKFZ)
| | - Bruno Kyewski
- Division of Developmental Immunology, German Cancer Research Center (DKFZ)
| |
Collapse
|
34
|
Papuga AY, Lukash LL. Different types of biotechnological wound coverages created with the application of alive human cells. ACTA ACUST UNITED AC 2015. [DOI: 10.7124/bc.0008d1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- A. Ye. Papuga
- Institute of Molecular Biology and Genetics, NAS of Ukraine
| | - L. L. Lukash
- Institute of Molecular Biology and Genetics, NAS of Ukraine
| |
Collapse
|
35
|
Soboleva AG, Mezentsev A, Zolotorenko A, Bruskin S, Pirusian E. Three-Dimensional Skin Models of Psoriasis. Cells Tissues Organs 2015; 199:301-10. [DOI: 10.1159/000369925] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2014] [Indexed: 11/19/2022] Open
|
36
|
Dermal matrices and bioengineered skin substitutes: a critical review of current options. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2015; 3:e284. [PMID: 25674365 PMCID: PMC4323388 DOI: 10.1097/gox.0000000000000219] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 09/16/2014] [Indexed: 12/22/2022]
Abstract
Background: Over recent decades, scientists and surgeons have collaborated to develop various bioengineered and synthetic products as an alternative to skin grafts. Despite the numerous articles and reviews written about dermal skin substitutes, there is no general consensus. Methods: This article reviews dermal skin scaffolds used in clinical applications and experimental settings. For scaffold evaluation, we focused on clinical and/or histological results, and conclusions are listed. Explanations for general trends were sought based on existing knowledge about tissue engineering principles and wound healing mechanisms. Results: Decellularized dermis seems to remain the best option with no other acellular scaffold being clinically proven to gain better results yet. In general, chemically cross-linked products were seen to be less effective in skin tissue engineering. Biocompatibility could be enhanced by preseeding substitutes with fibroblasts to allow some natural scaffold remodeling before product application. Conclusions: Skin substitutes are a useful tool in plastic and reconstructive surgery practices as an alternative to skin grafts. In the choice of substitute, the general plastic surgery principle of replacing like tissue with like tissue seems to be still standing, and products most resembling the natural dermal extracellular matrix should be preferred.
Collapse
|
37
|
Monteiro IP, Gabriel D, Timko BP, Hashimoto M, Karajanagi S, Tong R, Marques AP, Reis RL, Kohane DS. A two-component pre-seeded dermal-epidermal scaffold. Acta Biomater 2014; 10:4928-4938. [PMID: 25192821 PMCID: PMC4254066 DOI: 10.1016/j.actbio.2014.08.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 08/18/2014] [Accepted: 08/25/2014] [Indexed: 12/14/2022]
Abstract
We have developed a bilayered dermal-epidermal scaffold for application in the treatment of full-thickness skin defects. The dermal component gels in situ and adapts to the lesion shape, delivering human dermal fibroblasts in a matrix of fibrin and cross-linked hyaluronic acid modified with a cell adhesion-promoting peptide. Fibroblasts were able to form a tridimensional matrix due to material features such as tailored mechanical properties, presence of protease-degradable elements and cell-binding ligands. The epidermal component is a robust membrane containing cross-linked hyaluronic acid and poly-l-lysine, on which keratinocytes were able to attach and to form a monolayer. Amine-aldehyde bonding at the interface between the two components allows the formation of a tightly bound composite scaffold. Both parts of the scaffold were designed to provide cell-type-specific cues to allow for cell proliferation and form a construct that mimics the skin environment.
Collapse
Affiliation(s)
- I P Monteiro
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Taipas, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory University of Minho, Braga/Guimarães, Portugal
| | - D Gabriel
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - B P Timko
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - M Hashimoto
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - S Karajanagi
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - R Tong
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - A P Marques
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Taipas, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory University of Minho, Braga/Guimarães, Portugal
| | - R L Reis
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Taipas, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory University of Minho, Braga/Guimarães, Portugal
| | - D S Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
38
|
Longinotti C. The use of hyaluronic acid based dressings to treat burns: A review. BURNS & TRAUMA 2014; 2:162-8. [PMID: 27602379 PMCID: PMC5012021 DOI: 10.4103/2321-3868.142398] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/08/2014] [Accepted: 09/09/2014] [Indexed: 11/19/2022]
Abstract
Deep cutaneous lesions such as burns, traumas or ulcers are all conditions characterized by a massive loss of dermis, bringing several important consequences. For the treatment of these conditions, the evolution of material science has made available new dressings based on natural and synthetic polymers. Hyaluronic acid (HA) is involved in many steps of the wound healing process, such as inflammation, granulation and re-epithelialization. In order to overcome the poor physical properties of the native polymer, such as solubility and rapid degradation, insoluble molecules starting from the natural compound were produced via esterification. Thanks to their improved structural properties, the dressings based on these hyaluronic acid derivatives represent a valuable option for the treatment of deep burns. This narrative monograph describes the development and the outcome of the use of these products in burns. The currently available clinical experience suggests that these HA medical devices represent a safe therapeutic method useful for the treatment of acute wounds.
Collapse
Affiliation(s)
- Cristina Longinotti
- Research and Development, Anika Therapeutics S.r.l, via Ponte della Fabbrica 3b, 35031 Abano Terme, Italy
| |
Collapse
|
39
|
Guiraud B, Hernandez-Pigeon H, Ceruti I, Mas S, Palvadeau Y, Saint-Martory C, Castex-Rizzi N, Duplan H, Bessou-Touya S. Characterization of a human epidermis model reconstructed from hair follicle keratinocytes and comparison with two commercially models and native skin. Int J Cosmet Sci 2014; 36:485-93. [DOI: 10.1111/ics.12150] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 06/28/2014] [Indexed: 12/30/2022]
Affiliation(s)
- B. Guiraud
- Département de Pharmacologie In Vitro; Pierre Fabre Dermo-Cosmétique; 3 Avenue Hubert Curien BP13562 Toulouse CEDEX 31035 France
| | - H. Hernandez-Pigeon
- Département de Pharmacologie In Vitro; Pierre Fabre Dermo-Cosmétique; 3 Avenue Hubert Curien BP13562 Toulouse CEDEX 31035 France
| | - I. Ceruti
- Département de Pharmacologie In Vitro; Pierre Fabre Dermo-Cosmétique; 3 Avenue Hubert Curien BP13562 Toulouse CEDEX 31035 France
| | - S. Mas
- Département de Pharmacologie In Vitro; Pierre Fabre Dermo-Cosmétique; 3 Avenue Hubert Curien BP13562 Toulouse CEDEX 31035 France
| | - Y. Palvadeau
- Département de Pharmacologie In Vitro; Pierre Fabre Dermo-Cosmétique; 3 Avenue Hubert Curien BP13562 Toulouse CEDEX 31035 France
| | - C. Saint-Martory
- Pole Recherche, Exploration, Développement Clinique; Pierre Fabre Dermo-Cosmétique; 2, Rue Viguerie 31025 Toulouse Cedex France
| | - N. Castex-Rizzi
- Département de Pharmacologie In Vitro; Pierre Fabre Dermo-Cosmétique; 3 Avenue Hubert Curien BP13562 Toulouse CEDEX 31035 France
| | - H. Duplan
- Département de Pharmacologie In Vitro; Pierre Fabre Dermo-Cosmétique; 3 Avenue Hubert Curien BP13562 Toulouse CEDEX 31035 France
| | - S. Bessou-Touya
- Département de Pharmacologie In Vitro; Pierre Fabre Dermo-Cosmétique; 3 Avenue Hubert Curien BP13562 Toulouse CEDEX 31035 France
| |
Collapse
|
40
|
Monteiro IP, Shukla A, Marques AP, Reis RL, Hammond PT. Spray-assisted layer-by-layer assembly on hyaluronic acid scaffolds for skin tissue engineering. J Biomed Mater Res A 2014; 103:330-40. [PMID: 24659574 DOI: 10.1002/jbm.a.35178] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/11/2014] [Accepted: 03/19/2014] [Indexed: 01/12/2023]
Abstract
Tissue engineering approaches for the development of a single epidermal-dermal scaffold to treat full-thickness skin defects have been limited by difficulties in the fabrication of a bilayer scaffold combining the specific properties of the epidermis and the dermis. Here we present an innovative approach to developing a scaffold that holds promise for skin tissue engineering. We utilize the spray-assisted layer-by-layer assembly technique to deposit a polyelectrolyte multilayer film composed of hyaluronic acid and poly-L-lysine (the epidermal component) on a porous hyaluronic acid scaffold (the dermal component), in a rapid and controlled manner. The multilayer film promotes cell adhesion, contributing to regeneration of the epidermal barrier functions of skin. While human keratinocytes attached and proliferated on the coated porous scaffolds, they did not invade the porous dermal component, thus leaving room for seeding of relevant fibroblast cell types in this scaffold. This scaffold therefore holds promise for co-culture of different cells, which may be useful for treatment of full-thickness skin defects as well as other tissue engineering applications.
Collapse
Affiliation(s)
- Isa P Monteiro
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139; 3B's Research Group, Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Caldas das Taipas, Guimarães, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | | | | | | |
Collapse
|
41
|
Onesti MG, Fino P, Ponzo I, Ruggieri M, Scuderi N. Non-surgical treatment of deep wounds triggered by harmful physical and chemical agents: a successful combined use of collagenase and hyaluronic acid. Int Wound J 2014; 13:22-6. [PMID: 24698215 DOI: 10.1111/iwj.12215] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 12/19/2013] [Indexed: 11/30/2022] Open
Abstract
Some chronic ulcers often occur with slough, not progressing through the normal stages of wound healing. Treatment is long and other therapies need to be performed in addition to surgery. Patients not eligible for surgery because of ASA class (American Society of Anesthesiologists class) appear to benefit from chemical therapy with collagenase or hydrocolloids in order to prepare the wound bed, promoting the healing process. We describe four cases of traumatic, upper limb deep wounds caused by different physical and chemical agents, emphasising the effectiveness of treatment based on topical application of collagenase and hyaluronic acid (HA) before standardised surgical procedures. We performed careful disinfection of lesions combined with application of topical cream containing hyaluronic acid, bacterial fermented sodium hyaluronate (0·2%w/w) salt, and bacterial collagenase obtained from non-pathogenic Vibrio alginolyticus (>2·0 nkat1/g). In one patient a dermo-epidermal graft was used to cover the wide loss of substance. In two patients application of a HA-based dermal substitute was done. We obtained successful results in terms of wound healing, with satisfactory aesthetic result and optimal recovery of the affected limb functionality. Topical application of collagenase and HA, alone or before standardised surgical procedures allows faster wound healing.
Collapse
Affiliation(s)
- Maria G Onesti
- Department of Plastic, Reconstructive and Aesthetic Surgery, University of Rome La Sapienza, Rome, Italy
| | - Pasquale Fino
- Department of Plastic, Reconstructive and Aesthetic Surgery, University of Rome La Sapienza, Rome, Italy
| | - Ida Ponzo
- Department of Plastic, Reconstructive and Aesthetic Surgery, University of Rome La Sapienza, Rome, Italy
| | - Martina Ruggieri
- Department of Plastic, Reconstructive and Aesthetic Surgery, University of Rome La Sapienza, Rome, Italy
| | - Nicolò Scuderi
- Department of Plastic, Reconstructive and Aesthetic Surgery, University of Rome La Sapienza, Rome, Italy
| |
Collapse
|
42
|
Onesti MG, Fino P, Fioramonti P, Amorosi V, Scuderi N. Reconstruction after skin cancer excision through a dermal induction template: our experience. Int Wound J 2014; 13:198-203. [PMID: 24684743 DOI: 10.1111/iwj.12255] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Dermal substitutes offer alternative approaches for wounds of all thicknesses where sufficient donation sites are not available for self-grafts. Several dermal substitutes are described in literature. This study included 20 patients treated with a dermal induction template after the removal of malignant skin cancers situated in various parts of the body. The participants were especially aged patients with multiple skin cancers, and complex clinical conditions, often affected by pathologies such as cardiopathy, diabetes mellitus, and hypercholesterolaemia, and receiving pharmacological multi-therapies, particularly antiplatelets and anticoagulants. In many of these patients, the general complex clinical picture provided significant contraindication for complex reconstructive surgery because of the high risk involved. All patients achieved complete healing about 8 weeks after the first surgery. By using a dermal induction template, it was possible to cover substantial loss of substances without the need of autologous tissue, with smoother and less apparent scar, minor occurrence of hypertrophic and retracted scars, better flexibility of healed skin and therefore a better result from an aesthetic point of view.
Collapse
Affiliation(s)
- Maria G Onesti
- Department of Plastic and Reconstructive Surgery, "Sapienza" University, Policlinico Umberto I, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Pasquale Fino
- Department of Plastic and Reconstructive Surgery, "Sapienza" University, Policlinico Umberto I, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Paolo Fioramonti
- Department of Plastic and Reconstructive Surgery, "Sapienza" University, Policlinico Umberto I, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Vittoria Amorosi
- Department of Plastic and Reconstructive Surgery, "Sapienza" University, Policlinico Umberto I, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Nicolò Scuderi
- Department of Plastic and Reconstructive Surgery, "Sapienza" University, Policlinico Umberto I, Viale del Policlinico, 155, 00161 Rome, Italy
| |
Collapse
|
43
|
Lemper M, Snykers S, Vanhaecke T, De Paepe K, Rogiers V. Current Status of Healthy Human Skin Models: Can Histone Deacetylase Inhibitors Potentially Improve the Present Replacement Models? Skin Pharmacol Physiol 2014; 27:36-46. [DOI: 10.1159/000351363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 03/24/2013] [Indexed: 11/19/2022]
|
44
|
Guo Z, Higgins CA, Gillette BM, Itoh M, Umegaki N, Gledhill K, Sia SK, Christiano AM. Building a microphysiological skin model from induced pluripotent stem cells. Stem Cell Res Ther 2013; 4 Suppl 1:S2. [PMID: 24564920 PMCID: PMC4029476 DOI: 10.1186/scrt363] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The discovery of induced pluripotent stem cells (iPSCs) in 2006 was a major breakthrough for regenerative medicine. The establishment of patient-specific iPSCs has created the opportunity to model diseases in culture systems, with the potential to rapidly advance the drug discovery field. Current methods of drug discovery are inefficient, with a high proportion of drug candidates failing during clinical trials due to low efficacy and/or high toxicity. Many drugs fail toxicity testing during clinical trials, since the cells on which they have been tested do not adequately model three-dimensional tissues or their interaction with other organs in the body. There is a need to develop microphysiological systems that reliably represent both an intact tissue and also the interaction of a particular tissue with other systems throughout the body. As the port of entry for many drugs is via topical delivery, the skin is the first line of exposure, and also one of the first organs to demonstrate a reaction after systemic drug delivery. In this review, we discuss our strategy to develop a microphysiological system using iPSCs that recapitulates human skin for analyzing the interactions of drugs with the skin.
Collapse
|
45
|
Langbein L, Reichelt J, Eckhart L, Praetzel-Wunder S, Kittstein W, Gassler N, Schweizer J. New facets of keratin K77: interspecies variations of expression and different intracellular location in embryonic and adult skin of humans and mice. Cell Tissue Res 2013; 354:793-812. [PMID: 24057875 DOI: 10.1007/s00441-013-1716-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 07/19/2013] [Indexed: 01/08/2023]
Abstract
The differential expression of keratins is central to the formation of various epithelia and their appendages. Structurally, the type II keratin K77 is closely related to K1, the prototypical type II keratin of the suprabasal epidermis. Here, we perform a developmental study on K77 expression in human and murine skin. In both species, K77 is expressed in the suprabasal fetal epidermis. While K77 appears after K1 in the human epidermis, the opposite is true for the murine tissue. This species-specific pattern of expression is also found in conventional and organotypic cultures of human and murine keratinocytes. Ultrastructure investigation shows that, in contrast to K77 intermediate filaments of mice, those of the human ortholog are not attached to desmosomes. After birth, K77 disappears without deleterious consequences from human epidermis while it is maintained in the adult mouse epidermis, where its presence has so far gone unnoticed. After targeted Krt1 gene deletion in mice, K77 is normally expressed but fails to functionally replace K1. Besides the epidermis, both human and mouse K77 are present in luminal duct cells of eccrine sweat glands. The demonstration of a K77 ortholog in platypus but not in non-mammalian vertebrates identifies K77 as an evolutionarily ancient component of the mammalian integument that has evolved different patterns of intracellular distribution and adult tissue expression in primates.
Collapse
Affiliation(s)
- Lutz Langbein
- Genetics of Skin Carcinogenesis, A110, German Cancer Research Center, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany,
| | | | | | | | | | | | | |
Collapse
|
46
|
Kamel RA, Ong JF, Eriksson E, Junker JPE, Caterson EJ. Tissue engineering of skin. J Am Coll Surg 2013; 217:533-55. [PMID: 23816384 DOI: 10.1016/j.jamcollsurg.2013.03.027] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 03/15/2013] [Accepted: 03/18/2013] [Indexed: 11/18/2022]
Affiliation(s)
- Rami A Kamel
- Division of Plastic Surgery, Brigham and Women's Surgery, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
47
|
Breitkreutz D, Koxholt I, Thiemann K, Nischt R. Skin basement membrane: the foundation of epidermal integrity--BM functions and diverse roles of bridging molecules nidogen and perlecan. BIOMED RESEARCH INTERNATIONAL 2013; 2013:179784. [PMID: 23586018 PMCID: PMC3618921 DOI: 10.1155/2013/179784] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 01/18/2013] [Accepted: 01/28/2013] [Indexed: 02/06/2023]
Abstract
The epidermis functions in skin as first defense line or barrier against environmental impacts, resting on extracellular matrix (ECM) of the dermis underneath. Both compartments are connected by the basement membrane (BM), composed of a set of distinct glycoproteins and proteoglycans. Herein we are reviewing molecular aspects of BM structure, composition, and function regarding not only (i) the dermoepidermal interface but also (ii) the resident microvasculature, primarily focusing on the per se nonscaffold forming components perlecan and nidogen-1 and nidogen-2. Depletion or functional deficiencies of any BM component are lethal at some stage of development or around birth, though BM defects vary between organs and tissues. Lethality problems were overcome by developmental stage- and skin-specific gene targeting or by cell grafting and organotypic (3D) cocultures of normal or defective cells, which allows recapitulating BM formation de novo. Thus, evidence is accumulating that BM assembly and turnover rely on mechanical properties and composition of the adjacent ECM and the dynamics of molecular assembly, including further "minor" local components, nidogens largely functioning as catalysts or molecular adaptors and perlecan as bridging stabilizer. Collectively, orchestration of BM assembly, remodeling, and the role of individual players herein are determined by the developmental, tissue-specific, or functional context.
Collapse
Affiliation(s)
- Dirk Breitkreutz
- Department of Dermatology, University of Cologne, Kerpener Strasse 62, 50937 Cologne, Germany.
| | | | | | | |
Collapse
|
48
|
Pontiggia L, Klar A, Böttcher-Haberzeth S, Biedermann T, Meuli M, Reichmann E. Optimizing in vitro culture conditions leads to a significantly shorter production time of human dermo-epidermal skin substitutes. Pediatr Surg Int 2013; 29:249-56. [PMID: 23377785 DOI: 10.1007/s00383-013-3268-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2013] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Autologous dermo-epidermal skin substitutes (DESS) generated in vitro represent a promising therapeutic means to treat full-thickness skin defects in clinical practice. A serious drawback with regard to acute patients is the relatively long production time of 3-4 weeks. With this experimental study we aimed to decrease the production time of DESS without compromising their quality. METHODS Two in vitro steps of DESS construction were varied: the pre-cultivation time of fibroblasts in hydrogels (1, 3, and 6 days), and the culture time of keratinocytes (3, 6, and 12 days) before transplantation of DESS on nude rats. Additionally, the impact of the air-liquid interface culture during 3 days before transplantation was investigated. 3 weeks after transplantation, the macroscopic appearance was evaluated and histological sections were produced to analyze structure and thickness of epidermis and dermis, the stratification of the epidermis, and the presence of a basal lamina. RESULTS Optimal DESS formation was obtained with a fibroblast pre-cultivation time of 6 days. The minimal culture time of keratinocytes on hydrogels was also 6 days. The air-liquid interface culture did not improve graft quality. CONCLUSION By optimizing our in vitro culture conditions, it was possible to very substantially reduce the production time for DESS from 21 to 12 days. However, pre-cultivation of fibroblasts in the dermal equivalent and proliferation of keratinocytes before transplantation remain crucial for an equilibrated maturation of the epidermis and cannot be completely skipped.
Collapse
Affiliation(s)
- Luca Pontiggia
- Tissue Biology Research Unit, University Children's Hospital Zurich, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
49
|
Farrugia BL, Brown TD, Upton Z, Hutmacher DW, Dalton PD, Dargaville TR. Dermal fibroblast infiltration of poly(ε-caprolactone) scaffolds fabricated by melt electrospinning in a direct writing mode. Biofabrication 2013; 5:025001. [DOI: 10.1088/1758-5082/5/2/025001] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
50
|
Pinto S, Schmidt K, Egle S, Stark HJ, Boukamp P, Kyewski B. An organotypic coculture model supporting proliferation and differentiation of medullary thymic epithelial cells and promiscuous gene expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:1085-93. [PMID: 23269248 DOI: 10.4049/jimmunol.1201843] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Understanding intrathymic T cell differentiation has been greatly aided by the development of various reductionist in vitro models that mimic certain steps/microenvironments of this complex process. Most models focused on the faithful in vitro restoration of T cell differentiation and selection. In contrast, suitable in vitro models emulating the developmental pathways of the two major thymic epithelial cell lineages--cortical thymic epithelial cells and medullary thymic epithelial cells (mTECs)--are yet to be developed. In this regard, lack of an in vitro model mimicking the developmental biology of the mTEC lineage has hampered the molecular analysis of the so-called "promiscuous expression" of tissue-restricted genes, a key property of terminally differentiated mTECs. Based on the close biological relationship between the skin and thymus epithelial cell compartments, we adapted a three-dimensional organotypic coculture model, originally developed to provide a bona fide in vitro dermal equivalent, for the culture of isolated mTECs. This three-dimensional model preserves key features of mTECs: proliferation and terminal differentiation of CD80(lo), Aire(-) mTECs into CD80(hi), Aire(+) mTECs; responsiveness to RANKL; and sustained expression of FoxN1, Aire, and tissue-restricted genes in CD80(hi) mTECs. This in vitro culture model should facilitate the identification of molecular components and pathways involved in mTEC differentiation in general and in promiscuous gene expression in particular.
Collapse
Affiliation(s)
- Sheena Pinto
- Division of Developmental Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|