1
|
Lechner J, vonBaehr V, Doebis C, Notter F, Schick F. Platelet-Rich Fibrin (PRF) Analyzed for Cytokine Profiles - A Misguided Hope for Osteogenesis in Jawbone Defects? Research and Clinical Observational Study. Clin Cosmet Investig Dent 2024; 16:467-479. [PMID: 39583889 PMCID: PMC11585297 DOI: 10.2147/ccide.s488206] [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: 09/04/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024] Open
Abstract
Background Platelet-rich fibrin (PRF) blood concentrates are used in oral implantology and defect surgery to promote osteoneogenesis in Bone Marrow Defects in Jawbone (BMDJ), according to the morphology of fatty-degenerative osteonecrosis also called FDOJ. Question Can the benefit of PRF on alveolar osteoneogenesis be confirmed by cytokine analysis?. Methods The cytokine expressions of the PRF samples in 26 patients undergoing BMDJ/FDOJ surgery in the same session were analysed for seven cytokines (RANTES/CCL5; FGF-2; IL-1RA; Il-6; IL-8; MCP-1; TNF-a) by multiplex (Luminex). The FDOJ samples of these 26 BMDJ/FDOJ patients were analysed for the RANTES/CCL5 expression only. Results Cytokine expression in PRF is compared to reference values for healthy medullary bone of the jaw and BMDJ/FDOJ and shows that the cytokine expressions of the PRF samples do not compensate or counteract prima vista for the cytokine dysregulations present in the BMDJ/FDOJ areas. Discussion To define the aid of cytokines studied in PRF in the restoration of the immunological dysregulation in areas of BMDJ/FDOJ, literature is reviewed comparing RANTES/CCL5, IL-1ra, TNF-α and MCP-1/CCL2 expression in PRF and BMDJ/FDOJ. Immunoregulatory properties of PRF in alveolar bone restoration are evaluated. Summary PRF was mistakenly thought to be a cure for bone healing, which is here shown to be incorrect. Enoral Ultrasound Sonography of bone density is available for the clinical measurement of individually developed osteoneogenesis by PRF. Conclusion The multiplex analysis of PRF shows a dynamic and cytokine-based interaction with osteoneogenesis that is not yet fully clarified.
Collapse
Affiliation(s)
- Johann Lechner
- Department of Osteoimmunology, Clinic for Integrative Dentistry, Munich, Germany
| | - Volker vonBaehr
- Department of Immunology and Allergology, Institute for Medical Diagnostics, Berlin, Germany
| | - Cornelia Doebis
- Department of Analysis, Institute for Medical Diagnostics, Berlin, Germany
| | - Florian Notter
- Department of Implantology, Clinic for Integrative Dentistry, Munich, Germany
| | - Fabian Schick
- Department of Implantology, Clinic for Integrative Dentistry, Munich, Germany
| |
Collapse
|
2
|
Michielon E, Boninsegna M, Waaijman T, Fassini D, Spiekstra SW, Cramer J, Gaudriault P, Kodolányi J, de Gruijl TD, Homs-Corbera A, Gibbs S. Environmentally Controlled Microfluidic System Enabling Immune Cell Flow and Activation in an Endothelialised Skin-On-Chip. Adv Healthc Mater 2024; 13:e2400750. [PMID: 39370595 PMCID: PMC11582514 DOI: 10.1002/adhm.202400750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 09/17/2024] [Indexed: 10/08/2024]
Abstract
Integration of reconstructed human skin (RhS) into organ-on-chip (OoC) platforms addresses current limitations imposed by static culturing. This innovation, however, is not without challenges. Microfluidic devices, while powerful, often encounter usability, robustness, and gas bubble issues that hinder large-scale high-throughput setups. This study aims to develop a novel re-usable multi-well microfluidic adaptor (MMA) with the objective to provide a flexible tool for biologists implementing complex 3D biological models (e.g., skin) while enabling simultaneous user control over temperature, medium flow, oxigen (O2), nitrogen (N2), and carbon dioxide (CO2) without the need for an incubator. The presented MMA device is designed to be compatible with standard, commercially available 6-well multi-well plates (6MWPs) and 12-well transwells. This MMA-6MWP setup is employed to generate a skin-on-chip (SoC). RhS viability is maintained under flow for three days and their morphology closely resembles that of native human skin. A proof-of-concept study demonstrates the system's potential in toxicology applications by combining endothelialised RhS with flowing immune cells. This dynamic setting activates the monocyte-like MUTZ-3 cells (CD83 and CD86 upregulation) upon topical exposure of RhS to a sensitizer, revealing the MMA-6MWP's unique capabilities compared to static culturing, where such activation is absent.
Collapse
Affiliation(s)
- Elisabetta Michielon
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam UMC, Vrije Universiteit, Amsterdam, 1081 HV, The Netherlands
| | - Matteo Boninsegna
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
- Department of Physics, Bielefeld University, Universitätsstr 25, 33615, Bielefeld, Germany
| | - Taco Waaijman
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
| | - Dario Fassini
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - Sander W Spiekstra
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jeremy Cramer
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - Pierre Gaudriault
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - János Kodolányi
- Department of Dental Material Science, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, 1081 LA, The Netherlands
| | - Tanja D de Gruijl
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam UMC, Vrije Universiteit, Amsterdam, 1081 HV, The Netherlands
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
| | - Antoni Homs-Corbera
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, 1081 LA, The Netherlands
| |
Collapse
|
3
|
Sierra-Sánchez Á, Cabañas-Penagos J, Igual-Roger S, Martínez-Heredia L, Espinosa-Ibáñez O, Sanabria-de la Torre R, Quiñones-Vico MI, Ubago-Rodríguez A, Lizana-Moreno A, Fernández-González A, Guerrero-Calvo J, Fernández-Porcel N, Ramírez-Muñoz A, Arias-Santiago S. Biological properties and characterization of several variations of a clinical human plasma-based skin substitute model and its manufacturing process. Regen Biomater 2024; 11:rbae115. [PMID: 39469583 PMCID: PMC11513639 DOI: 10.1093/rb/rbae115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/07/2024] [Accepted: 09/11/2024] [Indexed: 10/30/2024] Open
Abstract
Human plasma is a natural biomaterial that due to their protein composition is widely used for the development of clinical products, especially in the field of dermatology. In this context, this biomaterial has been used as a scaffold alone or combined with others for the development of cellular human plasma-based skin substitutes (HPSSs). Herein, the biological properties (cell viability, cell metabolic activity, protein secretion profile and histology) of several variations of a clinical HPSS model, regarding the biomaterial composition (alone or combined with six secondary biomaterials - serine, fibronectin, collagen, two types of laminins and hyaluronic acid), the cellular structure (trilayer, bilayer, monolayer and control without cells) and their skin tissue of origin (abdominal or foreskin cells) and the manufacturing process [effect of partial dehydration process in cell viability and comparison between submerged (SUB) and air/liquid interface (ALI) methodologies] have been evaluated and compared. Results reveal that the use of human plasma as a main biomaterial determines the in vitro properties, rather than the secondary biomaterials added. Moreover, the characteristics are similar regardless of the skin cells used (from abdomen or foreskin). However, the manufacture of more complex cellular substitutes (trilayer and bilayer) has been demonstrated to be better in terms of cell viability, metabolic activity and wound healing protein secretion (bFGF, EGF, VEGF-A, CCL5) than monolayer HPSSs, especially when ALI culture methodology is applied. Moreover, the application of the dehydration, although required to achieve an appropriate clinical structure, reduce cell viability in all cases. These data indicate that this HPSS model is robust and reliable and that the several subtypes here analysed could be promising clinical approaches depending on the target dermatological disease.
Collapse
Affiliation(s)
- Álvaro Sierra-Sánchez
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, 18012, Spain
- Department of Dermatology, Virgen de las Nieves University Hospital, Granada, 18012, Spain
| | - Jorge Cabañas-Penagos
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
| | - Sandra Igual-Roger
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
| | - Luis Martínez-Heredia
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, 18012, Spain
| | - Olga Espinosa-Ibáñez
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, 18012, Spain
| | - Raquel Sanabria-de la Torre
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, 18012, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, University of Granada, Granada, 18071, Spain
| | - María I Quiñones-Vico
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, 18012, Spain
- Department of Dermatology, University of Granada, Granada, 18016, Spain
| | - Ana Ubago-Rodríguez
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, 18012, Spain
| | - Antonio Lizana-Moreno
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, 18012, Spain
| | - Ana Fernández-González
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, 18012, Spain
| | - Jorge Guerrero-Calvo
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, 18012, Spain
| | - Natividad Fernández-Porcel
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, 18012, Spain
| | - Arena Ramírez-Muñoz
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, 18012, Spain
| | - Salvador Arias-Santiago
- Andalusian Network of Design and Translation of Advanced Therapies, Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Granada, 18014, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, 18012, Spain
- Department of Dermatology, Virgen de las Nieves University Hospital, Granada, 18012, Spain
- Department of Dermatology, University of Granada, Granada, 18016, Spain
| |
Collapse
|
4
|
Chen B, Zhu X, Zhang D, Zhu Z, Ye Q, Guo J. Adipose-derived mesenchymal stem cells suppress fibroblast proliferation of hypertrophic scar through CCL5 and CXCL12. Arch Dermatol Res 2024; 316:527. [PMID: 39153095 DOI: 10.1007/s00403-024-03289-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/08/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND AND OBJECTIVE Adipose-derived mesenchymal stem cells (ADSCs) can accelerate wound healing, reduce scar formation, and inhibit hypertrophic scar (HTS). ADSCs can secrete a large amount of CCL5, and CCL5 has been proved to be pro-inflammatory and pro-fibrotic. CXCL12 (SDF-1) is a key chemokine that promotes stem cell migration and survival. Therefore, this study selected normal skin and HTS conditioned medium to simulate different microenvironments, and analyzed the effects of different microenvironments on the expression of CCL5 and CXCL12 in human ADSCs (hADSCs). MATERIALS AND METHODS hADSCs with silenced expression of CCL5 and CXCL12 were co-cultured with hypertrophic scar fibroblasts to verify the effects of CCL5 and CXCL12 in hADSCs on the proliferation ability of hypertrophic scar fibroblasts. A mouse model of hypertrophic scar was established to further confirm the effect of CCL5 and CXCL12 in hADSCs on hypertrophic scar formation. RESULTS CCL5 level was found to be significantly high in hADSCs cultured in HTS conditioned medium. CXCL12 in HTS group was prominently lowly expressed compared with the normal group. Inhibition of CCL5 in hADSCs enhanced the effects of untreated hADSCs on proliferation of HTS fibroblasts while CXCL12 knockdown exerted the opposite function. Inhibition of CCL5 in hADSCs increased the percentage of HTS fibroblasts in the G0/G1 phase while down-regulation of CXCL12 decreased those. Meanwhile, the down-regulated levels of fibroblast markers including collagen I, collagen III, and α-SMA induced by CCL5 knockdown were significantly up-regulated by CXCL12 inhibition. hADSCs alleviate the HTS of mice through CCL5 and CXCL12. CONCLUSION In summary, our results demonstrated that hADSCs efficiently cured HTS by suppressing proliferation of HTS fibroblasts, which may be related to the inhibition of CXCL12 and elevation of CCL5 in hADSCs, suggesting that hADSCs may provide an alternative therapeutic approach for the treatment of HTS.
Collapse
Affiliation(s)
- Bo Chen
- Department of Burns and Plastic Surgery, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen, 518000, Guangdong, China
| | - Xiongxiang Zhu
- Department of Burns and Plastic Surgery, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen, 518000, Guangdong, China
| | - Dongmei Zhang
- Department of Burns and Plastic Surgery, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen, 518000, Guangdong, China
| | - Zhensen Zhu
- Department of Burns and Plastic Surgery, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen, 518000, Guangdong, China
| | - Qian Ye
- Department of Orthopaedics, Shenzhen Hospital of Southern Medical University, Shenzhen, 518000, Guangdong, China
| | - Jingdong Guo
- Department of Burns and Plastic Surgery, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
5
|
Chen YC, Chuang EY, Tu YK, Hsu CL, Cheng NC. Human platelet lysate-cultured adipose-derived stem cell sheets promote angiogenesis and accelerate wound healing via CCL5 modulation. Stem Cell Res Ther 2024; 15:163. [PMID: 38853252 PMCID: PMC11163789 DOI: 10.1186/s13287-024-03762-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND A rising population faces challenges with healing-impaired cutaneous wounds, often leading to physical disabilities. Adipose-derived stem cells (ASCs), specifically in the cell sheet format, have emerged as a promising remedy for impaired wound healing. Human platelet lysate (HPL) provides an attractive alternative to fetal bovine serum (FBS) for culturing clinical-grade ASCs. However, the potential of HPL sheets in promoting wound healing has not been fully investigated. This study aimed to explore the anti-fibrotic and pro-angiogenic capabilities of HPL-cultured ASC sheets and delve into the molecular mechanism. METHODS A rat burn model was utilized to evaluate the efficacy of HPL-cultured ASC sheets in promoting wound healing. ASC sheets were fabricated with HPL, and those with FBS were included for comparison. Various analyses were conducted to assess the impact of HPL sheets on wound healing. Histological examination of wound tissues provided insights into aspects such as wound closure, collagen deposition, and overall tissue regeneration. Immunofluorescence was employed to assess the presence and distribution of transplanted ASCs after treatment. Further in vitro studies were conducted to decipher the specific factors in HPL sheets contributing to angiogenesis. RESULTS HPL-cultured ASC sheets significantly accelerated wound closure, fostering ample and organized collagen deposition in the neo-dermis. Significantly more retained ASCs were observed in wound tissues treated with HPL sheets compared to the FBS counterparts. Moreover, HPL sheets mitigated macrophage recruitment and decreased subsequent wound tissue fibrosis in vivo. Immunohistochemistry also indicated enhanced angiogenesis in the HPL sheet group. The in vitro analyses showed upregulation of C-C motif chemokine ligand 5 (CCL5) and angiogenin in HPL sheets, including both gene expression and protein secretion. Culturing endothelial cells in the conditioned media compared to media supplemented with CCL5 or angiogenin suggested a correlation between CCL5 and the pro-angiogenic effect of HPL sheets. Additionally, through neutralizing antibody experiments, we further validated the crucial role of CCL5 in HPL sheet-mediated angiogenesis in vitro. CONCLUSIONS The present study underscores CCL5 as an essential factor in the pro-angiogenic effect of HPL-cultured ASC sheets during the wound healing process. These findings highlight the potential of HPL-cultured ASC sheets as a promising therapeutic option for healing-impaired cutaneous wounds in clinical settings. Furthermore, the mechanism exploration yields valuable information for optimizing regenerative strategies with ASC products. BRIEF ACKNOWLEDGMENT This research was supported by the National Science and Technology Council, Taiwan (NSTC112-2321-B-002-018), National Taiwan University Hospital (111C-007), and E-Da Hospital-National Taiwan University Hospital Joint Research Program (111-EDN0001, 112-EDN0002).
Collapse
Affiliation(s)
- Yueh-Chen Chen
- Department of Surgery, National Taiwan University Hospital and College of Medicine, 7 Chung-Shan S. Rd, Taipei, 100, Taiwan
| | - Er-Yuan Chuang
- International Ph.D. Program in Biomedical Engineering, Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei, Taiwan
| | - Yuan-Kun Tu
- Department of Orthopedics, E-Da Hospital/I-Shou University, Kaohsiung, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Nai-Chen Cheng
- Department of Surgery, National Taiwan University Hospital and College of Medicine, 7 Chung-Shan S. Rd, Taipei, 100, Taiwan.
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
6
|
Jäger J, Vahav I, Thon M, Waaijman T, Spanhaak B, de Kok M, Bhogal RK, Gibbs S, Koning JJ. Reconstructed Human Skin with Hypodermis Shows Essential Role of Adipose Tissue in Skin Metabolism. Tissue Eng Regen Med 2024; 21:499-511. [PMID: 38367122 PMCID: PMC10987437 DOI: 10.1007/s13770-023-00621-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/17/2023] [Accepted: 08/27/2023] [Indexed: 02/19/2024] Open
Abstract
BACKGROUND Dysregulation of skin metabolism is associated with a plethora of diseases such as psoriasis and dermatitis. Until now, reconstructed human skin (RhS) models lack the metabolic potential of native human skin, thereby limiting their relevance to study human healthy and diseased skin. We aimed to determine whether incorporation of an adipocyte-containing hypodermis into RhS improves its metabolic potential and to identify major metabolic pathways up-regulated in adipose-RhS. METHODS Primary human keratinocytes, fibroblasts and differentiated adipose-derived stromal cells were co-cultured in a collagen/fibrin scaffold to create an adipose-RhS. The model was extensively characterized structurally in two- and three-dimensions, by cytokine secretion and RNA-sequencing for metabolic enzyme expression. RESULTS Adipose-RhS showed increased secretion of adipokines. Both RhS and adipose-RhS expressed 29 of 35 metabolic genes expressed in ex vivo native human skin. Addition of the adipose layer resulted in up-regulation of 286 genes in the dermal-adipose fraction of which 7 were involved in phase I (CYP19A1, CYP4F22, CYP3A5, ALDH3B2, EPHX3) and phase II (SULT2B1, GPX3) metabolism. Vitamin A, D and carotenoid metabolic pathways were enriched. Additionally, pro-inflammatory (IL-1β, IL-18, IL-23, IL-33, IFN-α2, TNF-α) and anti-inflammatory cytokine (IL-10, IL-12p70) secretion was reduced in adipose-RhS. CONCLUSIONS Adipose-RhS mimics healthy native human skin more closely than traditional RhS since it has a less inflamed phenotype and a higher metabolic activity, indicating the contribution of adipocytes to tissue homeostasis. Therefore it is better suited to study onset of skin diseases and the effect of xenobiotics.
Collapse
Affiliation(s)
- Jonas Jäger
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands
| | - Irit Vahav
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Tissue Function & Regeneration, Amsterdam, The Netherlands
| | - Maria Thon
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands
| | - Taco Waaijman
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands
| | - Bas Spanhaak
- Systems Biology Lab, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Michael de Kok
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands
| | | | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands
| | - Jasper J Koning
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Santolaya JL, Schweer DS, Cardenas-Goicoechea J, Bukowski R, Santolaya-Forgas J. Bioavailability of the tumor necrosis factor alpha/regulated on activation, normal T cell expressed and secreted (RANTES) biosystem inside the gestational sac during the pre-immune stages of embryo development. J Perinat Med 2023; 51:891-895. [PMID: 37067543 DOI: 10.1515/jpm-2022-0542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/06/2023] [Indexed: 04/18/2023]
Abstract
OBJECTIVES In-vivo studies of the bioavailability of major components of the tumor necrosis factor alpha (TNFα) biosystem inside the gestational sac during embryogenesis have not been reported. We sought to determine the concentration of TNFα, soluble (s) TNFα receptors (sTNFR1, sTNFR2), and RANTES in the primate extraembryonic celomic fluid (ECF). METHODS A validated timed-pregnant baboon animal model (N: 10) for experimental research in pregnancy was used to collect paired maternal blood and ECF samples in ongoing pregnancies. The concentrations (pg/dL) of TNFα, sTNFR1, sTNFR2, and RANTES were then determined by ELISA immunoassays. RESULTS All animals delivered at term healthy newborns. The differential concentration of TNFα, sTNFR1, sTNFR2, and RANTES between the maternal plasma and the ECF could be determined with ratios for TNFα (5.4), sTNFR2 (1.85) and RANTES (3.59) that contrasted with that of sTNFR1 (0.07), which favored the gestational sac compartment. No significant correlations were noted between maternal plasma and ECF TNFR1, sTNFR2 and RANTES. There was a trend for a correlation between TNFα in maternal plasma and ECF (R=0.74; p=0.07). CONCLUSIONS We report the physiological concentrations of TNFα, sTNFR1, sTNFR2, and RANTES in extraembryonic celomic fluid during embryogenesis in primates.
Collapse
Affiliation(s)
- Jacobo L Santolaya
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David S Schweer
- Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY, USA
| | | | - Radek Bukowski
- Department of Obstetrics and Gynecology, University of Texas, Austin, TX, USA
| | | |
Collapse
|
8
|
Schaffrick L, Ding J, Kwan P, Tredget E. The dynamic changes of monocytes and cytokines during wound healing post-burn injury. Cytokine 2023; 168:156231. [PMID: 37247448 DOI: 10.1016/j.cyto.2023.156231] [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: 03/02/2023] [Revised: 05/01/2023] [Accepted: 05/08/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND Burn injury is a sudden and traumatic injury that affects a large part of the population worldwide, who are placed at high risk of developing hypertrophic scars (HTS). HTS are a fibrotic scar resulting in painful contracted and raised scarring, affecting mobility in joints and work life, as well as cosmetically. The aim of this research was to enhance our understanding of the systematic response of monocytes and cytokines in wound healing after burn injury, in order to develop novel approaches to prevention and treatment of HTS. METHODS Twenty-seven burn patients and thirteen healthy individuals were recruited in this study. Burn patients were stratified by burn total body surface area (TBSA). Peripheral blood samples were taken post-burn injury. Serum and peripheral blood mononuclear cells (PBMCs) were separated from the blood samples. This research investigated cytokines IL-6, IL-8, IL1RA, IL-10, and chemokine pathways SDF-1/CXCR4, MCP-1/CCR2, RANTES/CCR5 during the wound healing process in burn patients with varying severity of injuries by using enzyme-linked immunosorbent assays. PBMCs were stained for monocytes and the chemokine receptors by flow cytometry. Statistical analysis was done by one-way ANOVA with a Tukey correction, and regression analysis was performed using Pearson's Correlation analysis. RESULTS The CD14+CD16- monocyte subpopulation is larger in patients who developed HTS at 4-7 days. The CD14+CD16+ monocyte subpopulation is smaller in the first week of injury, where it is similar after 8 days. Burn injury increased CXCR4, CCR2, and CCR5 expressions in CD14+ CD16+ monocytes. Increases in MCP-1 at 0-3 days after burn injury was positively correlated with burn severity. IL-6, IL-8, RANTES, and MCP-1 significantly increased with increasing burn severity. CONCLUSIONS Monocytes and their chemokine receptors, as well as systemic levels of cytokines in wound healing of burn patients and scar development will require ongoing assessment to enhance our understanding of the abnormal wound healing after burn injury.
Collapse
Affiliation(s)
- Lindy Schaffrick
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.
| | - Jie Ding
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.
| | - Peter Kwan
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada; Division of Critical Care, University of Alberta, Edmonton, Alberta, Canada
| | - Edward Tredget
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada; Division of Critical Care, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
9
|
Michielon E, López González M, Stolk DA, Stolwijk JGC, Roffel S, Waaijman T, Lougheed SM, de Gruijl TD, Gibbs S. A Reconstructed Human Melanoma-in-Skin Model to Study Immune Modulatory and Angiogenic Mechanisms Facilitating Initial Melanoma Growth and Invasion. Cancers (Basel) 2023; 15:2849. [PMID: 37345186 DOI: 10.3390/cancers15102849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 06/23/2023] Open
Abstract
Invasion, immune modulation, and angiogenesis are crucial in melanoma progression. Studies based on animals or two-dimensional cultures poorly recapitulate the tumor-microenvironmental cross-talk found in humans. This highlights a need for more physiological human models to better study melanoma features. Here, six melanoma cell lines (A375, COLO829, G361, MeWo, RPMI-7951, and SK-MEL-28) were used to generate an in vitro three-dimensional human melanoma-in-skin (Mel-RhS) model and were compared in terms of dermal invasion and immune modulatory and pro-angiogenic capabilities. A375 displayed the most invasive phenotype by clearly expanding into the dermal compartment, whereas COLO829, G361, MeWo, and SK-MEL-28 recapitulated to different extent the initial stages of melanoma invasion. No nest formation was observed for RPMI-7951. Notably, the integration of A375 and SK-MEL-28 cells into the model resulted in an increased secretion of immune modulatory factors (e.g., M-CSF, IL-10, and TGFβ) and pro-angiogenic factors (e.g., Flt-1 and VEGF). Mel-RhS-derived supernatants induced endothelial cell sprouting in vitro. In addition, observed A375-RhS tissue contraction was correlated to increased TGFβ release and α-SMA expression, all indicative of differentiation of fibroblasts into cancer-associated fibroblast-like cells and reminiscent of epithelial-to-mesenchymal transition, consistent with A375's most prominent invasive behavior. In conclusion, we successfully generated several Mel-RhS models mimicking different stages of melanoma progression, which can be further tailored for future studies to investigate individual aspects of the disease and serve as three-dimensional models to assess efficacy of therapeutic strategies.
Collapse
Affiliation(s)
- Elisabetta Michielon
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location Vrije Universiteit, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
| | - Marta López González
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| | - Dorian A Stolk
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| | - Joeke G C Stolwijk
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| | - Sanne Roffel
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location Vrije Universiteit, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| | - Taco Waaijman
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location Vrije Universiteit, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
| | - Sinéad M Lougheed
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location Vrije Universiteit, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, 1105 AZ Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
10
|
Pereira B, Duque K, Ramos-Gonzalez G, Díaz-Solano D, Wittig O, Zamora M, Gledhill T, Cardier JE. Wound healing by transplantation of mesenchymal stromal cells loaded on polyethylene terephthalate scaffold: Implications for skin injury treatment. Injury 2023; 54:1071-1081. [PMID: 36801131 DOI: 10.1016/j.injury.2023.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/25/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND Several clinical studies have shown that cellular therapy based on mesenchymal stromal cells (MSCs) transplantation may accelerate wound healing. One major challenge is the delivery system used for MSCs transplantation. In this work, we evaluated the capacity of a scaffold based on polyethylene terephthalate (PET) to maintain the viability and biological functions of MSCs, in vitro. We examined the capacity of MSCs loaded on PET (MSCs/PET) to induce wound healing in an experimental model of full-thickness wound. METHODS Human MSCs were seeded and cultured on PET membranes at 37 °C for 48 h. Adhesion, viability, proliferation, migration, multipotential differentiation and chemokine production were evaluated in cultures of MSCs/PET. The possible therapeutic effect of MSCs/PET on the re-epithelialization of full thickness wounds was examined at day 3 post-wounding in C57BL/6 mice. Histological and immunohistochemical (IH) studies were performed to evaluate wound re-epithelialization and the presence of epithelial progenitor cells (EPC). As controls, wounds without treatment or treated with PET were established. RESULTS We observed MSCs adhered to PET membranes and maintained their viability, proliferation and migration. They preserved their multipotential capacity of differentiation and ability of chemokine production. MSCs/PET implants promoted an accelerated wound re-epithelialization, after three days post-wounding. It was associated with the presence of EPC Lgr6+ and K6+. DISCUSSION Our results show that MSCs/PET implants induce a rapid re-epithelialization of deep- and full-thickness wounds. MSCs/PET implants constitute a potential clinical therapy for treating cutaneous wounds.
Collapse
Affiliation(s)
- Betzabeth Pereira
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela; Laboratorio de Neurofarmacología Celular, Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela
| | - Kharelys Duque
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela
| | - Giselle Ramos-Gonzalez
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela
| | - Dylana Díaz-Solano
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela
| | - Olga Wittig
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela
| | - Mariela Zamora
- Departamento de Dermatologia, Hospital Militar "Dr Carlos Arvelo, Venezuela
| | - Teresa Gledhill
- Servicio de Anatomía Patológica, Hospital Vargas, Caracas 1010-A, Venezuela
| | - José E Cardier
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela.
| |
Collapse
|
11
|
Hoch M, Smita S, Cesnulevicius K, Schultz M, Lescheid D, Wolkenhauer O, Gupta S. Network analyses reveal new insights into the effect of multicomponent Tr14 compared to single-component diclofenac in an acute inflammation model. J Inflamm (Lond) 2023; 20:12. [PMID: 36973809 PMCID: PMC10044762 DOI: 10.1186/s12950-023-00335-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 02/21/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Modifying the acute inflammatory response has wide clinical benefits. Current options include non-steroidal anti-inflammatory drugs (NSAIDs) and therapies that may resolve inflammation. Acute inflammation involves multiple cell types and various processes. We, therefore, investigated whether an immunomodulatory drug that acts simultaneously at multiple sites shows greater potential to resolve acute inflammation more effectively and with fewer side effects than a common anti-inflammatory drug developed as a small molecule for a single target. In this work, we used time-series gene expression profiles from a wound healing mouse model to compare the effects of Traumeel (Tr14), a multicomponent natural product, to diclofenac, a single component NSAID on inflammation resolution. RESULTS We advance previous studies by mapping the data onto the "Atlas of Inflammation Resolution", followed by in silico simulations and network analysis. We found that Tr14 acts primarily on the late phase of acute inflammation (during resolution) compared to diclofenac, which suppresses acute inflammation immediately after injury. CONCLUSIONS Our results provide new insights how network pharmacology of multicomponent drugs may support inflammation resolution in inflammatory conditions.
Collapse
Affiliation(s)
- Matti Hoch
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, 18055, Germany
| | - Suchi Smita
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, 18055, Germany
| | | | | | | | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, 18055, Germany
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Freising, 85354, Germany
- Stellenbosch Institute of Advanced Study, Wallenberg Research Centre, Stellenbosch University, Stellenbosch, 7602, South Africa
| | - Shailendra Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, 18055, Germany.
| |
Collapse
|
12
|
Duddu S, Bhattacharya A, Chakrabarti R, Chakravorty N, Shukla PC. Regeneration and Tissue Microenvironment. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
13
|
CXCL4 drives fibrosis by promoting several key cellular and molecular processes. Cell Rep 2022; 38:110189. [PMID: 34986347 DOI: 10.1016/j.celrep.2021.110189] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 02/28/2021] [Accepted: 12/08/2021] [Indexed: 01/05/2023] Open
Abstract
Fibrosis is a major cause of mortality worldwide, characterized by myofibroblast activation and excessive extracellular matrix deposition. Systemic sclerosis is a prototypic fibrotic disease in which CXCL4 is increased and strongly correlates with skin and lung fibrosis. Here we aim to elucidate the role of CXCL4 in fibrosis development. CXCL4 levels are increased in multiple inflammatory and fibrotic mouse models, and, using CXCL4-deficient mice, we demonstrate the essential role of CXCL4 in promoting fibrotic events in the skin, lungs, and heart. Overexpressing human CXCL4 in mice aggravates, whereas blocking CXCL4 reduces, bleomycin-induced fibrosis. Single-cell ligand-receptor analysis predicts CXCL4 to affect endothelial cells and fibroblasts. In vitro, we confirm that CXCL4 directly induces myofibroblast differentiation and collagen synthesis in different precursor cells, including endothelial cells, by stimulating endothelial-to-mesenchymal transition. Our findings identify a pivotal role of CXCL4 in fibrosis, further substantiating the potential role of neutralizing CXCL4 as a therapeutic strategy.
Collapse
|
14
|
Abstract
The introduction of antiretroviral therapy (ART) and highly active antiretroviral therapy (HAART) has transformed human immunodeficiency virus (HIV)-1 into a chronic, well-managed disease. However, these therapies do not eliminate all infected cells from the body despite suppressing viral load. Viral rebound is largely due to the presence of cellular reservoirs which support long-term persistence of HIV-1. A thorough understanding of the HIV-1 reservoir will facilitate the development of new strategies leading to its detection, reduction, and elimination, ultimately leading to curative therapies for HIV-1. Although immune cells derived from lymphoid and myeloid progenitors have been thoroughly studied as HIV-1 reservoirs, few studies have examined whether mesenchymal stromal/stem cells (MSCs) can assume this function. In this review, we evaluate published studies which have assessed whether MSCs contribute to the HIV-1 reservoir. MSCs have been found to express the receptors and co-receptors required for HIV-1 entry, albeit at levels of expression and receptor localisation that vary considerably between studies. Exposure to HIV-1 and HIV-1 proteins alters MSC properties in vitro, including their proliferation capacity and differentiation potential. However, in vitro and in vivo experiments investigating whether MSCs can become infected with and harbour latent integrated proviral DNA are lacking. In conclusion, MSCs appear to have the potential to contribute to the HIV-1 reservoir. However, further studies are needed using techniques such as those used to prove that cluster of differentiation (CD)4+ T cells constitute an HIV-1 reservoir before a reservoir function can definitively be ascribed to MSCs.
Collapse
|
15
|
Savitri C, Kwon JW, Drobyshava V, Ha SS, Park K. M2 Macrophage-Derived Concentrated Conditioned Media Significantly Improves Skin Wound Healing. Tissue Eng Regen Med 2021; 19:617-628. [PMID: 34962626 PMCID: PMC9130431 DOI: 10.1007/s13770-021-00414-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Macrophages, with many different phenotypes play a major role during wound healing process, secreting the cytokines crucial to angiogenesis, cell recruitment and ECM remodeling. Therefore, macrophage-derived cytokines may be attractive therapeutic resource for wound healing. METHODS To obtain a conditioned media (CM) from macrophages, human monocyte THP-1 cells were seeded on TCP or human fibroblast-derived matrix (hFDM) and they were differentiated into M1 or M2 phenotype using distinct protocols. A combination of different substrates and macrophage phenotypes produced M1- and M2-CM or M1-hFDM- and M2-hFDM-CM, respectively. Proteome microarray determines the cytokine contents in those CMs. CMs-treated human dermal fibroblast (hDFB) was analyzed using collagen synthesis and wound scratch assay. Concentrated form of the CM (CCM), obtained by high-speed centrifugation, was administered to a murine full-thickness wound model using alginate patch, where alginate patch was incubated in the M2-CCM overnight at 4 °C before transplantation. On 14 day post-treatment, examination was carried out through H&E and Herovici staining. Keratinocyte and M2 macrophages were also evaluated via immunofluorescence staining. RESULTS Cytokine analysis of CMs found CCL1, CCL5, and G-CSF, where CCL5 is more dominant. We found increased collagen synthesis and faster wound closure in hDFB treated with M2-CM. Full-thickness wounds treated by M2-hFDM-CCM containing alginate patch showed early wound closure, larger blood vessels, increased mature collagen deposition, enhanced keratinocyte maturation and more M2-macrophage population. CONCLUSION Our study demonstrated therapeutic potential of the CM derived from M2 macrophages, where the cytokines in the CM may have played an active role for enhanced wound healing.
Collapse
Affiliation(s)
- Cininta Savitri
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea ,Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792 Republic of Korea
| | - Jae Won Kwon
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea ,Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792 Republic of Korea
| | - Valeryia Drobyshava
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea ,Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792 Republic of Korea
| | - Sang Su Ha
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea ,Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792 Republic of Korea
| |
Collapse
|
16
|
Torrecillas-Baena B, Gálvez-Moreno MÁ, Quesada-Gómez JM, Dorado G, Casado-Díaz A. Influence of Dipeptidyl Peptidase-4 (DPP4) on Mesenchymal Stem-Cell (MSC) Biology: Implications for Regenerative Medicine - Review. Stem Cell Rev Rep 2021; 18:56-76. [PMID: 34677817 DOI: 10.1007/s12015-021-10285-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2021] [Indexed: 12/16/2022]
Abstract
Dipeptidyl peptidase IV (DPP4) is a ubiquitous protease that can be found in membrane-anchored or soluble form. Incretins are one of the main DPP4 substrates. These hormones regulate glucose levels, by stimulating insulin secretion and decreasing glucagon production. Because DPP4 levels are high in diabetes, DPP4 inhibitor (DPP4i) drugs derived from gliptin are widespread used as hypoglycemic agents for its treatment. However, as DPP4 recognizes other substrates such as chemokines, growth factors and neuropeptides, pleiotropic effects have been observed in patients treated with DPP4i. Several of these substrates are part of the stem-cell niche. Thus, they may affect different physiological aspects of mesenchymal stem-cells (MSC). They include viability, differentiation, mobilization and immune response. MSC are involved in tissue homeostasis and regeneration under both physiological and pathological conditions. Therefore, such cells and their secretomes have a high clinical potential in regenerative medicine. In this context, DPP4 activity may modulate different aspects of MSC regenerative capacity. Therefore, the aim of this review is to analyze the effect of different DPP4 substrates on MSC. Likewise, how the regulation of DPP4 activity by DPP4i can be applied in regenerative medicine. That includes treatment of cardiovascular and bone pathologies, cutaneous ulcers, organ transplantation and pancreatic beta-cell regeneration, among others. Thus, DPP4i has an important clinical potential as a complement to therapeutic strategies in regenerative medicine. They involve enhancing the differentiation, immunomodulation and mobilization capacity of MSC for regenerative purposes.
Collapse
Affiliation(s)
- Bárbara Torrecillas-Baena
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - María Ángeles Gálvez-Moreno
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - José Manuel Quesada-Gómez
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - Gabriel Dorado
- Dep. Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, CIBERFES, 14071, Córdoba, Spain
| | - Antonio Casado-Díaz
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain.
| |
Collapse
|
17
|
Cuesta-Gomez N, Graham GJ, Campbell JDM. Chemokines and their receptors: predictors of the therapeutic potential of mesenchymal stromal cells. J Transl Med 2021; 19:156. [PMID: 33865426 PMCID: PMC8052819 DOI: 10.1186/s12967-021-02822-5] [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: 12/21/2020] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) are promising cellular therapeutics for the treatment of inflammatory and degenerative disorders due to their anti-inflammatory, immunomodulatory and regenerative potentials. MSCs can be sourced from a variety of tissues within the body, but bone marrow is the most frequently used starting material for clinical use. The chemokine family contains many regulators of inflammation, cellular function and cellular migration-all critical factors in understanding the potential potency of a novel cellular therapeutic. In this review, we focus on expression of chemokine receptors and chemokine ligands by MSCs isolated from different tissues. We discuss the differential migratory, angiogenetic and immunomodulatory potential to understand the role that tissue source of MSC may play within a clinical context. Furthermore, this is strongly associated with leukocyte recruitment, immunomodulatory potential and T cell inhibition potential and we hypothesize that chemokine profiling can be used to predict the in vivo therapeutic potential of MSCs isolated from new sources and compare them to BM MSCs.
Collapse
Affiliation(s)
- Nerea Cuesta-Gomez
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Gerard J Graham
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - John D M Campbell
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK. .,Tissues, Cells and Advanced Therapeutics, Scottish National Blood Transfusion Service, The Jack Copland Centre, Research Avenue North, Edinburgh, UK.
| |
Collapse
|
18
|
Sierra-Sánchez Á, Montero-Vilchez T, Quiñones-Vico MI, Sanchez-Diaz M, Arias-Santiago S. Current Advanced Therapies Based on Human Mesenchymal Stem Cells for Skin Diseases. Front Cell Dev Biol 2021; 9:643125. [PMID: 33768095 PMCID: PMC7985058 DOI: 10.3389/fcell.2021.643125] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/18/2021] [Indexed: 12/17/2022] Open
Abstract
Skin disease may be related with immunological disorders, external aggressions, or genetic conditions. Injuries or cutaneous diseases such as wounds, burns, psoriasis, and scleroderma among others are common pathologies in dermatology, and in some cases, conventional treatments are ineffective. In recent years, advanced therapies using human mesenchymal stem cells (hMSCs) from different sources has emerged as a promising strategy for the treatment of many pathologies. Due to their properties; regenerative, immunomodulatory and differentiation capacities, they could be applied for the treatment of cutaneous diseases. In this review, a total of thirteen types of hMSCs used as advanced therapy have been analyzed, considering the last 5 years (2015-2020). The most investigated types were those isolated from umbilical cord blood (hUCB-MSCs), adipose tissue (hAT-MSCs) and bone marrow (hBM-MSCs). The most studied diseases were wounds and ulcers, burns and psoriasis. At preclinical level, in vivo studies with mice and rats were the main animal models used, and a wide range of types of hMSCs were used. Clinical studies analyzed revealed that cell therapy by intravenous administration was the advanced therapy preferred except in the case of wounds and burns where tissue engineering was also reported. Although in most of the clinical trials reviewed results have not been posted yet, safety was high and only local slight adverse events (mild nausea or abdominal pain) were reported. In terms of effectiveness, it was difficult to compare the results due to the different doses administered and variables measured, but in general, percentage of wound's size reduction was higher than 80% in wounds, Psoriasis Area and Severity Index and Severity Scoring for Atopic Dermatitis were significantly reduced, for scleroderma, parameters such as Modified Rodnan skin score (MRSC) or European Scleroderma Study Group activity index reported an improvement of the disease and for hypertrophic scars, Vancouver Scar Scale (VSS) score was decreased after applying these therapies. On balance, hMSCs used for the treatment of cutaneous diseases is a promising strategy, however, the different experimental designs and endpoints stablished in each study, makes necessary more research to find the best way to treat each patient and disease.
Collapse
Affiliation(s)
- Álvaro Sierra-Sánchez
- Cell Production and Tissue Engineering Unit, Andalusian Network of Design and Translation of Advanced Therapies, Virgen de las Nieves University Hospital, Granada, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada, Spain
| | - Trinidad Montero-Vilchez
- Biosanitary Institute of Granada (ibs.GRANADA), Granada, Spain.,Department of Dermatology, Virgen de las Nieves University Hospital, Granada, Spain
| | - María I Quiñones-Vico
- Cell Production and Tissue Engineering Unit, Andalusian Network of Design and Translation of Advanced Therapies, Virgen de las Nieves University Hospital, Granada, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada, Spain.,Department of Dermatology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Manuel Sanchez-Diaz
- Biosanitary Institute of Granada (ibs.GRANADA), Granada, Spain.,Department of Dermatology, Virgen de las Nieves University Hospital, Granada, Spain
| | - Salvador Arias-Santiago
- Cell Production and Tissue Engineering Unit, Andalusian Network of Design and Translation of Advanced Therapies, Virgen de las Nieves University Hospital, Granada, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada, Spain.,Department of Dermatology, Virgen de las Nieves University Hospital, Granada, Spain.,Department of Dermatology, Faculty of Medicine, University of Granada, Granada, Spain
| |
Collapse
|
19
|
Savcı Y, Kırbaş OK, Bozkurt BT, Abdik EA, Taşlı PN, Şahin F, Abdik H. Grapefruit-derived extracellular vesicles as a promising cell-free therapeutic tool for wound healing. Food Funct 2021; 12:5144-5156. [PMID: 33977960 DOI: 10.1039/d0fo02953j] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Due to the prevalence of individuals suffering from chronic wounds, developing safe and effective wound care agents are one of the more prominent fields of research in biology. However, wound healing is a complex, multi-stage biological process, involving multiple sequences of biological responses from different types of cells, secreted mediators, and extracellular matrix elements. Plants have a long history of use in the treatment of wounds. Plant-derived extracellular vesicles, which are secreted nano vesicle messengers responsible for intercellular communications, show promise as a new, biotechnological wound-care agent. In this study, we assessed the wound healing potential of extracellular vesicles isolated from grapefruits - a plant with well-known anti-inflammatory and wound healing properties. Grapefruit extracellular vesicles (GEVs) increased cell viability and cell migration while reducing intracellular ROS production in a dose-dependent manner in HaCaT cells. Expression of proliferation and migration-related genes were raised by GEV treatment in a dose dependent manner. Additionally, GEV treatment increased the tube formation capabilities of treated HUVEC cells. These findings suggest that GEVs can be used as plant-derived wound healing agents, and have shown potential as a biotechnological agent for wound healing. Further development and study of plant-derived extracellular vesicles may lead to the realization of their full potential.
Collapse
Affiliation(s)
- Yağız Savcı
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Oğuz Kaan Kırbaş
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Batuhan Turhan Bozkurt
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Ezgi Avşar Abdik
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Pakize Neslihan Taşlı
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Hüseyin Abdik
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Istanbul Sabahattin Zaim University, Istanbul, Turkey.
| |
Collapse
|
20
|
Liang W, Chen X, Dong Y, Zhou P, Xu F. Recent advances in biomaterials as instructive scaffolds for stem cells in tissue repair and regeneration. INT J POLYM MATER PO 2020. [DOI: 10.1080/00914037.2020.1848832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, P. R. China
| | - Xuerong Chen
- Department of Orthopaedics, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, P. R. China
| | - Yongqiang Dong
- Department of Orthopaedics, Xinchang People’s Hospital, Shaoxing, P. R. China
| | - Ping Zhou
- Department of Orthopaedics, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, P. R. China
| | - Fangming Xu
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, P. R. China
| |
Collapse
|
21
|
Gwak J, Jeong H, Lee K, Shin JY, Sim T, Na J, Kim J, Ju BG. SFMBT2-Mediated Infiltration of Preadipocytes and TAMs in Prostate Cancer. Cancers (Basel) 2020; 12:E2718. [PMID: 32971847 PMCID: PMC7565541 DOI: 10.3390/cancers12092718] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/07/2020] [Accepted: 08/16/2020] [Indexed: 12/22/2022] Open
Abstract
Infiltration of diverse cell types into tumor microenvironment plays a critical role in cancer progression including metastasis. We previously reported that SFMBT2 (Scm-like with four mbt domains 2) regulates the expression of matrix metalloproteinases (MMPs) and migration and invasion of cancer cells in prostate cancer. Here we investigated whether the down-regulation of SFMBT2 regulates the infiltration of preadipocytes and tumor-associated macrophages (TAMs) in prostate cancer. We found that the down-regulation of SFMBT2 promotes the infiltration of preadipocytes and TAMs through up-regulation of CXCL8, CCL2, CXCL10, and CCL20 expression in prostate cancer. Expression of CXCL8, CCL2, CXCL10, and CCL20 was also elevated in prostate cancer patients having a higher Gleason score (≥8), which had substantially lower SFMBT2 expression. We also found that the up-regulation of CXCL8, CCL2, CXCL10, and CCL20 expression is dependent on NF-κB activation in prostate cancer cells expressing a low level of SFMBT2. Moreover, increased IL-6 from infiltrated preadipocytes and TAMs promoted migration and invasion of prostate cancer cells expressing a low level of SFMBT2. Our study may suggest that SFMBT2 a critical regulator for the infiltration of preadipocytes and TAMs into the prostate tumor microenvironment. Thus, the regulation of SFMBT2 may provide a new therapeutic strategy to inhibit prostate cancer metastasis, and SFMBT2 could be used as a potential biomarker in prostate cancer metastasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Bong-Gun Ju
- Department of Life Science, Sogang University, Seoul 04107, Korea; (J.G.); (H.J.); (K.L.); (J.Y.S.); (T.S.); (J.N.); (J.K.)
| |
Collapse
|
22
|
Rodrigues Neves CT, Spiekstra SW, de Graaf NPJ, Rustemeyer T, Feilzer AJ, Kleverlaan CJ, Gibbs S. Titanium salts tested in reconstructed human skin with integrated MUTZ-3-derived Langerhans cells show an irritant rather than a sensitizing potential. Contact Dermatitis 2020; 83:337-346. [PMID: 32677096 PMCID: PMC7689826 DOI: 10.1111/cod.13666] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The nature of clinically related adverse reactions to titanium is still unknown. OBJECTIVE To determine whether titanium salts have irritant or sensitizing potential in a reconstructed human skin (RHS) model with integrated Langerhans cells (LCs). METHODS RHS-LCs (ie, reconstructed epidermis) containing primary differentiated keratinocytes and CFSE+ CD1a+ -LCs generated from the MUTZ-3 cell line on a primary fibroblast-populated collagen hydrogel (dermis) were topically exposed to titanium(IV) bis(ammonium lactato)dihydroxide (TiALH). LC migration and plasticity were determined. RESULTS TiALH resulted in CFSE+ CD1a+ -LC migration out of the epidermis. Neutralizing antibodies to CCL5 and CXCL12 showed that LC migration was CCL5 and not CXCL12 mediated. LCs accumulating within the dermis after TiALH exposure were CFSE+ Lang+ CD68+ which is characteristic of a phenotypic switch of MUTZ-LC to a macrophage-like cell. Furthermore, TiALH did not result in increased interleukin (IL)-1β or CCR7 messenger RNA (mRNA) in the dermis, but did result in increased IL-10 mRNA. In addition, monocultures of MUTZ-LCs failed to increase LC maturation biomarkers CD83, CD86, and CXCL-8 when exposed to noncytotoxic concentrations of four different titanium salts. CONCLUSION These results classify titanium salts as irritants rather than sensitizers and indicate that titanium implant-related complaints could be due to localized irritant-mediated inflammation arising from leachable agents rather than a titanium metal allergy.
Collapse
Affiliation(s)
- Charlotte T Rodrigues Neves
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sander W Spiekstra
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Niels P J de Graaf
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Oral Cell Biology, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Thomas Rustemeyer
- Department of Dermatology, Amsterdam University Medical Centre (location AMC), Amsterdam, The Netherlands
| | - Albert J Feilzer
- Department of Dental Materials Science, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Cees J Kleverlaan
- Department of Oral Cell Biology, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Oral Cell Biology, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Limandjaja GC, Niessen FB, Scheper RJ, Gibbs S. The Keloid Disorder: Heterogeneity, Histopathology, Mechanisms and Models. Front Cell Dev Biol 2020; 8:360. [PMID: 32528951 PMCID: PMC7264387 DOI: 10.3389/fcell.2020.00360] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/22/2020] [Indexed: 12/22/2022] Open
Abstract
Keloids constitute an abnormal fibroproliferative wound healing response in which raised scar tissue grows excessively and invasively beyond the original wound borders. This review provides a comprehensive overview of several important themes in keloid research: namely keloid histopathology, heterogeneity, pathogenesis, and model systems. Although keloidal collagen versus nodules and α-SMA-immunoreactivity have been considered pathognomonic for keloids versus hypertrophic scars, conflicting results have been reported which will be discussed together with other histopathological keloid characteristics. Importantly, histopathological keloid abnormalities are also present in the keloid epidermis. Heterogeneity between and within keloids exists which is often not considered when interpreting results and may explain discrepancies between studies. At least two distinct keloid phenotypes exist, the superficial-spreading/flat keloids and the bulging/raised keloids. Within keloids, the periphery is often seen as the actively growing margin compared to the more quiescent center, although the opposite has also been reported. Interestingly, the normal skin directly surrounding keloids also shows partial keloid characteristics. Keloids are most likely to occur after an inciting stimulus such as (minor and disproportionate) dermal injury or an inflammatory process (environmental factors) at a keloid-prone anatomical site (topological factors) in a genetically predisposed individual (patient-related factors). The specific cellular abnormalities these various patient, topological and environmental factors generate to ultimately result in keloid scar formation are discussed. Existing keloid models can largely be divided into in vivo and in vitro systems including a number of subdivisions: human/animal, explant/culture, homotypic/heterotypic culture, direct/indirect co-culture, and 3D/monolayer culture. As skin physiology, immunology and wound healing is markedly different in animals and since keloids are exclusive to humans, there is a need for relevant human in vitro models. Of these, the direct co-culture systems that generate full thickness keloid equivalents appear the most promising and will be key to further advance keloid research on its pathogenesis and thereby ultimately advance keloid treatment. Finally, the recent change in keloid nomenclature will be discussed, which has moved away from identifying keloids solely as abnormal scars with a purely cosmetic association toward understanding keloids for the fibroproliferative disorder that they are.
Collapse
Affiliation(s)
- Grace C. Limandjaja
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center (location VUmc), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Frank B. Niessen
- Department of Plastic Surgery, Amsterdam University Medical Center (location VUmc), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Rik J. Scheper
- Department of Pathology, Amsterdam University Medical Center (location VUmc), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center (location VUmc), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
24
|
Vahav I, van den Broek LJ, Thon M, Monsuur HN, Spiekstra SW, Atac B, Scheper RJ, Lauster R, Lindner G, Marx U, Gibbs S. Reconstructed human skin shows epidermal invagination towards integrated neopapillae indicating early hair follicle formation in vitro. J Tissue Eng Regen Med 2020; 14:761-773. [PMID: 32293116 PMCID: PMC7317351 DOI: 10.1002/term.3039] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 03/02/2020] [Accepted: 03/23/2020] [Indexed: 01/06/2023]
Abstract
Application of reconstructed human Skin (RhS) is a promising approach for the treatment of extensive wounds and for drug efficacy and safety testing. However, incorporating appendages, such as hair follicles, into RhS still remains a challenge. The hair follicle plays a critical role in thermal regulation, dispersion of sweat and sebum, sensory and tactile functions, skin regeneration, and repigmentation. The aim of this study was to determine whether human neopapilla could be incorporated into RhS (differentiated epidermis on fibroblast and endothelial cell populated dermis) and whether the neopapillae maintain their inductive follicular properties in vitro. Neopapillae spheroids, constructed from expanded and self‐aggregating dermal papilla cells, synthesized extracellular matrix typically found in follicular papillae. Compared with dermal fibroblasts, neopapillae showed increased expression of multiple genes (Wnt5a, Wnt10b, and LEF1) known to regulate hair development and also increased secretion of CXCL1, which is a strong keratinocyte chemoattractant. When neopapillae were incorporated into the dermis of RhS, they stimulated epidermal down‐growth resulting in engulfment of the neopapillae sphere. Similar to the native hair follicle, the differentiated invaginating epidermis inner side was keratin 10 positive and the undifferentiated outer side keratin 10 negative. The outer side was keratin 15 positive confirming the undifferentiated nature of these keratinocytes aligning a newly formed collagen IV, laminin V positive basement membrane within the hydrogel. In conclusion, we describe a RhS model containing neopapillae with hair follicle‐inductive properties. Importantly, epidermal invagination occurred to engulf the neopapillae, thus demonstrating in vitro the first steps towards hair follicle morphogenesis in RhS.
Collapse
Affiliation(s)
- Irit Vahav
- TissUse GmbH, Berlin, Germany.,Department of Molecular Cell Biology and Immunology, Amsterdam Movement Sciences, VU University Medical Centre, Amsterdam UMC, Amsterdam, The Netherlands
| | - Lenie J van den Broek
- Department of Molecular Cell Biology and Immunology, Amsterdam Movement Sciences, VU University Medical Centre, Amsterdam UMC, Amsterdam, The Netherlands.,A-Skin BV, Amsterdam, The Netherlands
| | - Maria Thon
- Department of Molecular Cell Biology and Immunology, Amsterdam Movement Sciences, VU University Medical Centre, Amsterdam UMC, Amsterdam, The Netherlands
| | - Hanneke N Monsuur
- Department of Molecular Cell Biology and Immunology, Amsterdam Movement Sciences, VU University Medical Centre, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sander W Spiekstra
- Department of Molecular Cell Biology and Immunology, Amsterdam Movement Sciences, VU University Medical Centre, Amsterdam UMC, Amsterdam, The Netherlands
| | - Beren Atac
- TissUse GmbH, Berlin, Germany.,Department of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | | | - Roland Lauster
- Department of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Gerd Lindner
- TissUse GmbH, Berlin, Germany.,Department of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | | | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam Movement Sciences, VU University Medical Centre, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Dhoke NR, Kaushik K, Das A. Cxcr6-Based Mesenchymal Stem Cell Gene Therapy Potentiates Skin Regeneration in Murine Diabetic Wounds. Mol Ther 2020; 28:1314-1326. [PMID: 32112713 DOI: 10.1016/j.ymthe.2020.02.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 01/14/2020] [Accepted: 02/10/2020] [Indexed: 01/09/2023] Open
Abstract
Mesenchymal stem cell (MSC) therapies for wound healing are often compromised due to low recruitment and engraftment of transplanted cells, as well as delayed differentiation into cell lineages for skin regeneration. An increased expression of chemokine ligand CXCL16 in wound bed and its cognate receptor, CXCR6, on murine bone-marrow-derived MSCs suggested a putative therapeutic relevance of exogenous MSC transplantation therapy. Induction of the CXCL16-CXCR6 axis led to activation of focal adhesion kinase (FAK), Src, and extracellular signal-regulated kinases 1/2 (ERK1/2)-mediated matrix metalloproteinases (MMP)-2 promoter regulation and expression, the migratory signaling pathways in MSC. CXCL16 induction also increased the transdifferentiation of MSCs into endothelial-like cells and keratinocytes. Intravenous transplantation of allogenic stable MSCs with Cxcr6 gene therapy potentiated skin tissue regeneration by increasing recruitment and engraftment as well as neovascularization and re-epithelialization at the wound site in excisional splinting wounds of type I and II diabetic mice. This study suggests that activation of the CXCL16-CXCR6 axis in bioengineered MSCs with Cxcr6 overexpression provides a promising therapeutic approach for the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Neha R Dhoke
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Hyderabad 500 007, TS, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IICT Campus, Hyderabad 500 007, TS, India
| | - Komal Kaushik
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Hyderabad 500 007, TS, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IICT Campus, Hyderabad 500 007, TS, India
| | - Amitava Das
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Hyderabad 500 007, TS, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IICT Campus, Hyderabad 500 007, TS, India.
| |
Collapse
|
26
|
Caseiro AR, Santos Pedrosa S, Ivanova G, Vieira Branquinho M, Almeida A, Faria F, Amorim I, Pereira T, Maurício AC. Mesenchymal Stem/ Stromal Cells metabolomic and bioactive factors profiles: A comparative analysis on the umbilical cord and dental pulp derived Stem/ Stromal Cells secretome. PLoS One 2019; 14:e0221378. [PMID: 31774816 PMCID: PMC6881058 DOI: 10.1371/journal.pone.0221378] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/21/2019] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal Stem/ Stromal Cells assume a supporting role to the intrinsic mechanisms of tissue regeneration, a feature mostly assigned to the contents of their secretome. A comparative study on the metabolomic and bioactive molecules/factors content of the secretome of Mesenchymal Stem/ Stromal Cells derived from two expanding sources: the umbilical cord stroma and the dental pulp is presented and discussed. The metabolic profile (Nuclear Magnetic Resonance Spectroscopy) evidenced some differences in the metabolite dynamics through the conditioning period, particularly on the glucose metabolism. Despite, overall similar profiles are suggested. More prominent differences are highlighted for the bioactive factors (Multiplexing Laser Bear Analysis), in which Follistatin, Growth Regulates Protein, Hepatocyte Growth Factor, Interleukin-8 and Monocyte Chemotactic Protein-1 dominate in Umbilical Cord Mesenchymal Stem/ Stromal Cells secretion, while in Dental Pulp Stem/ Stromal Cells the Vascular Endothelial Growth Factor-A and Follistatin are more evident. The distinct secretory cocktail did not result in significantly different effects on endothelial cell populations dynamics including proliferation, migration, tube formation capacity and in vivo angiogenesis, or in chemotaxis for both Mesenchymal Stem/ Stromal Cells populations.
Collapse
Affiliation(s)
- Ana Rita Caseiro
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado, Porto, Portugal
- Escola Universitária Vasco da Gama (EUVG), Lordemão, Coimbra, Portugal
| | - Sílvia Santos Pedrosa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado, Porto, Portugal
| | - Galya Ivanova
- REQUIMTE- LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, Porto, Portugal
| | - Mariana Vieira Branquinho
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado, Porto, Portugal
| | - André Almeida
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado, Porto, Portugal
- Indústria Transformadora de Subprodutos—I.T.S, SA, Grupo ETSA, Rua Padre Adriano, Olivais do Machio, Santo Antão do Tojal, Loures, Portugal
| | - Fátima Faria
- Departamento de Patologia e Imunologia Molecular, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
| | - Irina Amorim
- Departamento de Patologia e Imunologia Molecular, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
- i3S - Instituto de Investigação e Inovação da Universidade do Porto, Rua Alfredo Allen, Porto, Portugal
| | - Tiago Pereira
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado, Porto, Portugal
| | - Ana Colette Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado, Porto, Portugal
- * E-mail: ,
| |
Collapse
|
27
|
Ma H, Duan S, Yan F, Yang H, Cao Y, Ge L, Gao R. Enhancer of zeste homolog 2 enhances the migration and chemotaxis of dental mesenchymal stem cells. J Int Med Res 2019; 48:300060519882149. [PMID: 31642363 PMCID: PMC7262854 DOI: 10.1177/0300060519882149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Objective To investigate the function of enhancer of zeste homolog 2 (EZH2) in the migration and chemotaxis of human dental tissue-derived mesenchymal stem cells. Methods The expression of EZH2, C-X-C motif chemokine ligand 11 (CXCL11), CXCL16, and CXCR1 in stem cells from the apical papilla (SCAPs) was determined by real-time reverse transcription PCR and western blotting. The effects of EZH2 on the homing of SCAPs and the effects of EZH2-overexpressing SCAP culture supernatant on periodontal ligament stem cells (PDLSCs) were tested by scratch migration assays and transwell chemotaxis assays. Results EZH2 overexpression significantly enhanced the migration and chemotaxis of SCAPs and upregulated the expression of CXCL11, CXCL16, and CXCR1 in SCAPs. EZH2 depletion had the opposite effect, impairing the migration and chemotaxis of SCAPs and downregulating the expression of CXCL11, CXCL16, and CXCR1. The culture supernatant of EZH2-overexpressing SCAPs advanced the migration and chemotaxis of PDLSCs. Conclusions EZH2 evidently promoted the migration and chemotaxis of SCAPs by upregulating the expression of CXCL11, CXCL16, and CXCR1. Moreover, EZH2-overexpressing SCAPs enhanced the homing, migration, and chemotaxis of PDLSCs via paracrine signaling.
Collapse
Affiliation(s)
- Huarui Ma
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shaoyu Duan
- Department of Stomatology, Beijing Electric Power Hospital, Beijing, China
| | - Fei Yan
- Xiangya Stomatology Hospital and School of Stomatology, Central South University, Changsha, Hunan, China
| | - Haoqing Yang
- Capital Medical University School of Stomatology, No. 4 Tian Tan Xi Li, Beijing, China
| | - Yangyang Cao
- Capital Medical University School of Stomatology, No. 4 Tian Tan Xi Li, Beijing, China
| | - Lihua Ge
- Capital Medical University School of Stomatology, No. 4 Tian Tan Xi Li, Beijing, China
| | - Runtao Gao
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Collagen I Promotes Adipocytogenesis in Adipose-Derived Stem Cells In Vitro. Cells 2019; 8:cells8040302. [PMID: 30939867 PMCID: PMC6523348 DOI: 10.3390/cells8040302] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 01/19/2023] Open
Abstract
A hallmark of ageing is the redistribution of body fat. Particularly, subcutaneous fat decreases paralleled by a decrease of skin collagen I are typical for age-related skin atrophy. In this paper, we hypothesize that collagen I may be a relevant molecule stimulating the differentiation of adipose-derived stem cells (ASCs) into adipocytes augmenting subcutaneous fat. In this context lipogenesis, adiponectin, and collagen I receptor expression were determined. Freshly isolated ASCs were characterized by stemness-associated surface markers by FACS analysis and then transdifferentiated into adipocytes by specific medium supplements. Lipogenesis was evaluated using Nile Red staining and documented by fluorescence microscopy or quantitatively measured by using a multiwell spectrofluorometer. Expression of adiponectin was measured by real-time RT-PCR and in cell-free supernatants by ELISA, and expression of collagen I receptors was observed by western blot analysis. It was found that supports coated with collagen I promote cell adhesion and lipogenesis of ASCs. Interestingly, a reverse correlation to adiponectin expression was observed. Moreover, we found upregulation of the collagen receptor, discoidin domain-containing receptor 2; receptors of the integrin family were absent or downregulated. These findings indicate that collagen I is able to modulate lipogenesis and adiponectin expression and therefore may contribute to metabolic dysfunctions associated with ageing.
Collapse
|
29
|
Versteeg B, van den Broek LJ, Bruisten SM, Mullender M, de Vries HJC, Gibbs S. An Organotypic Reconstructed Human Urethra to Study Chlamydia trachomatis Infection. Tissue Eng Part A 2018; 24:1663-1671. [PMID: 29792385 DOI: 10.1089/ten.tea.2017.0511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Organotypic models to investigate host-microbiome interactions are still a challenge for the field of tissue engineering. This is particularly the case for organs such as the urethra. Several cell line, animal, and tissue models are available to study Chlamydia trachomatis infections, but none fully reflects natural infection in native human tissue. Therefore, we developed an organotypic reconstructed human urethral model (RhU) to study invasive and noninvasive strains of C. trachomatis. Primary urethra cells were used to reconstruct epithelium on a fibroblast populated collagen-fibrin hydrogel, yielding a RhU. Immunohistochemistry was used to compare RhU with native urethral tissue and to visualize the location of C. trachomatis bacteria in RhU after 10-day exposure. RhU closely resembled native urethral tissue with respect to proliferation and differentiation markers (keratins 6, 10, 13, 17, involucrin, SKALP [skin-derived antileucoproteinase], vimentin, and CD31). Exposure of RhU to noninvasive and invasive C. trachomatis strains revealed relevant differences in infection ability because inclusions were observed (indicating active infection) in the epithelial layer after 10 days exposure only to the invasive strain. The noninvasive strain remained localized on the surface of the epithelial layer. Human primary urethral fibroblasts and keratinocytes can be used to construct RhU that closely resembles native tissue and can be used to investigate active C. trachomatis infections. RhU provides a promising model to investigate host-microbiome interactions such as, but not limited to, the human pathogenesis of C. trachomatis.
Collapse
Affiliation(s)
- Bart Versteeg
- 1 Department of Infectious Diseases, Public Health Service Amsterdam , Amsterdam, The Netherlands
| | - Lenie J van den Broek
- 2 Department of Molecular Cell Biology and Immunology, VU University Medical Center (VUMC) , Amsterdam, The Netherlands
| | - Sylvia M Bruisten
- 1 Department of Infectious Diseases, Public Health Service Amsterdam , Amsterdam, The Netherlands .,3 Amsterdam Infection and Immunity Institute, Academic Medical Center, University of Amsterdam , Amsterdam, The Netherlands
| | - Margriet Mullender
- 4 Department of Plastic, Reconstructive and Hand Surgery, VU University Medical Center (VUMC) , Amsterdam, The Netherlands
| | - Henry J C de Vries
- 1 Department of Infectious Diseases, Public Health Service Amsterdam , Amsterdam, The Netherlands .,3 Amsterdam Infection and Immunity Institute, Academic Medical Center, University of Amsterdam , Amsterdam, The Netherlands .,5 Department of Dermatology, Academic Medical Center, University of Amsterdam , Amsterdam, The Netherlands
| | - Susan Gibbs
- 2 Department of Molecular Cell Biology and Immunology, VU University Medical Center (VUMC) , Amsterdam, The Netherlands .,6 Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University , Amsterdam, The Netherlands
| |
Collapse
|
30
|
Impaired cutaneous T-cell attracting chemokine elevation and adipose-derived stromal cell migration in a high-glucose environment cause poor diabetic wound healing. Kaohsiung J Med Sci 2018; 34:539-546. [PMID: 30309481 DOI: 10.1016/j.kjms.2018.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/26/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022] Open
Abstract
Diabetic wound care is a major health care concern. The major cause of non-healing of wounds in patients with diabetes mellitus (DM) patients mainly involves poor glycemic control, which hinders the migration of progenitor cells including mesenchymal stem cells to the wound site. In this study, we introduced adipose-derived stromal cells (ADSCs) into wound sites and demonstrated that the local transplantation of ADSCs accelerated DM-related wound healing. Furthermore, the migration ability of ADSCs, which diminishes in a high-glucose environment, was partially restored by the exogenous replenishment of the cutaneous T-cell attracting chemokine (CTACK/CCL27). Our findings suggest that CTACK is a potential novel therapeutic target in DM-related wound healing.
Collapse
|
31
|
Petry L, Kippenberger S, Meissner M, Kleemann J, Kaufmann R, Rieger UM, Wellenbrock S, Reichenbach G, Zöller N, Valesky E. Directing adipose-derived stem cells into keratinocyte-like cells: impact of medium composition and culture condition. J Eur Acad Dermatol Venereol 2018; 32:2010-2019. [PMID: 29705993 DOI: 10.1111/jdv.15010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/22/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Adipose-derived stem cells (ASC) are known to transdifferentiate into a wide range of different cell species in vitro including along the epidermal lineage. This property makes them a promising tool for regenerative medicine to restore the epidermal barrier. OBJECTIVE This study is dedicated to identify in vitro conditions enabling transdifferentiation to a keratinocyte-like phenotype. In particular, the impact of different culture conditions (media compositions, 2D, 3D cultures) and extracellular matrix (ECM) molecules was evaluated. METHODS Adipose-derived stem cells derived from subcutaneous abdominal fat were characterized by stemness-associated markers and subjected to different media. Epithelial differentiation in 2D cultures was monitored by pan-cytokeratin expression using flow cytometry and immunocytochemistry. To evaluate the impact of different ECM molecules on epidermal stratification, 3D cultures were produced, lifted to the air-liquid interface (ALI) and examined by histological analysis and quantitative real-time RT-PCR. RESULTS We identified a medium composition containing retinoic acid, hydrocortisone, ascorbic acid and BMP-4 enabling maximum pan-cytokeratin expression in 2D cultures. Moreover, adhesion to type IV collagen further promotes the pan-cytokeratin expression. When cultures were lifted to the ALI, significant stratification was observed, particularly in supports coated with type IV collagen or fibronectin. Moreover, epidermal differentiation markers (involucrin, cytokeratin 1 and 14) become induced. CONCLUSION Conditions with hampered wound healing such as non-healing ulcers demand new treatment regimes. The here introduced optimized protocols for transdifferentiation of ASC into keratinocyte-like cells may help to establish more effective treatment procedures.
Collapse
Affiliation(s)
- L Petry
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - S Kippenberger
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - M Meissner
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - J Kleemann
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - R Kaufmann
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - U M Rieger
- Department of Plastic & Aesthetic, Reconstructive & Hand Surgery, AGAPLESION Markus Hospital, Frankfurt/Main, Germany
| | - S Wellenbrock
- Department of Plastic & Aesthetic, Reconstructive & Hand Surgery, AGAPLESION Markus Hospital, Frankfurt/Main, Germany
| | - G Reichenbach
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - N Zöller
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| | - E Valesky
- Dermatology, Venereology and Allergology, Clinic of the Johann Wolfgang Goethe University, Frankfurt/Main, Germany
| |
Collapse
|
32
|
Monsuur HN, van den Broek LJ, Koolwijk P, Niessen FB, Gibbs S. Endothelial cells enhance adipose mesenchymal stromal cell-mediated matrix contraction via ALK receptors and reduced follistatin: Potential role of endothelial cells in skin fibrosis. J Cell Physiol 2018; 233:6714-6722. [PMID: 29345319 PMCID: PMC6056025 DOI: 10.1002/jcp.26494] [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: 10/06/2017] [Accepted: 01/17/2018] [Indexed: 12/18/2022]
Abstract
Abnormal cutaneous wound healing can lead to formation of fibrotic hypertrophic scars. Although several clinical risk factors have been described, the cross‐talk between different cell types resulting in hypertrophic scar formation is still poorly understood. The aim of this in vitro study was to investigate whether endothelial cells (EC) may play a role in skin fibrosis, for example, hypertrophic scar formation after full‐thickness skin trauma. Using a collagen/elastin matrix, we developed an in vitro fibrosis model to study the interaction between EC and dermal fibroblasts or adipose tissue‐derived mesenchymal stromal cells (ASC). Tissue equivalents containing dermal fibroblasts and EC displayed a normal phenotype. In contrast, tissue equivalents containing ASC and EC displayed a fibrotic phenotype indicated by contraction of the matrix, higher gene expression of ACTA2, COL1A, COL3A, and less secretion of follistatin. The contraction was in part mediated via the TGF‐β pathway, as both inhibition of the ALK4/5/7 receptors and the addition of recombinant follistatin resulted in decreased matrix contraction (75 ± 11% and 24 ± 8%, respectively). In conclusion, our study shows that EC may play a critical role in fibrotic events, as seen in hypertrophic scars, by stimulating ASC‐mediated matrix contraction via regulation of fibrosis‐related proteins.
Collapse
Affiliation(s)
- Hanneke N Monsuur
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Lenie J van den Broek
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Pieter Koolwijk
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Frank B Niessen
- Department of Plastic, Reconstructive and Hand Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands.,Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Buskermolen JK, Reijnders CMA, Spiekstra SW, Steinberg T, Kleverlaan CJ, Feilzer AJ, Bakker AD, Gibbs S. Development of a Full-Thickness Human Gingiva Equivalent Constructed from Immortalized Keratinocytes and Fibroblasts. Tissue Eng Part C Methods 2017; 22:781-91. [PMID: 27406216 PMCID: PMC4991602 DOI: 10.1089/ten.tec.2016.0066] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Organotypic models make it possible to investigate the unique properties of oral mucosa in vitro. For gingiva, the use of human primary keratinocytes (KC) and fibroblasts (Fib) is limited due to the availability and size of donor biopsies. The use of physiologically relevant immortalized cell lines would solve these problems. The aim of this study was to develop fully differentiated human gingiva equivalents (GE) constructed entirely from cell lines, to compare them with the primary cell counterpart (Prim), and to test relevance in an in vitro wound healing assay. Reconstructed gingiva epithelium on a gingiva fibroblast-populated collagen hydrogel was constructed from cell lines (keratinocytes: TERT or HPV immortalized; fibroblasts: TERT immortalized) and compared to GE-Prim and native gingiva. GE were characterized by immunohistochemical staining for proliferation (Ki67), epithelial differentiation (K10, K13), and basement membrane (collagen type IV and laminin 5). To test functionality of GE-TERT, full-thickness wounds were introduced. Reepithelialization, fibroblast repopulation of hydrogel, metabolic activity (MTT assay), and (pro-)inflammatory cytokine release (enzyme-linked immunosorbent assay) were assessed during wound closure over 7 days. Significant differences in basal KC cytokine secretion (IL-1α, IL-18, and CXCL8) were only observed between KC-Prim and KC-HPV. When Fib-Prim and Fib-TERT were stimulated with TNF-α, no differences were observed regarding cytokine secretion (IL-6, CXCL8, and CCL2). GE-TERT histology, keratin, and basement membrane protein expression very closely represented native gingiva and GE-Prim. In contrast, the epithelium of GE made with HPV-immortalized KC was disorganized, showing suprabasal proliferating cells, limited keratinocyte differentiation, and the absence of basement membrane proteins. When a wound was introduced into the more physiologically relevant GE-TERT model, an immediate inflammatory response (IL-6, CCL2, and CXCL8) was observed followed by complete reepithelialization. Seven days after wounding, tissue integrity, metabolic activity, and cytokine levels had returned to the prewounded state. In conclusion, immortalized human gingiva KC and fibroblasts can be used to make physiologically relevant GE, which resemble either the healthy gingiva or a neoplastic disease model. These organotypic models will provide valuable tools to investigate oral mucosa biology and can also be used as an animal alternative for drug targeting, vaccination studies, microbial biofilm studies, and testing new therapeutics.
Collapse
Affiliation(s)
- Jeroen K Buskermolen
- 1 Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , MOVE Research Institute Amsterdam, Amsterdam, The Netherlands .,2 Department of Dental Materials Science, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | | | - Sander W Spiekstra
- 3 Department of Dermatology, VU University Medical Centre , Amsterdam, The Netherlands
| | - Thorsten Steinberg
- 4 Department of Oral Biotechnology, University Medical Center Freiburg , Freiburg, Germany
| | - Cornelis J Kleverlaan
- 2 Department of Dental Materials Science, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Albert J Feilzer
- 2 Department of Dental Materials Science, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Astrid D Bakker
- 1 Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Susan Gibbs
- 1 Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , MOVE Research Institute Amsterdam, Amsterdam, The Netherlands .,3 Department of Dermatology, VU University Medical Centre , Amsterdam, The Netherlands
| |
Collapse
|
34
|
Buskermolen JK, Roffel S, Gibbs S. Stimulation of oral fibroblast chemokine receptors identifies CCR3 and CCR4 as potential wound healing targets. J Cell Physiol 2017; 232:2996-3005. [PMID: 28387445 PMCID: PMC5575500 DOI: 10.1002/jcp.25946] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/05/2017] [Indexed: 02/02/2023]
Abstract
The focus of this study was to determine which chemokine receptors are present on oral fibroblasts and whether these receptors influence proliferation, migration, and/or the release of wound healing mediators. This information may provide insight into the superior wound healing characteristics of the oral mucosa. The gingiva fibroblasts expressed 12 different chemokine receptors (CCR3, CCR4, CCR6, CCR9, CCR10, CXCR1, CXCR2, CXCR4, CXCR5, CXCR7, CX3CR1, and XCR1), as analyzed by flow cytometry. Fourteen corresponding chemokines (CCL5, CCL15, CCL20, CCL22, CCL25, CCL27, CCL28, CXCL1, CXCL8, CXCL11, CXCL12, CXCL13, CX3CL1, and XCL1) were used to study the activation of these receptors on gingiva fibroblasts. Twelve of these fourteen chemokines stimulated gingiva fibroblast migration (all except for CXCL8 and CXCL12). Five of the chemokines stimulated proliferation (CCL5/CCR3, CCL15/CCR3, CCL22/CCR4, CCL28/CCR3/CCR10, and XCL1/XCR1). Furthermore, CCL28/CCR3/CCR10 and CCL22/CCR4 stimulation increased IL-6 secretion and CCL28/CCR3/CCR10 together with CCL27/CCR10 upregulated HGF secretion. Moreover, TIMP-1 secretion was reduced by CCL15/CCR3. In conclusion, this in-vitro study identifies chemokine receptor-ligand pairs which may be used in future targeted wound healing strategies. In particular, we identified the chemokine receptors CCR3 and CCR4, and the mucosa specific chemokine CCL28, as having an predominant role in oral wound healing by increasing human gingiva fibroblast proliferation, migration, and the secretion of IL-6 and HGF and reducing the secretion of TIMP-1.
Collapse
Affiliation(s)
- Jeroen K. Buskermolen
- Department of Oral Cell BiologyAcademic Centre for Dentistry (ACTA)University of Amsterdam and Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Sanne Roffel
- Department of Oral Cell BiologyAcademic Centre for Dentistry (ACTA)University of Amsterdam and Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Susan Gibbs
- Department of Oral Cell BiologyAcademic Centre for Dentistry (ACTA)University of Amsterdam and Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of DermatologyVU University Medical CenterAmsterdamThe Netherlands
| |
Collapse
|
35
|
Konstantinow A, Arnold A, Djabali K, Kempf W, Gutermuth J, Fischer T, Biedermann T. Therapy of ulcus cruris of venous and mixed venous arterial origin with autologous, adult, native progenitor cells from subcutaneous adipose tissue: a prospective clinical pilot study. J Eur Acad Dermatol Venereol 2017; 31:2104-2118. [PMID: 28750144 DOI: 10.1111/jdv.14489] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/11/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND The stromal vascular fraction (SVF) of adipose tissue consists of cellular subpopulations with distinct regenerative potential. OBJECTIVE To investigate the regenerative capacities of autologous SVF cells in the treatment of chronic leg ulcers of venous (VLU) and arterial-venous (AVLU) origin. METHODS Multimorbid ulcer patients received a singular topical treatment with 9-15 × 106 SVF cells, separated from abdominal lipoaspirates by digestion with collagenase and neutral protease and applied immediately after isolation. The primary endpoints were the change in wound size 12 weeks after treatment and evaluation of adverse events. Secondary endpoints included the time to complete wound epithelialization and change in pain levels. Postoperative wound treatment modalities and treatment of comorbidities were not intensified compared with pre-operative management. Follow-up period was at least 6 months. RESULTS Sixteen elderly ulcer patients (seven with VLU, nine with AVLU) were treated as described. All VLU patients (median ulcer size: 48.25 cm2 ) and four of nine AVLU patients showed complete epithelialization of the ulcers within 71-174 days. In three patients with large ulcerations on both legs, ulcerations on the non-treated, contralateral leg also epithelialized. Patients reported a considerable rapid decrease in pain intensity by 2.5 points on average on a visual scale from 1 to 5 within the first 2 weeks after treatment. The patients were followed up for 9-44 months (median: 30 months). No severe side-effects were observed. CONCLUSIONS The use of SVF cells presents an effective, minimally invasive option for the treatment of VLU and AVLU even in multimorbid patients. In patients with larger predominantly ischaemic AVLU and comorbidities, one-time application of the used amounts of SVF cells was not sufficient in the majority of cases.
Collapse
Affiliation(s)
- A Konstantinow
- Department of Dermatology and Allergology, Technical University Munich, Munich, Germany
| | - A Arnold
- Department of Dermatology and Allergology, Technical University Munich, Munich, Germany
| | - K Djabali
- School of Medicine, Epigenetic of Aging, Technical University Munich, Garching, Germany
| | - W Kempf
- Department of Dermatology and Allergology, Technical University Munich, Munich, Germany
| | - J Gutermuth
- Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - T Fischer
- Department of Dermatology and Allergology, Technical University Munich, Munich, Germany
| | - T Biedermann
- Department of Dermatology and Allergology, Technical University Munich, Munich, Germany
| |
Collapse
|
36
|
Burn Eschar Stimulates Fibroblast and Adipose Mesenchymal Stromal Cell Proliferation and Migration but Inhibits Endothelial Cell Sprouting. Int J Mol Sci 2017; 18:ijms18081790. [PMID: 28820426 PMCID: PMC5578178 DOI: 10.3390/ijms18081790] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 07/31/2017] [Accepted: 08/12/2017] [Indexed: 12/03/2022] Open
Abstract
The majority of full-thickness burn wounds heal with hypertrophic scar formation. Burn eschar most probably influences early burn wound healing, since granulation tissue only forms after escharotomy. In order to investigate the effect of burn eschar on delayed granulation tissue formation, burn wound extract (BWE) was isolated from the interface between non-viable eschar and viable tissue. The influence of BWE on the activity of endothelial cells derived from dermis and adipose tissue, dermal fibroblasts and adipose tissue-derived mesenchymal stromal cells (ASC) was determined. It was found that BWE stimulated endothelial cell inflammatory cytokine (CXCL8, IL-6 and CCL2) secretion and migration. However, BWE had no effect on endothelial cell proliferation or angiogenic sprouting. Indeed, BWE inhibited basic Fibroblast Growth Factor (bFGF) induced endothelial cell proliferation and sprouting. In contrast, BWE stimulated fibroblast and ASC proliferation and migration. No difference was observed between cells isolated from dermis or adipose tissue. The inhibitory effect of BWE on bFGF-induced endothelial proliferation and sprouting would explain why excessive granulation tissue formation is prevented in full-thickness burn wounds as long as the eschar is still present. Identifying the eschar factors responsible for this might give indications for therapeutic targets aimed at reducing hypertrophic scar formation which is initiated by excessive granulation tissue formation once eschar is removed.
Collapse
|
37
|
Alexeev V, Salas-Alanis JC, Palisson F, Mukhtarzada L, Fortuna G, Uitto J, South A, Igoucheva O. Pro-Inflammatory Chemokines and Cytokines Dominate the Blister Fluid Molecular Signature in Patients with Epidermolysis Bullosa and Affect Leukocyte and Stem Cell Migration. J Invest Dermatol 2017; 137:2298-2308. [PMID: 28736230 DOI: 10.1016/j.jid.2017.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 06/05/2017] [Accepted: 07/03/2017] [Indexed: 12/21/2022]
Abstract
Hereditary epidermolysis bullosa (EB) is associated with skin blistering and the development of chronic nonhealing wounds. Although clinical studies have shown that cell-based therapies improve wound healing, the recruitment of therapeutic cells to blistering skin and to more advanced skin lesions remains a challenge. Here, we analyzed cytokines and chemokines in blister fluids of patients affected by dystrophic, junctional, and simplex EB. Our analysis revealed high levels of CXCR1, CXCR2, CCR2, and CCR4 ligands, particularly dominant in dystrophic and junctional EB. In vitro migration assays demonstrated the preferential recruitment of CCR4+ lymphocytes and CXCR1+, CXCR2+, and CCR2+ myeloid cells toward EB-derived blister fluids. Immunophenotyping of skin-infiltrating leukocytes confirmed substantial infiltration of EB-affected skin with resting (CD45RA+) and activated (CD45RO+) T cells and CXCR2+ CD11b+ cells, many of which were identified as CD16b+ neutrophils. Our studies also showed that abundance of CXCR2 ligand in blister fluids also creates a favorable milieu for the recruitment of the CXCR2+ stem cells, as validated by in vitro and in-matrix migration assays. Collectively, this study identified several chemotactic pathways that control the recruitment of leukocytes to the EB-associated skin lesions. These chemotactic axes could be explored for the refinement of the cutaneous homing of the therapeutic stem cells.
Collapse
Affiliation(s)
- Vitali Alexeev
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Julio Cesar Salas-Alanis
- Department of Basic Sciences, Health Sciences Division, University of Monterrey, Monterrey, Mexico
| | - Francis Palisson
- Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Lila Mukhtarzada
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Giulio Fortuna
- Department of Diagnostic Science, Louisiana State University School of Dentistry, New Orleans, Louisiana, USA
| | - Jouni Uitto
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Andrew South
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Olga Igoucheva
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
38
|
Boink MA, Roffel S, Breetveld M, Thon M, Haasjes MSP, Waaijman T, Scheper RJ, Blok CS, Gibbs S. Comparison of advanced therapy medicinal product gingiva and skin substitutes and their in vitro wound healing potentials. J Tissue Eng Regen Med 2017; 12:e1088-e1097. [PMID: 28388010 PMCID: PMC5836907 DOI: 10.1002/term.2438] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 01/25/2017] [Accepted: 04/03/2017] [Indexed: 01/23/2023]
Abstract
Skin and oral mucosa substitutes are a therapeutic option for closing hard‐to‐heal skin and oral wounds. Our aim was to develop bi‐layered skin and gingiva substitutes, from 3 mm diameter biopsies, cultured under identical conditions, which are compliant with current European regulations for advanced therapy medicinal products. We present in vitro mode of action methods to (i) determine viability: epithelial expansion, proliferation (Ki‐67), metabolic activity (MTT assay); (ii) characterize skin and gingiva substitutes: histology and immunohistochemistry; and (iii) determine potency: soluble wound healing mediator release (enzyme‐linked immunosorbent assay). Both skin and gingiva substitutes consist of metabolically active autologous reconstructed differentiated epithelium expanding from the original biopsy sheet on a fibroblast populated connective tissue matrix (donor dermis). Gingival epithelium expanded 1.7‐fold more than skin epithelium during the 3 week culture period. The percentage of proliferating Ki‐67‐positive cells located in the basal layer of the gingiva substitute was >1.5‐fold higher than in the skin substitute. Keratins 16 and 17, which are upregulated during normal wound healing, were expressed in both the skin and gingiva substitutes. Notably, the gingiva substitute secreted higher amounts of key cytokines involved in mitogenesis, motogenesis and chemotaxis (interleukin‐6 > 23‐fold, CXCL8 > 2.5‐fold) as well as higher amounts of the anti‐fibrotic growth factor, hepatocyte growth factor (>7‐fold), compared with the skin substitute. In conclusion, while addressing the viability, characterization and potency of the tissue substitutes, important intrinsic differences between skin and gingiva were discovered that may explain in part the superior quality of wound healing observed in the oral mucosa compared with skin.
Collapse
Affiliation(s)
- Mireille A Boink
- Department of Oral Biochemistry, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, the Netherlands.,Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Sanne Roffel
- Department of Oral Biochemistry, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, the Netherlands.,Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Melanie Breetveld
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Maria Thon
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands.,A-Skin BV, Amsterdam, the Netherlands
| | - Michiel S P Haasjes
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Taco Waaijman
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Rik J Scheper
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Chantal S Blok
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Susan Gibbs
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, the Netherlands.,Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, the Netherlands
| |
Collapse
|
39
|
Lee J, Lee J, Cha J, Choi E, Park S, Cho K, Kim C. Chemokine in inflamed periodontal tissues activates healthy periodontal‐ligament stem cell migration. J Clin Periodontol 2017; 44:530-539. [DOI: 10.1111/jcpe.12710] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2017] [Indexed: 01/09/2023]
Affiliation(s)
- Jung‐Seok Lee
- Department of PeriodontologyResearch Institute for Periodontal RegenerationCollege of DentistryYonsei UniversitySeoulKorea
| | - Jong‐Bin Lee
- Department of PeriodontologyResearch Institute for Periodontal RegenerationCollege of DentistryYonsei UniversitySeoulKorea
| | - Jae‐Kook Cha
- Department of PeriodontologyResearch Institute for Periodontal RegenerationCollege of DentistryYonsei UniversitySeoulKorea
| | - Eun‐Young Choi
- Department of PeriodontologyResearch Institute for Periodontal RegenerationCollege of DentistryYonsei UniversitySeoulKorea
| | - So‐Yon Park
- Department of PeriodontologyResearch Institute for Periodontal RegenerationCollege of DentistryYonsei UniversitySeoulKorea
| | - Kyoo‐Sung Cho
- Department of PeriodontologyResearch Institute for Periodontal RegenerationCollege of DentistryYonsei UniversitySeoulKorea
| | - Chang‐Sung Kim
- Department of PeriodontologyResearch Institute for Periodontal RegenerationCollege of DentistryYonsei UniversitySeoulKorea
- Department of Applied Life ScienceBK21 PLUS ProjectCollege of DentistryYonsei UniversitySeoulKorea
| |
Collapse
|
40
|
Extensive Characterization and Comparison of Endothelial Cells Derived from Dermis and Adipose Tissue: Potential Use in Tissue Engineering. PLoS One 2016; 11:e0167056. [PMID: 27902740 PMCID: PMC5130240 DOI: 10.1371/journal.pone.0167056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 11/08/2016] [Indexed: 02/04/2023] Open
Abstract
Tissue-engineered constructs need to become quickly vascularized in order to ensure graft take. One way of achieving this is to incorporate endothelial cells (EC) into the construct. The adipose tissue stromal vascular fraction (adipose-SVF) might provide an alternative source for endothelial cells as adipose tissue can easily be obtained by liposuction. Since adipose-EC are now gaining more interest in tissue engineering, we aimed to extensively characterize endothelial cells from adipose tissue (adipose-EC) and compare them with endothelial cells from dermis (dermal-EC). The amount of endothelial cells before purification varied between 4–16% of the total stromal population. After MACS selection for CD31 positive cells, a >99% pure population of endothelial cells was obtained within two weeks of culture. Adipose- and dermal-EC expressed the typical endothelial markers PECAM-1, ICAM-1, Endoglin, VE-cadherin and VEGFR2 to a similar extent, with 80–99% of the cell population staining positive. With the exception of CXCR4, which was expressed on 29% of endothelial cells, all other chemokine receptors (CXCR1, 2, 3, and CCR2) were expressed on less than 5% of the endothelial cell populations. Adipose-EC proliferated similar to dermal-EC, but responded less to the mitogens bFGF and VEGF. A similar migration rate was found for both adipose-EC and dermal-EC in response to bFGF. Sprouting of adipose-EC and dermal-EC was induced by bFGF and VEGF in a 3D fibrin matrix. After stimulation of adipose-EC and dermal-EC with TNF-α an increased secretion was seen for PDGF-BB, but not uPA, PAI-1 or Angiopoietin-2. Furthermore, secretion of cytokines and chemokines (IL-6, CCL2, CCL5, CCL20, CXCL1, CXCL8 and CXCL10) was also upregulated by both adipose- and dermal-EC. The similar characteristics of adipose-EC compared to their dermal-derived counterpart make them particularly interesting for skin tissue engineering. In conclusion, we show here that adipose tissue provides for an excellent source of endothelial cells for tissue engineering purposes, since they are readily available, and easily isolated and amplified.
Collapse
|
41
|
Methods to study differences in cell mobility during skin wound healing in vitro. J Biomech 2016; 49:1381-1387. [DOI: 10.1016/j.jbiomech.2016.01.040] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 01/19/2016] [Accepted: 01/28/2016] [Indexed: 01/19/2023]
|
42
|
Boink MA, van den Broek LJ, Roffel S, Nazmi K, Bolscher JGM, Gefen A, Veerman ECI, Gibbs S. Different wound healing properties of dermis, adipose, and gingiva mesenchymal stromal cells. Wound Repair Regen 2015; 24:100-9. [PMID: 26542883 DOI: 10.1111/wrr.12380] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/03/2015] [Indexed: 12/15/2022]
Abstract
Oral wounds heal faster and with better scar quality than skin wounds. Deep skin wounds where adipose tissue is exposed, have a greater risk of forming hypertrophic scars. Differences in wound healing and final scar quality might be related to differences in mesenchymal stromal cells (MSC) and their ability to respond to intrinsic (autocrine) and extrinsic signals, such as human salivary histatin, epidermal growth factor, and transforming growth factor beta1. Dermis-, adipose-, and gingiva-derived MSC were compared for their regenerative potential with regards to proliferation, migration, and matrix contraction. Proliferation was assessed by cell counting and migration using a scratch wound assay. Matrix contraction and alpha smooth muscle actin was assessed in MSC populated collagen gels, and also in skin and gingival full thickness tissue engineered equivalents (reconstructed epithelium on MSC populated matrix). Compared to skin-derived MSC, gingiva MSC showed greater proliferation and migration capacity, and less matrix contraction in full thickness tissue equivalents, which may partly explain the superior oral wound healing. Epidermal keratinocytes were required for enhanced adipose MSC matrix contraction and alpha smooth muscle actin expression, and may therefore contribute to adverse scarring in deep cutaneous wounds. Histatin enhanced migration without influencing proliferation or matrix contraction in all three MSC, indicating that salivary peptides may have a beneficial effect on wound closure in general. Transforming growth factor beta1 enhanced contraction and alpha smooth muscle actin expression in all three MSC types when incorporated into collagen gels. Understanding the mechanisms responsible for the superior oral wound healing will aid us to develop advanced strategies for optimal skin regeneration, wound healing and scar formation.
Collapse
Affiliation(s)
- Mireille A Boink
- Department of Oral Biochemistry, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, The Netherlands.,Department of Dermatology, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Sanne Roffel
- Department of Oral Biochemistry, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, The Netherlands.,Department of Dermatology, VU University Medical Center, Amsterdam, The Netherlands
| | - Kamran Nazmi
- Department of Oral Biochemistry, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, The Netherlands
| | - Jan G M Bolscher
- Department of Oral Biochemistry, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, The Netherlands
| | - Amit Gefen
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Enno C I Veerman
- Department of Oral Biochemistry, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Dermatology, VU University Medical Center, Amsterdam, The Netherlands.,Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Hocking AM. The Role of Chemokines in Mesenchymal Stem Cell Homing to Wounds. Adv Wound Care (New Rochelle) 2015; 4:623-630. [PMID: 26543676 DOI: 10.1089/wound.2014.0579] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Significance: Mesenchymal stem cells (MSCs) are being administered to cutaneous wounds with the goal of accelerating wound closure and promoting regeneration instead of scar formation. An ongoing challenge for cell-based therapies is achieving effective and optimal targeted delivery and engraftment at the site of injury. Contributing to this challenge is our incomplete understanding of endogenous MSC homing to sites of injury. Recent Advances: Chemokines and their receptors are now recognized as important mediators of stem cell homing. To date, the most studied chemokine-chemokine receptor axis in MSC homing to wounds is CXCL12-CXCR4 but recent work suggests that CCL27-CCR10 and CCL21-CCR7 may also be involved. Critical Issues: Strategies to enhance chemokine-mediated MSC homing to wounds are using a variety of approaches to amplify the chemokine signal at the wound site and/or overexpress specific chemokine receptors on the surface of the MSC. Future Directions: Harnessing chemokine signaling may enhance the therapeutic effects of stem cell therapy by increasing the number of both exogenous and endogenous stem cells recruited to the site of injury. Alternatively, chemokine-based therapies directly targeting endogenous stem cells may circumvent the need for the time-consuming and costly isolation and expansion of autologous stem cells prior to therapeutic administration.
Collapse
Affiliation(s)
- Anne M. Hocking
- Department of Surgery, University of Washington, Seattle, Washington
| |
Collapse
|
44
|
Gingiva Equivalents Secrete Negligible Amounts of Key Chemokines Involved in Langerhans Cell Migration Compared to Skin Equivalents. J Immunol Res 2015; 2015:627125. [PMID: 26539556 PMCID: PMC4619927 DOI: 10.1155/2015/627125] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/05/2015] [Indexed: 11/18/2022] Open
Abstract
Both oral mucosa and skin have the capacity to maintain immune homeostasis or regulate immune responses upon environmental assault. Whereas much is known about key innate immune events in skin, little is known about oral mucosa. Comparative studies are limited due to the scarce supply of oral mucosa for ex vivo studies. Therefore, we used organotypic tissue equivalents (reconstructed epithelium on fibroblast-populated collagen hydrogel) to study cross talk between cells. Oral mucosa and skin equivalents were compared regarding secretion of cytokines and chemokines involved in LC migration and general inflammation. Basal secretion, representative of homeostasis, and also secretion after stimulation with TNFα, an allergen (cinnamaldehyde), or an irritant (SDS) were assessed. We found that proinflammatory IL-18 and chemokines CCL2, CCL20, and CXCL12, all involved in LC migration, were predominantly secreted by skin as compared to gingiva. Furthermore, CCL27 was predominantly secreted by skin whereas CCL28 was predominantly secreted by gingiva. In contrast, general inflammatory cytokines IL-6 and CXCL8 were secreted similarly by skin and gingiva. These results indicate that the cytokines and chemokines triggering innate immunity and LC migration are different in skin and gingiva. This differential regulation should be figured into novel therapy or vaccination strategies in the context of skin versus mucosa.
Collapse
|
45
|
Lu L, Zhang X, Zhang M, Zhang H, Liao L, Yang T, Zhang J, Xian L, Chen D, Wang M. RANTES and SDF-1 Are Keys in Cell-based Therapy of TMJ Osteoarthritis. J Dent Res 2015; 94:1601-9. [PMID: 26377571 DOI: 10.1177/0022034515604621] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The present study aimed to investigate the therapeutic effect of injections of local bone marrow mesenchymal stem cells (BMSCs) on osteoarthritis (OA) of the temporomandibular joint (TMJ) and to explore the role of stromal cell-derived factor 1 (SDF-1) and regulated on activation, normal T-cell expressed and secreted (RANTES) in this effect. Fundamentally, OA of the TMJ was induced by unilateral anterior crossbite in mice. Exogenous green fluorescent protein-labeled BMSCs (GFP-BMSCs) were weekly injected into the TMJ region for 4, 8, and 12 wk. The reparative effects of exogenous GFP-BMSCs were investigated by morphological observation and micro-computed tomography. The differentiation of GFP-BMSCs in the cartilage was examined by double immunofluorescence of GFPs with type II collagen, and the expression of related factors in the condylar cartilage was quantified by real-time polymerase chain reaction. The role of RANTES and SDF-1 in the therapeutic effect of exogenous BMSCs was examined by both in vitro and in vivo studies. The OA cartilage of the TMJ displays a synchronous increase in SDF-1 and RANTES expression and a higher capability of attracting the migration of GFP-BMSCs. The implanted GFP-BMSCs differentiated into type II collagen-positive cells and reversed cartilage degradation and subchondral bone loss in mice with OA of the TMJ. The migration of GFP-BMSCs towards OA cartilage and the rescuing effect of GFP-BMSC injections were impaired by the inhibitors of C-X-C chemokine receptor type 4 (CXCR4) and C-C chemokine receptor type 1 (CCR1), which are the receptors of SDF-1 and RANTES, respectively. Our data indicated that SDF-1/CXCR4 and RANTES/CCR1 signals are pivotal and function synergistically in the recruitment of GFP-BMSCs towards degraded cartilage in mice OA of the TMJ.
Collapse
Affiliation(s)
- L Lu
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - X Zhang
- Department of Stomatology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - M Zhang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - H Zhang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - L Liao
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - T Yang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - J Zhang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - L Xian
- Department of Hematology, Johns Hopkins University, Baltimore, MD, USA
| | - D Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - M Wang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
46
|
Reijnders CMA, van Lier A, Roffel S, Kramer D, Scheper RJ, Gibbs S. Development of a Full-Thickness Human Skin Equivalent In Vitro Model Derived from TERT-Immortalized Keratinocytes and Fibroblasts. Tissue Eng Part A 2015; 21:2448-59. [PMID: 26135533 PMCID: PMC4554934 DOI: 10.1089/ten.tea.2015.0139] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Currently, human skin equivalents (HSEs) used for in vitro assays (e.g., for wound healing) make use of primary human skin cells. Limitations of primary keratinocytes and fibroblasts include availability of donor skin and donor variation. The use of physiologically relevant cell lines could solve these limitations. The aim was to develop a fully differentiated HSE constructed entirely from human skin cell lines, which could be applied for in vitro wound-healing assays. Skin equivalents were constructed from human TERT-immortalized keratinocytes and fibroblasts (TERT-HSE) and compared with native skin and primary HSEs. HSEs were characterized by hematoxylin–eosin and immunohistochemical stainings with markers for epidermal proliferation and differentiation, basement membrane (BM), fibroblasts, and the extracellular matrix (ECM). Ultrastructure was determined with electron microscopy. To test the functionality of the TERT-HSE, burn and cold injuries were applied, followed by immunohistochemical stainings, measurement of reepithelialization, and determination of secreted wound-healing mediators. The TERT-HSE was composed of a fully differentiated epidermis and a fibroblast-populated dermis comparable to native skin and primary HSE. The epidermis consisted of proliferating keratinocytes within the basal layer, followed by multiple spinous layers, a granular layer, and cornified layers. Within the TERT-HSE, the membrane junctions such as corneosomes, desmosomes, and hemidesmosomes were well developed as shown by ultrastructure pictures. Furthermore, the BM consisted of a lamina lucida and lamina densa comparable to native skin. The dermal matrix of the TERT-HSE was more similar to native skin than the primary construct, since collagen III, an ECM marker, was present in TERT-HSEs and absent in primary HSEs. After wounding, the TERT-HSE was able to reepithelialize and secrete inflammatory wound-healing mediators. In conclusion, the novel TERT-HSE, constructed entirely from human cell lines, provides an excellent opportunity to study in vitro skin biology and can also be used for drug targeting and testing new therapeutics, and ultimately, for incorporating into skin-on-a chip in the future.
Collapse
Affiliation(s)
| | - Amanda van Lier
- 1 Department of Dermatology, VU University Medical Centre , Amsterdam, The Netherlands
| | - Sanne Roffel
- 1 Department of Dermatology, VU University Medical Centre , Amsterdam, The Netherlands
| | - Duco Kramer
- 2 Department of Dermatology, University Medical Centre Groningen , Groningen, The Netherlands
| | - Rik J Scheper
- 3 Department of Pathology, VU University Medical Centre , Amsterdam, The Netherlands
| | - Susan Gibbs
- 1 Department of Dermatology, VU University Medical Centre , Amsterdam, The Netherlands .,4 Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam , Amsterdam, The Netherlands
| |
Collapse
|
47
|
van den Broek LJ, Limandjaja GC, Niessen FB, Gibbs S. Human hypertrophic and keloid scar models: principles, limitations and future challenges from a tissue engineering perspective. Exp Dermatol 2015; 23:382-6. [PMID: 24750541 PMCID: PMC4369123 DOI: 10.1111/exd.12419] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2014] [Indexed: 12/23/2022]
Abstract
Most cutaneous wounds heal with scar formation. Ideally, an inconspicuous normotrophic scar is formed, but an abnormal scar (hypertrophic scar or keloid) can also develop. A major challenge to scientists and physicians is to prevent adverse scar formation after severe trauma (e.g. burn injury) and understand why some individuals will form adverse scars even after relatively minor injury. Currently, many different models exist to study scar formation, ranging from simple monolayer cell culture to 3D tissue-engineered models even to humanized mouse models. Currently, these high-/medium-throughput test models avoid the main questions referring to why an adverse scar forms instead of a normotrophic scar and what causes a hypertrophic scar to form rather than a keloid scar and also, how is the genetic predisposition of the individual and the immune system involved. This information is essential if we are to identify new drug targets and develop optimal strategies in the future to prevent adverse scar formation. This viewpoint review summarizes the progress on in vitro and animal scar models, stresses the limitations in the current models and identifies the future challenges if scar-free healing is to be achieved in the future.
Collapse
Affiliation(s)
- Lenie J van den Broek
- Department of Dermatology, VU University Medical Center, Amsterdam, The Netherlands; Research Institute MOVE, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
48
|
Van de Laar E, Clifford M, Hasenoeder S, Kim BR, Wang D, Lee S, Paterson J, Vu NM, Waddell TK, Keshavjee S, Tsao MS, Ailles L, Moghal N. Cell surface marker profiling of human tracheal basal cells reveals distinct subpopulations, identifies MST1/MSP as a mitogenic signal, and identifies new biomarkers for lung squamous cell carcinomas. Respir Res 2014; 15:160. [PMID: 25551685 PMCID: PMC4343068 DOI: 10.1186/s12931-014-0160-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 12/17/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The large airways of the lungs (trachea and bronchi) are lined with a pseudostratified mucociliary epithelium, which is maintained by stem cells/progenitors within the basal cell compartment. Alterations in basal cell behavior can contribute to large airway diseases including squamous cell carcinomas (SQCCs). Basal cells have traditionally been thought of as a uniform population defined by basolateral position, cuboidal cell shape, and expression of pan-basal cell lineage markers like KRT5 and TP63. While some evidence suggests that basal cells are not all functionally equivalent, few heterogeneously expressed markers have been identified to purify and study subpopulations. In addition, few signaling pathways have been identified that regulate their cell behavior. The goals of this work were to investigate tracheal basal cell diversity and to identify new signaling pathways that regulate basal cell behavior. METHODS We used flow cytometry (FACS) to profile cell surface marker expression at a single cell level in primary human tracheal basal cell cultures that maintain stem cell/progenitor activity. FACS results were validated with tissue staining, in silico comparisons with normal basal cell and lung cancer datasets, and an in vitro proliferation assay. RESULTS We identified 105 surface markers, with 47 markers identifying potential subpopulations. These subpopulations generally fell into more (~ > 13%) or less abundant (~ < 6%) groups. Microarray gene expression profiling supported the heterogeneous expression of these markers in the total population, and immunostaining of large airway tissue suggested that some of these markers are relevant in vivo. 24 markers were enriched in lung SQCCs relative to adenocarcinomas, with four markers having prognostic significance in SQCCs. We also identified 33 signaling receptors, including the MST1R/RON growth factor receptor, whose ligand MST1/MSP was mitogenic for basal cells. CONCLUSION This work provides the largest description to date of molecular diversity among human large airway basal cells. Furthermore, these markers can be used to further study basal cell function in repair and disease, and may aid in the classification and study of SQCCs.
Collapse
Affiliation(s)
- Emily Van de Laar
- />Department of Medical Biophysics, Ontario Cancer Institute/Campbell Family Cancer Research Institute/Princess Margaret Cancer Centre/University Health Network, University of Toronto, Toronto, ON M5G 1 L7 Canada
| | - Monica Clifford
- />Department of Medical Biophysics, Ontario Cancer Institute/Campbell Family Cancer Research Institute/Princess Margaret Cancer Centre/University Health Network, University of Toronto, Toronto, ON M5G 1 L7 Canada
| | - Stefan Hasenoeder
- />Department of Medical Biophysics, Ontario Cancer Institute/Campbell Family Cancer Research Institute/Princess Margaret Cancer Centre/University Health Network, University of Toronto, Toronto, ON M5G 1 L7 Canada
- />Present address: Helmholtz Zentrum München, Institute of Stem Cell Research, Ingolstädter Landstrasse 1, 85746 Neuherberg, Germany
| | - Bo Ram Kim
- />Department of Medical Biophysics, Ontario Cancer Institute/Campbell Family Cancer Research Institute/Princess Margaret Cancer Centre/University Health Network, University of Toronto, Toronto, ON M5G 1 L7 Canada
| | - Dennis Wang
- />Department of Medical Biophysics, Ontario Cancer Institute/Campbell Family Cancer Research Institute/Princess Margaret Cancer Centre/University Health Network, University of Toronto, Toronto, ON M5G 1 L7 Canada
| | - Sharon Lee
- />Department of Medical Biophysics, Ontario Cancer Institute/Campbell Family Cancer Research Institute/Princess Margaret Cancer Centre/University Health Network, University of Toronto, Toronto, ON M5G 1 L7 Canada
- />Department of Applied Mathematics, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1 Canada
| | - Josh Paterson
- />Department of Medical Biophysics, Ontario Cancer Institute/Campbell Family Cancer Research Institute/Princess Margaret Cancer Centre/University Health Network, University of Toronto, Toronto, ON M5G 1 L7 Canada
| | - Nancy M Vu
- />Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112 USA
- />Present address: University of Utah School of Medicine, Salt Lake City, UT 84132 USA
| | - Thomas K Waddell
- />Toronto Lung Transplant Program, University Health Network, University of Toronto, Toronto, ON M5G 1 L7 Canada
| | - Shaf Keshavjee
- />Toronto Lung Transplant Program, University Health Network, University of Toronto, Toronto, ON M5G 1 L7 Canada
| | - Ming-Sound Tsao
- />Department of Medical Biophysics, Ontario Cancer Institute/Campbell Family Cancer Research Institute/Princess Margaret Cancer Centre/University Health Network, University of Toronto, Toronto, ON M5G 1 L7 Canada
| | - Laurie Ailles
- />Department of Medical Biophysics, Ontario Cancer Institute/Campbell Family Cancer Research Institute/Princess Margaret Cancer Centre/University Health Network, University of Toronto, Toronto, ON M5G 1 L7 Canada
| | - Nadeem Moghal
- />Department of Medical Biophysics, Ontario Cancer Institute/Campbell Family Cancer Research Institute/Princess Margaret Cancer Centre/University Health Network, University of Toronto, Toronto, ON M5G 1 L7 Canada
- />Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112 USA
- />Present address: Ontario Cancer Institute and Princess Margaret Hospital, University Health Network, Toronto, ON M5G 1 L7 Canada
| |
Collapse
|
49
|
Fetoni AR, Lattanzi W, Eramo SLM, Barba M, Paciello F, Moriconi C, Rolesi R, Michetti F, Troiani D, Paludetti G. Grafting and early expression of growth factors from adipose-derived stem cells transplanted into the cochlea, in a Guinea pig model of acoustic trauma. Front Cell Neurosci 2014; 8:334. [PMID: 25368551 PMCID: PMC4202717 DOI: 10.3389/fncel.2014.00334] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/30/2014] [Indexed: 01/13/2023] Open
Abstract
Noise exposure causes damage of multiple cochlear cell types producing permanent hearing loss with important social consequences. In mammals, no regeneration of either damaged hair cells or auditory neurons has been observed and no successful treatment is available to achieve a functional recovery. Loads of evidence indicate adipose-derived stem cells (ASCs) as promising tools in diversified regenerative medicine applications, due to the high degree of plasticity and trophic features. This study was aimed at identifying the path of in vivo cell migration and expression of trophic growth factors, upon ASCs transplantation into the cochlea, following noise-induced injury. ASCs were isolated in primary culture from the adipose tissue of a guinea pig, transduced using a viral vector to express the green fluorescent protein, and implanted into the scala tympani of deafened animals. Auditory function was assessed 3 and 7 days after surgery. The expression of trophic growth factors was comparatively analyzed using real-time PCR in control and noise-injured cochlear tissues. Immunofluorescence was used to assess the in vivo localization and expression of trophic growth factors in ASCs and cochleae, 3 and 7 days following homologous implantation. ASC implantation did not modify auditory function. ASCs migrated from the perilymphatic to the endolymphatic compartment, during the analyzed time course. Upon noise exposure, the expression of chemokine ligands and receptors related to the PDGF, VEGF, and TGFbeta pathways, increased in the cochlear tissues, possibly guiding in vivo cell migration. Immunofluorescence confirmed the increased expression, which appeared to be further strengthened by ASCs’ implantation. These results indicated that ASCs are able to migrate at the site of tissue damage and express trophic factors, upon intracochlear implantation, providing an original proof of principle, which could pave the way for further developments of ASC-based treatments of deafness.
Collapse
Affiliation(s)
- Anna Rita Fetoni
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Wanda Lattanzi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore , Rome , Italy ; Latium Musculoskeletal Tissue Bank , Rome , Italy
| | | | - Marta Barba
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Fabiola Paciello
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Chiara Moriconi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Rolando Rolesi
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore , Rome , Italy ; Latium Musculoskeletal Tissue Bank , Rome , Italy
| | - Diana Troiani
- Institute of Physiology, Università Cattolica del Sacro Cuore , Rome , Italy
| | - Gaetano Paludetti
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore , Rome , Italy
| |
Collapse
|
50
|
Chen Z, Wang Y, Shi C. Therapeutic Implications of Newly Identified Stem Cell Populations From the Skin Dermis. Cell Transplant 2014; 24:1405-22. [PMID: 24972091 DOI: 10.3727/096368914x682431] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Skin, the largest organ of the body, is a promising reservoir for adult stem cells. The epidermal stem cells and hair follicle stem cells have been well studied for their important roles in homeostasis, regeneration, and repair of the epidermis and appendages for decades. However, stem cells residing in dermis were not identified until the year 2001, when a variety of stem cell subpopulations have been isolated and identified from the dermis of mammalian skin such as neural crest stem cells, mesenchymal stem cell-like dermal stem cells, and dermal hematopoietic cells. These stem cell subpopulations exhibited capabilities of self-renewing, multipotent differentiating, and immunosuppressive properties. Hence, the dermis-derived stem cells showed extensive potential applications in regenerative medicine, especially for wound healing/tissue repair, neural repair, and hematopoietic recovery. Here we summarized current research on the stem cell subpopulations derived from the dermis and aimed to provide a comprehensive review on their isolation, specific markers, differentiation capacity, and the functional activities in homeostasis, regeneration, and tissue repair.
Collapse
Affiliation(s)
- Zelin Chen
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | | | | |
Collapse
|