1
|
Zielinska D, Micka-Michalak K, Ademi H, Fisch P, Boeni R, Linder T, Moehrlen U, Biedermann T, Klar AS. Adipose-mesenchymal stem cells enhance the formation of auricular cartilage in vitro and in vivo. Stem Cells Transl Med 2025; 14:szae098. [PMID: 40304399 PMCID: PMC12042163 DOI: 10.1093/stcltm/szae098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 10/11/2024] [Indexed: 05/02/2025] Open
Abstract
Patients suffering from microtia have limited treatment options for auricular reconstruction due to donor-site morbidity, complications, and unaesthetic outcome. Therefore, tissue engineering emerged as an alternative therapeutic option. Here, we generated and characterized human auricular cartilage using differentiated human adipose mesenchymal stem cells (hASCs) combined with human auricular chondrocytes. The differentiated hASCs were analysed for their morphology, phenotype, gene, and protein expression of chondrogenic markers, and biochemical composition at different time points in 2D and 3D in vitro. Importantly, we improved conditions for chondrogenic differentiation of hASCs in vitro to enhance their proliferation, survival, and deposition of cartilaginous-matrix proteins. In particular, gene expression analysis revealed an upregulation of cartilage oligomeric matrix protein (COMP) and aggrecan core protein (ACAN) in hASCs using the improved differentiation protocol in vitro. Additionally, we observed that co-seeding of hASCs with chondrocytes in a 1:5 ratio significantly enhanced the de novo auricular cartilage formation in a collagen-I bioink after 8 weeks on immunodeficient rat. In particular, the co-culture resulted in reduced shrinkage, and increased cartilage matrix production as confirmed by GAG deposition in vivo. Our results demonstrate that in co-cultures, hASCs stimulate cartilage formation due to a synergistic effect: hASCs' differentiation into chondrocytes and a trophic effect of hASCs on human auricular chondrocytes. Here we demonstrate the successful use of an hASC-chondrocyte co-culture technique for auricular cartilage tissue engineering in 3D collagen-I bioink. This co-culture approach omits the major drawbacks of traditional cartilage transplantation and thus, represents a fundamental step towards clinical translation.
Collapse
Affiliation(s)
- Dominika Zielinska
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
- Medical Faculty, University of Zurich, 8006 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
| | - Katarzyna Micka-Michalak
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
- Medical Faculty, University of Zurich, 8006 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
| | - Hyrije Ademi
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
- Medical Faculty, University of Zurich, 8006 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
| | - Philip Fisch
- Tissue Engineering and Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zurich, 8093 Zurich, Switzerland
| | - Roland Boeni
- White House Centre for Liposuction, 8044 Zurich, Switzerland
| | - Thomas Linder
- Otorhinolaryngology Clinic, Cantonal Hospital Luzern, 6000 Luzern, Switzerland
| | - Ueli Moehrlen
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
- Medical Faculty, University of Zurich, 8006 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
- Department of Pediatric Surgery, University Children’s Hospital Zurich, University of Zurich, 8008 Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
- Medical Faculty, University of Zurich, 8006 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
| | - Agnes S Klar
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
- Medical Faculty, University of Zurich, 8006 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8008 Zurich, Switzerland
| |
Collapse
|
2
|
Schiestl C, Neuhaus K, Meuli M, Farkas M, Hartmann-Fritsch F, Elrod J, Bressan J, Reichmann E, Böttcher-Haberzeth S. Long-Term Outcomes of a Cultured Autologous Dermo-Epidermal Skin Substitute in Children: 5-Year Results of a Phase I Clinical Trial. J Burn Care Res 2025; 46:326-334. [PMID: 39115183 PMCID: PMC11879726 DOI: 10.1093/jbcr/irae150] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Limited donor sites and poor long-term outcomes with standard treatment for large skin defects remain a huge problem. An autologous, bilayered, laboratory-grown skin substitute (denovoSkin) was developed to overcome this problem and has shown to be safe in 10 pediatric patients in a Phase I clinical trial after transplantation. The goal of this article was to report on 48-month long-term results. The pediatric participants of the phase I clinical trial were followed up at yearly visits up to 5 years after transplantation. Safety parameters, including the occurrence of adverse events, possible deviations of vital signs, and changes in concomitant therapy as well as additional parameters regarding skin stability, scar quality, and tumor formation, were assessed. Furthermore, scar maturation was photographically documented. Of the 10 patients treated with denovoSkin in this phase I clinical trial, 7 completed the 5-year follow-up period. Skin substitutes continued to be deemed safe, remained stable, and practically unchanged, with no sign of fragility and no tumor formation at clinical examination. Scar quality, captured using the Patient and Observer Scar Assessment Scale, was evaluated as close to normal skin. Transplantation of this laboratory-grown skin substitute in children is to date considered safe and shows encouraging functional and aesthetical long-term results close to normal skin. These results are promising and highlight the potential of a life-saving therapy for large skin defects. A multicentre, prospective, randomized, phase II clinical trial is currently ongoing to further evaluate the safety and efficacy of this novel skin substitute. Clinicaltrials.gov identifier NCT02145130.
Collapse
Affiliation(s)
- Clemens Schiestl
- Department of Surgery, Pediatric Burn Center, Children’s Skin Center, University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Department of Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Children’s Research Center (CRC), University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Faculty of Medicine, University of Zurich (UZH), Zurich 8006, Switzerland
| | - Kathrin Neuhaus
- Department of Surgery, Pediatric Burn Center, Children’s Skin Center, University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Department of Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Children’s Research Center (CRC), University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Faculty of Medicine, University of Zurich (UZH), Zurich 8006, Switzerland
| | - Martin Meuli
- Faculty of Medicine, University of Zurich (UZH), Zurich 8006, Switzerland
- CUTISS Ltd., Schlieren 8952, Switzerland
| | - Melinda Farkas
- Department of Surgery, Pediatric Burn Center, Children’s Skin Center, University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Department of Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Children’s Research Center (CRC), University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Faculty of Medicine, University of Zurich (UZH), Zurich 8006, Switzerland
| | - Fabienne Hartmann-Fritsch
- Faculty of Medicine, University of Zurich (UZH), Zurich 8006, Switzerland
- CUTISS Ltd., Schlieren 8952, Switzerland
| | - Julia Elrod
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Jenny Bressan
- Department of Surgery, Pediatric Burn Center, Children’s Skin Center, University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Department of Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Children’s Research Center (CRC), University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Faculty of Medicine, University of Zurich (UZH), Zurich 8006, Switzerland
| | - Ernst Reichmann
- Children’s Research Center (CRC), University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Faculty of Medicine, University of Zurich (UZH), Zurich 8006, Switzerland
- Department of Surgery, Tissue Biology Research Unit, University Children’s Hospital Zurich, University of Zurich, Schlieren 8952, Switzerland
| | - Sophie Böttcher-Haberzeth
- Department of Surgery, Pediatric Burn Center, Children’s Skin Center, University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Department of Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Children’s Research Center (CRC), University Children’s Hospital Zurich, University of Zurich, Zurich 8032, Switzerland
- Faculty of Medicine, University of Zurich (UZH), Zurich 8006, Switzerland
| |
Collapse
|
3
|
Sun M, Angelillo J, Hugues S. Lymphatic transport in anti-tumor immunity and metastasis. J Exp Med 2025; 222:e20231954. [PMID: 39969537 PMCID: PMC11837853 DOI: 10.1084/jem.20231954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/18/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Although lymphatic vessels (LVs) are present in many tumors, their importance in cancer has long been underestimated. In contrast to the well-studied tumor-associated blood vessels, LVs were previously considered to function as passive conduits for tumor metastasis. However, emerging evidence over the last two decades has shed light on their critical role in locally shaping the tumor microenvironment (TME). Here we review the involvement of LVs in tumor progression, metastasis, and modulation of anti-tumor immune response.
Collapse
Affiliation(s)
- Mengzhu Sun
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| | - Julien Angelillo
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| | - Stéphanie Hugues
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
4
|
Chiu A, Rutkowski JM, Zhang Q, Zhao F. Tissue-Engineered Therapeutics for Lymphatic Regeneration: Solutions for Myocardial Infarction and Secondary Lymphedema. Adv Healthc Mater 2025; 14:e2403551. [PMID: 39806804 PMCID: PMC11936459 DOI: 10.1002/adhm.202403551] [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: 09/18/2024] [Revised: 12/12/2024] [Indexed: 01/16/2025]
Abstract
The lymphatic system, which regulates inflammation and fluid homeostasis, is damaged in various diseases including myocardial infarction (MI) and breast-cancer-related lymphedema (BCRL). Mounting evidence suggests that restoring tissue fluid drainage and clearing excess immune cells by regenerating damaged lymphatic vessels can aid in cardiac repair and lymphedema amelioration. Current treatments primarily address symptoms rather than underlying causes due to a lack of regenerative therapies, highlighting the importance of the lymphatic system as a promising novel therapeutic target. Here cutting-edge research on engineered lymphatic tissues, growth factor therapies, and cell-based approaches designed to enhance lymphangiogenesis and restore lymphatic function is explored. Special focus is placed on how therapies with potential for immediate lymphatic reconstruction, originally designed for treating BCRL, can be applied to MI to augment cardiac repair and reduce heart failure risk. The integration of these novel treatments can significantly improve patient outcomes by promoting lymphatic repair, preventing pathological remodeling, and offering new avenues for managing lymphatic-associated diseases.
Collapse
Affiliation(s)
- Alvis Chiu
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 5045 Emerging Technologies Building 3120 TAMU, College Station, TX 77843-3120
| | - Joseph M. Rutkowski
- Department of Medical Physiology, College of Medicine, Texas A&M University, Medical Research and Education Building, 8447 Riverside Pkwy, Bryan, TX 77807-3260
| | - Qixu Zhang
- Department of Plastic Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, Texas 77030
| | - Feng Zhao
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 5045 Emerging Technologies Building 3120 TAMU, College Station, TX 77843-3120
| |
Collapse
|
5
|
Sierra-Sánchez Á, Sanabria-de la Torre R, Ubago-Rodríguez A, Quiñones-Vico MI, Montero-Vílchez T, Sánchez-Díaz M, Arias-Santiago S. Blood Plasma, Fibrinogen or Fibrin Biomaterial for the Manufacturing of Skin Tissue-Engineered Products and Other Dermatological Treatments: A Systematic Review. J Funct Biomater 2025; 16:79. [PMID: 40137358 PMCID: PMC11942893 DOI: 10.3390/jfb16030079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/09/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
The use of blood plasma, fibrinogen or fibrin, a natural biomaterial, has been widely studied for the development of different skin tissue-engineered products and other dermatological treatments. This systematic review reports the preclinical and clinical studies which use it alone or combined with other biomaterials and/or cells for the treatment of several dermatological conditions. Following the PRISMA 2020 Guidelines, 147 preclinical studies have revealed that the use of this biomaterial as a wound dressing or as a monolayer (one cell type) skin substitute are the preferred strategies, mainly for the treatment of excisional or surgical wounds. Moreover, blood plasma is mainly used alone although its combination with other biomaterials such as agarose, polyethylene glycol or collagen has also been reported to increase its wound healing potential. However, most of the 17 clinical reviewed evaluated its use for the treatment of severely burned patients as a wound dressing or bilayer (two cell types) skin substitute. Although the number of preclinical studies evaluating the use of blood plasma as a dermatological treatment has increased during the last fifteen years, this has not been correlated with a wide variety of clinical studies. Its safety and wound healing potential have been proved; however, the lack of a standard model and the presence of several approaches have meant that its translation to a clinical environment is still limited. A higher number of clinical studies should be carried out in the coming years to set a standard wound healing strategy for each dermatological disease.
Collapse
Affiliation(s)
- Álvaro Sierra-Sánchez
- Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Andalusian Network of Design and Translation of Advanced Therapies, 18014 Granada, Spain; (Á.S.-S.); (S.A.-S.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Dermatology, Virgen de las Nieves University Hospital, 18012 Granada, Spain
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NA 27101, USA
| | - Raquel Sanabria-de la Torre
- Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Andalusian Network of Design and Translation of Advanced Therapies, 18014 Granada, Spain; (Á.S.-S.); (S.A.-S.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Dermatology, Virgen de las Nieves University Hospital, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology IIi and Immunology, University of Granada, 18071 Granada, Spain
| | - Ana Ubago-Rodríguez
- Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Andalusian Network of Design and Translation of Advanced Therapies, 18014 Granada, Spain; (Á.S.-S.); (S.A.-S.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - María I. Quiñones-Vico
- Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Andalusian Network of Design and Translation of Advanced Therapies, 18014 Granada, Spain; (Á.S.-S.); (S.A.-S.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Dermatology, University of Granada, 18016 Granada, Spain
| | - Trinidad Montero-Vílchez
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Dermatology, Virgen de las Nieves University Hospital, 18012 Granada, Spain
| | - Manuel Sánchez-Díaz
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Dermatology, Virgen de las Nieves University Hospital, 18012 Granada, Spain
| | - Salvador Arias-Santiago
- Unidad de Producción Celular e Ingeniería Tisular, Virgen de las Nieves University Hospital, Andalusian Network of Design and Translation of Advanced Therapies, 18014 Granada, Spain; (Á.S.-S.); (S.A.-S.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Dermatology, Virgen de las Nieves University Hospital, 18012 Granada, Spain
- Department of Dermatology, University of Granada, 18016 Granada, Spain
| |
Collapse
|
6
|
Dravid AA, Singh A, García AJ. Biomaterial-Based Therapeutic Delivery of Immune Cells. Adv Healthc Mater 2025; 14:e2400586. [PMID: 38813869 PMCID: PMC11607182 DOI: 10.1002/adhm.202400586] [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: 02/16/2024] [Revised: 05/15/2024] [Indexed: 05/31/2024]
Abstract
Immune cell therapy (ICT) is a transformative approach used to treat a wide range of diseases including type 1 diabetes, sickle cell disease, disorders of the hematopoietic system, and certain forms of cancers. Despite excellent clinical successes, the scope of adoptively transferred immune cells is limited because of toxicities like cytokine release syndrome and immune effector cell-associated neurotoxicity in patients. Furthermore, reports suggest that such treatment can impact major organ systems including cardiac, renal, pulmonary, and hepatic systems in the long term. Additionally, adoptively transferred immune cells cannot achieve significant penetration into solid tissues, thus limiting their therapeutic potential. Recent studies suggest that biomaterial-assisted delivery of immune cells can address these challenges by reducing toxicity, improving localization, and maintaining desired phenotypes to eventually regain tissue function. In this review, recent efforts in the field of biomaterial-based immune cell delivery for the treatment of diseases, their pros and cons, and where these approaches stand in terms of clinical treatment are highlighted.
Collapse
Affiliation(s)
- Ameya A. Dravid
- Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Ankur Singh
- Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
- Petit Institute for Bioengineering and BioscienceGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Andrés J. García
- Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- Petit Institute for Bioengineering and BioscienceGeorgia Institute of TechnologyAtlantaGA30332USA
| |
Collapse
|
7
|
Rütsche D, Nanni M, Cheng P, Caflisch N, Tastanova A, Jenni C, Levesque MP, Moehrlen U, Klar AS, Biedermann T. Human Dermal Microvascular Arterial and Venous Blood Endothelial Cells and Their Use in Bioengineered Dermo-Epidermal Skin Substitutes. SMALL METHODS 2025:e2401588. [PMID: 39871784 DOI: 10.1002/smtd.202401588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/04/2025] [Indexed: 01/29/2025]
Abstract
The bioengineering of vascular networks is pivotal to create complex tissues and organs for regenerative medicine applications. However, bioengineered tissues comprising an arterial and venous plexus alongside a lymphatic capillary network have not been explored yet. Here, scRNA-seq is first employed to investigate the arterio-venous endothelial cell marker patterning in human fetal and juvenile skin. Transcriptomic analysis reveals that arterial and venous endothelial cell markers NRP1 (neuropilin 1) and NR2F2 (nuclear receptor subfamily 2 group F member 2) are broadly expressed in fetal and juvenile skin. In contrast, expression of NRP1 and NR2F2 on the protein level is cell-type specific and is retained in 2D (2-dimensional) cultures in vitro. Finally, distinct arterial and venous capillaries are bioengineered in 3D (3-dimensional) hydrogels and rapid anastomosis is demonstrated with the host vasculature in vivo. In summary, the bioengineering of human arterial, venous, and lymphatic capillaries is established, hence paving the way for these cells to be used in regenerative medicine and future clinical applications.
Collapse
Affiliation(s)
- Dominic Rütsche
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
| | - Monica Nanni
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Leonhardstrasse 21, Zurich, 8092, Switzerland
| | - Phil Cheng
- Department of Dermatology, University Hospital Zurich, Wagistrasse 18, Schlieren, 8952, Switzerland
| | - Nicolà Caflisch
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
| | - Aizhan Tastanova
- Department of Dermatology, University Hospital Zurich, Wagistrasse 18, Schlieren, 8952, Switzerland
| | - Céline Jenni
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, Wagistrasse 18, Schlieren, 8952, Switzerland
- Medical Faculty, University of Zurich, Rämistrasse 71, Zurich, 8006, Switzerland
| | - Ueli Moehrlen
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
- Medical Faculty, University of Zurich, Rämistrasse 71, Zurich, 8006, Switzerland
| | - Agnes S Klar
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
- Medical Faculty, University of Zurich, Rämistrasse 71, Zurich, 8006, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Lenggstrasse 30, Zurich, 8008, Switzerland
- Medical Faculty, University of Zurich, Rämistrasse 71, Zurich, 8006, Switzerland
| |
Collapse
|
8
|
Teertam SK, Setaluri V, Ayuso JM. Advances in Microengineered Platforms for Skin Research. JID INNOVATIONS 2025; 5:100315. [PMID: 39525704 PMCID: PMC11550131 DOI: 10.1016/j.xjidi.2024.100315] [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: 12/28/2023] [Revised: 08/01/2024] [Accepted: 08/26/2024] [Indexed: 11/16/2024] Open
Abstract
The skin plays a critical role in human physiology, acting both as a barrier to environmental insults and as a window to environmental stimuli. Disruption of this homeostasis leads to numerous skin disorders. Human and animal skin differ significantly, limiting the translational potential of animal-based investigations to advance therapeutics to human skin diseases. Hence, there is a critical need for physiologically relevant human skin models to explore novel treatment strategies. Recent advances in microfluidic technologies now allow design and generation of organ-on-chip devices that mimic critical features of tissue architecture. Skin-on-a-chip and microfluidic platforms hold promise as useful models for diverse dermatology applications. Compared with traditional in vitro models, microfluidic platforms offer improved control of fluid flow, which in turn allows precise manipulation of cell and molecular distribution. These properties enable the generation of multilayered in vitro models that mimic human skin structure while simultaneously offering superior control over nutrient and drug distribution. Researchers have used microfluidic platforms for a variety of applications in skin research, including epidermal-dermal cellular crosstalk, cell migration, mechanobiology, microbiome-immune response interactions, vascular biology, and wound healing. In this review, we comprehensively review state-of-the-art microfluidic models for skin research. We discuss the challenges and promise of current skin-on-a-chip technologies and provide a roadmap for future research in this active field.
Collapse
Affiliation(s)
- Sireesh Kumar Teertam
- Department of Dermatology, University of Wisconsin-Madison, Wisconsin, USA
- UW Carbone Cancer Center, Madison, Wisconsin, USA
| | - Vijayasaradhi Setaluri
- Department of Dermatology, University of Wisconsin-Madison, Wisconsin, USA
- UW Carbone Cancer Center, Madison, Wisconsin, USA
- William S. Middleton Memorial VA Hospital. Madison, Wisconsin, USA
| | - Jose M. Ayuso
- Department of Dermatology, University of Wisconsin-Madison, Wisconsin, USA
- UW Carbone Cancer Center, Madison, Wisconsin, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Wisconsin, USA
| |
Collapse
|
9
|
Fuchs B, Mert S, Hofmann D, Kuhlmann C, Birt A, Wiggenhauser PS, Giunta RE, Chavez MN, Nickelsen J, Schenck TL, Moellhoff N. Bioactivated scaffolds promote angiogenesis and lymphangiogenesis for dermal regeneration in vivo. J Tissue Eng 2025; 16:20417314251317542. [PMID: 40078220 PMCID: PMC11898032 DOI: 10.1177/20417314251317542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/17/2025] [Indexed: 03/14/2025] Open
Abstract
Chronic wounds represent an unresolved medical challenge with significant impact for patients' quality of life and global healthcare. Diverse in origin, ischemic-hypoxic and inflammatory conditions play central roles as pathological features that impede proper tissue regeneration. In this study, we propose an innovative approach to address this challenge. Our novel strategy utilizes photosynthetic biomaterials to restore the wound healing process firstly by promoting a normoxic, regeneration-supporting environment and secondly by mitigating inflammation through restoring lymphatic fluid transport and improving blood perfusion. We designed bioartificial scaffolds with photosynthetic cyanobacteria (Synechococcus sp. PCC 7002) and assessed their functional integration in a bilateral full-thickness skin defect on the backs of mice over a period of 7 days. Illuminated photosynthetic cyanobacteria facilitated local tissue oxygenation independent of blood perfusion. Additionally, genetic modification enabled the secretion of lymphangiogenic hyaluronic acid (HA) into the wound area. After 7 days, the scaffolds were explanted and histologically examined, assessing cell migration (HE staining) and protein expression (CD31, LYVE-1, VEGFR3, Ly6G, and F4/80). Results demonstrated successful colonization of bioartificial scaffolds with cyanobacteria. Following implantation into bilateral full-thickness skin defects, we observed an adherent vascularized basal layer beneath the bioactivated scaffolds after 7 days. Substantial increases in cell migration within bacteria-loaden scaffolds were noted, accompanied by a heightened expression of lymphatic (LYVE-1 and VEGFR3) and endothelial cell markers (CD31). Simultaneously, an augmented expression of acute (Ly6G) and late (F4/80) inflammatory proteins was observed. In summary, we developed a viable photosynthetic scaffold by integrating cyanobacteria into dermal regeneration materials (DRM), promoting the expression of lymphatic, endothelial, and inflammatory proteins under hypoxic conditions. The findings from this study represent a significant advancement in establishing autotrophic tissue engineering approaches, advocating for the use of photosynthetic cells in treating a broad spectrum of hypoxic conditions.
Collapse
Affiliation(s)
- Benedikt Fuchs
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sinan Mert
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Daniel Hofmann
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Constanze Kuhlmann
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Alexandra Birt
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Paul Severin Wiggenhauser
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Riccardo E Giunta
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Myra N Chavez
- Molecular Plant Science, Department Biology I, LMU Munich, Munich, Germany
| | - Jörg Nickelsen
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | | | - Nicholas Moellhoff
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
10
|
Chatzopoulou E, Bousaidi N, Guilbert T, Rucher G, Rose J, Germain S, Rouzet F, Chaussain C, Muller L, Gorin C. Multiscale Imaging to Monitor Functional SHED-Supported Engineered Vessels. J Dent Res 2024; 103:1392-1402. [PMID: 39290146 DOI: 10.1177/00220345241271122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Regeneration of orofacial tissues is hampered by the lack of adequate vascular supply. Implantation of in vitro engineered, prevascularized constructs has emerged as a strategy to allow the rapid vascularization of the entire graft. Given the angiogenic properties of dental pulp stem cells, we hereby established a preclinical model of prevascularized constructs loaded with stem cells from human exfoliating deciduous teeth (SHED) in a 3-dimensional-printed material and provided a functional analysis of their in vivo angiogenesis, vascular perfusion, and permeability. Three different cell-loaded collagen hydrogels (SHED-human umbilical vein endothelial cell [HUVEC], HUVEC with SHED-conditioned medium, and SHED alone) were cast in polylactic acid (PLA) grids and ectopically implanted in athymic mice. At day 10, in vivo positron emission tomography (PETscan) revealed a significantly increased uptake of radiotracer targeting activated endothelial cells in the SHED-HUVEC group compared to the other groups. At day 30, ex vivo micro-computed tomography imaging confirmed that SHED-HUVEC constructs had a significantly increased vascular volume compared to the other ones. Injection of species-specific lectins analyzed by 2-photon microscopy demonstrated blood perfusion of the engineered human vessels in both prevascularized groups. However, in vivo quantification showed increased vessel density in the SHED-HUVEC group. In addition, coinjection of fluorescent lectin and dextran revealed that prevascularization with SHED prevented vascular leakage, demonstrating the active role of SHED in the maturation of human-engineered microvascular networks. This preclinical study introduces a novel PLA prevascularized and implantable construct, along with an array of imaging techniques, to validate the ability of SHED to promote functional human-engineered vessels, further highlighting the interest of SHED for orofacial tissue engineering. Furthermore, this study validates the use of PETscan for the early detection of in vivo angiogenesis, which may be applied in the clinic to monitor the performance of prevascularized grafts.
Collapse
Affiliation(s)
- E Chatzopoulou
- Université Paris Cité, URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, Montrouge, France
- AP-HP, Services de médecine bucco-dentaire, FHU DDS-Net, GH Paris Nord et Paris Est, France
| | - N Bousaidi
- Université Paris Cité, URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, Montrouge, France
| | - T Guilbert
- Université Paris Cité, CNRS, INSERM U1016, Institut Cochin, Paris, France
| | - G Rucher
- Université Paris Cité, LVTS, INSERM U1148, France
- Université Paris Cité, UMS 34-FRIM, France
| | - J Rose
- AP-HP, Département de médecine nucléaire, Hôpital Bichat, Paris, France
| | - S Germain
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR7241, INSERM U1050, Université PSL, Paris, France
| | - F Rouzet
- Université Paris Cité, LVTS, INSERM U1148, France
- AP-HP, Département de médecine nucléaire, Hôpital Bichat, Paris, France
| | - C Chaussain
- Université Paris Cité, URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, Montrouge, France
- AP-HP, Services de médecine bucco-dentaire, FHU DDS-Net, GH Paris Nord et Paris Est, France
| | - L Muller
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR7241, INSERM U1050, Université PSL, Paris, France
| | - C Gorin
- Université Paris Cité, URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, Montrouge, France
- AP-HP, Services de médecine bucco-dentaire, FHU DDS-Net, GH Paris Nord et Paris Est, France
| |
Collapse
|
11
|
Zhou X, Yu X, You T, Zhao B, Dong L, Huang C, Zhou X, Xing M, Qian W, Luo G. 3D Printing-Based Hydrogel Dressings for Wound Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404580. [PMID: 39552255 DOI: 10.1002/advs.202404580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 10/21/2024] [Indexed: 11/19/2024]
Abstract
Skin wounds have become an important issue that affects human health and burdens global medical care. Hydrogel materials similar to the natural extracellular matrix (ECM) are one of the best candidates for ideal wound dressings and the most feasible choices for printing inks. Distinct from hydrogels made by traditional technologies, which lack bionic and mechanical properties, 3D printing can promptly and accurately create hydrogels with complex bioactive structures and the potential to promote tissue regeneration and wound healing. Herein, a comprehensive review of multi-functional 3D printing-based hydrogel dressings for wound healing is presented. The review first summarizes the 3D printing techniques for wound hydrogel dressings, including photo-curing, extrusion, inkjet, and laser-assisted 3D printing. Then, the properties and design approaches of a series of bioinks composed of natural, synthetic, and composite polymers for 3D printing wound hydrogel dressings are described. Thereafter, the application of multi-functional 3D printing-based hydrogel dressings in a variety of wound environments is discussed in depth, including hemostasis, anti-inflammation, antibacterial, skin appendage regeneration, intelligent monitoring, and machine learning-assisted therapy. Finally, the challenges and prospects of 3D printing-based hydrogel dressings for wound healing are presented.
Collapse
Affiliation(s)
- Xuan Zhou
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China
| | - Xunzhou Yu
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China
| | - Tingting You
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China
| | - Baohua Zhao
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China
| | - Lanlan Dong
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China
| | - Can Huang
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China
| | - Xiaoqing Zhou
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Wei Qian
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China
| | - Gaoxing Luo
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China
| |
Collapse
|
12
|
Meng S, Borjihan Q, Xiao D, Wang Y, Chen M, Cheng C, Dong A. Biosynthesis of positively charged bacterial cellulose hydrogel with antibacterial and anti-inflammatory function for efficient wound healing. Int J Biol Macromol 2024; 279:135263. [PMID: 39244128 DOI: 10.1016/j.ijbiomac.2024.135263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/25/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
In bacterial cellulose (BC)-based living materials, the effective and permanent incorporation of bactericidal agents into BC remains a persistent challenge. In this study, midazole quaternary ammonium salt was grafted onto a dispersion of bacterial cellulose, which was subsequently directly added to the fermentation medium of BC-producing bacteria to obtain BC-based hydrogel materials (BC/BC-[PQVI]Br) with inherent antibacterial properties. The BC/BC-[PQVI]Br hydrogel prepared in this study exhibits favorable tensile properties, with a maximum tensile stress of 970 KPa and water retention for up to 6 h. Moreover, it demonstrates acceptable antibacterial activity against S. aureus (93 %) and E. coli (71 %), respectively. Additionally, the hydrogel displays a high cell survival rate of 98 % after contact with NIH 3T3 cells, indicating its non-cytotoxic nature. Furthermore, the mouse wound experiment confirms the excellent wound healing effect of the hydrogel. This research presents an innovative approach towards developing environmentally friendly active wound dressings with microbial-derived antibacterial functionality.
Collapse
Affiliation(s)
- Suriguga Meng
- Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010021, China
| | - Qinggele Borjihan
- Key Laboratory of Dairy Biotechnology and Engineering, Education Ministry of People's Republic of China, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Douxin Xiao
- Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010021, China.
| | - Yutian Wang
- Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010021, China
| | - Mei Chen
- Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010021, China
| | - Chunzu Cheng
- China Textile Academy, State Key Laboratory of Biobased Fiber Manufacturing Technology, Beijing 100025, China.
| | - Alideertu Dong
- Engineering Research Center of Dairy Quality and Safety Control Technology, Ministry of Education, College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot 010021, China.
| |
Collapse
|
13
|
Iqbal MZ, Riaz M, Biedermann T, Klar AS. Breathing new life into tissue engineering: exploring cutting-edge vascularization strategies for skin substitutes. Angiogenesis 2024; 27:587-621. [PMID: 38842751 PMCID: PMC11564345 DOI: 10.1007/s10456-024-09928-6] [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: 07/20/2023] [Accepted: 05/02/2024] [Indexed: 06/07/2024]
Abstract
Tissue-engineered skin substitutes (TESS) emerged as a new therapeutic option to improve skin transplantation. However, establishing an adequate and rapid vascularization in TESS is a critical factor for their clinical application and successful engraftment in patients. Therefore, several methods have been applied to improve the vascularization of skin substitutes including (i) modifying the structural and physicochemical properties of dermal scaffolds; (ii) activating biological scaffolds with growth factor-releasing systems or gene vectors; and (iii) developing prevascularized skin substitutes by loading scaffolds with capillary-forming cells. This review provides a detailed overview of the most recent and important developments in the vascularization strategies for skin substitutes. On the one hand, we present cell-based approaches using stem cells, microvascular fragments, adipose tissue derived stromal vascular fraction, endothelial cells derived from blood and skin as well as other pro-angiogenic stimulation methods. On the other hand, we discuss how distinct 3D bioprinting techniques and microfluidics, miRNA manipulation, cell sheet engineering and photosynthetic scaffolds like GelMA, can enhance skin vascularization for clinical applications. Finally, we summarize and discuss the challenges and prospects of the currently available vascularization techniques that may serve as a steppingstone to a mainstream application of skin tissue engineering.
Collapse
Affiliation(s)
- M Zohaib Iqbal
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Mahrukh Riaz
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Agnes S Klar
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland.
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
| |
Collapse
|
14
|
Rhee S, Xia C, Chandra A, Hamon M, Lee G, Yang C, Guo Z, Sun B. Full-Thickness Perfused Skin-on-a-Chip with In Vivo-Like Drug Response for Drug and Cosmetics Testing. Bioengineering (Basel) 2024; 11:1055. [PMID: 39593715 PMCID: PMC11591533 DOI: 10.3390/bioengineering11111055] [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: 10/03/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
In this study, we present a novel 3D perfused skin-on-a-chip model fabricated using micro-precision 3D printing, which offers a streamlined and reproducible approach for incorporating perfusion. Perfused skin models are well-regarded for their advantages, such as improved nutrient supply, enhanced barrier function, and prolonged tissue viability. However, current models often require complex setups, such as self-assembled endothelial cells or sacrificial rods, which are prone to variability and time-consuming. Our model uses projection micro-stereolithography 3D printing to create precise microcapillary-like channels using a biocompatible resin, overcoming the drug-absorbing properties of PDMS. A customized chip holder allows for the simultaneous culture of six perfused chips, enabling high-throughput testing. The engineered skin-on-a-chip features distinct dermis and epidermis layers, confirmed via H&E staining and immunostaining. To evaluate drug screening capabilities, inflammation was induced using TNF-α and treated with dexamethasone, with cytokine levels compared to 2D cultures and human skin biopsies. Our 3D model exhibited drug response trends similar to human skin, while showing reduced cytotoxicity over time compared to biopsies. This perfused skin-on-a-chip provides a reliable, physiologically relevant alternative for drug and cosmetics screening, simplifying perfusion setup while preserving key benefits.
Collapse
Affiliation(s)
- Stephen Rhee
- BMF Biotechnology, Inc., San Diego, CA 92121, USA
| | - Chunguang Xia
- BMF Biotechnology, Inc., San Diego, CA 92121, USA
- BMF Nano Material Technology Co., Ltd., Shenzhen 518100, China; (C.Y.)
| | | | - Morgan Hamon
- BMF Biotechnology, Inc., San Diego, CA 92121, USA
| | - Geonhui Lee
- BMF Biotechnology, Inc., San Diego, CA 92121, USA
| | - Chen Yang
- BMF Nano Material Technology Co., Ltd., Shenzhen 518100, China; (C.Y.)
| | - Zaixun Guo
- BMF Nano Material Technology Co., Ltd., Shenzhen 518100, China; (C.Y.)
| | - Bingjie Sun
- BMF Biotechnology, Inc., San Diego, CA 92121, USA
| |
Collapse
|
15
|
Alderfer L, Saha S, Fan F, Wu J, Littlepage LE, Hanjaya-Putra D. Multi-parameter tunable synthetic matrix for engineering lymphatic vessels. Commun Biol 2024; 7:1262. [PMID: 39367247 PMCID: PMC11452684 DOI: 10.1038/s42003-024-06935-7] [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: 11/22/2023] [Accepted: 09/20/2024] [Indexed: 10/06/2024] Open
Abstract
Controlling the formation of new lymphatic vessels has been postulated as an innovative therapeutic strategy for various disease phenotypes, including neurodegenerative diseases, metabolic syndrome, cardiovascular disease, and lymphedema. Yet, compared to the blood vascular system, little is known about the molecular regulation that controls lymphatic tube formation in a synthetic matrix. In this study, we utilize hyaluronic acid (HA)-hydrogels to design a novel platform for decoupled investigation into how mechanical and biochemical cues regulate lymphatic vessel formation in a synthetic matrix. Using HA and controlling the degree of modification provides a method to preserve and modulate key lymphatic markers Prox1, LYVE-1, and Pdpn. The chemistry of the system allows for spatial and temporal patterning of specific peptides and substrate stiffnesses, and an MMP-sensitive crosslinker allowed cells to degrade and remodel their matrix. Through systematic optimization of multiple parameters, we have designed a system that allows human lymphatic endothelial cells (LECs) to self-assemble into vessels in vitro within 3 days. These engineered vessels can be cultured for up to 3 weeks and can be used for high-throughput mechanistic studies, or can be implanted into immunodeficient mice where they have demonstrated the ability to integrate and mature. Collectively, these studies report a novel, fully-defined 3D synthetic matrix system capable of generating lymphatic vessels in vitro that provide promise as an in vitro screening platform and as a therapeutic vessel transplant, which to our knowledge, is the first ever 3D lymphatic tissue engineering approach to not require the use of support cells.
Collapse
Affiliation(s)
- Laura Alderfer
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, USA
| | - Sanjoy Saha
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, USA
| | - Fei Fan
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, USA
| | - Junmin Wu
- Department of Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, USA
| | - Laurie E Littlepage
- Department of Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, USA
| | - Donny Hanjaya-Putra
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, USA.
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, USA.
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, USA.
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA.
| |
Collapse
|
16
|
Jian Y, Li Y, Zhang Y, Tang M, Deng M, Liu C, Cheng M, Xiao S, Deng C, Wei Z. Lymphangiogenesis: novel strategies to promote cutaneous wound healing. BURNS & TRAUMA 2024; 12:tkae040. [PMID: 39328366 PMCID: PMC11427083 DOI: 10.1093/burnst/tkae040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 09/28/2024]
Abstract
The cutaneous lymphatic system regulates tissue inflammation, fluid balance and immunological responses. Lymphangiogenesis or lymphatic dysfunction may lead to lymphedema, immune deficiency, chronic inflammation etc. Tissue regeneration and healing depend on angiogenesis and lymphangiogenesis during wound healing. Tissue oedema and chronic inflammation can slow wound healing due to impaired lymphangiogenesis or lymphatic dysfunction. For example, impaired lymphangiogenesis or lymphatic dysfunction has been detected in nonhealing wounds such as diabetic ulcers, venous ulcers and bedsores. This review summarizes the structure and function of the cutaneous lymphatic vessel system and lymphangiogenesis in wounds. Furthermore, we review wound lymphangiogenesis processes and remodelling, especially the influence of the inflammatory phase. Finally, we outline how to control lymphangiogenesis to promote wound healing, assess the possibility of targeting lymphangiogenesis as a novel treatment strategy for chronic wounds and provide an analysis of the possible problems that need to be addressed.
Collapse
Affiliation(s)
- Yang Jian
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Yanqi Li
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Yanji Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Mingyuan Tang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Mingfu Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Chenxiaoxiao Liu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Maolin Cheng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
| | - Shune Xiao
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Chengliang Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| | - Zairong Wei
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Hui chuan District, Zunyi, Guizhou, 563003, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi, Guizhou, 563003, China
| |
Collapse
|
17
|
Arriola-Alvarez I, Jaunarena I, Izeta A, Lafuente H. Progenitor Cell Sources for 3D Bioprinting of Lymphatic Vessels and Potential Clinical Application. Tissue Eng Part A 2024; 30:353-366. [PMID: 37950710 DOI: 10.1089/ten.tea.2023.0204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2023] Open
Abstract
The lymphatic system maintains tissue fluid homeostasis and it is involved in the transport of nutrients and immunosurveillance. It also plays a pivotal role in both pathological and regenerative processes. Lymphatic development in the embryo occurs by polarization and proliferation of lymphatic endothelial cells from the lymph sacs, that is, lymphangiogenesis. Alternatively, lymphvasculogenesis further contributes to the formation of lymphatic vessels. In adult tissues, lymphatic formation rarely occurs under physiological conditions, being restricted to pathological processes. In lymphvasculogenesis, progenitor cells seem to be a source of lymphatic vessels. Indeed, mesenchymal stem cells, adipose stem cells, endothelial progenitor cells, and colony-forming endothelial cells are able to promote lymphatic regeneration by different mechanisms, such as direct differentiation and paracrine effects. In this review, we summarize what is known on the diverse stem/progenitor cell niches available for the lymphatic system, emphasizing the potential that these cells hold for lymphatic tissue engineering through 3D bioprinting and their translation to clinical application.
Collapse
Affiliation(s)
- Inazio Arriola-Alvarez
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
| | - Ibon Jaunarena
- Gynecology Oncology Unit, Donostia University Hospital, Donostia-San Sebastián, Spain
- Obstetrics and Gynaecology Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
- University of the Basque Country (UPV/EHU), Department of Medical Surgical Specialties, Leioa, Spain
| | - Ander Izeta
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
- Department of Biomedical Engineering and Sciences, Tecnun-University of Navarra, Donostia-San Sebastián, Spain
| | - Héctor Lafuente
- Tissue Engineering Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain
| |
Collapse
|
18
|
Pal D, Das P, Mukherjee P, Roy S, Chaudhuri S, Kesh SS, Ghosh D, Nandi SK. Biomaterials-Based Strategies to Enhance Angiogenesis in Diabetic Wound Healing. ACS Biomater Sci Eng 2024; 10:2725-2741. [PMID: 38630965 DOI: 10.1021/acsbiomaterials.4c00216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Amidst the present healthcare issues, diabetes is unique as an emerging class of affliction with chronicity in a majority of the population. To check and control its effects, there have been huge turnover and constant development of management strategies, and though a bigger part of the health care area is involved in achieving its control and the related issues such as the effect of diabetes on wound healing and care and many of the works have reached certain successful outcomes, still there is a huge lack in managing it, with maximum effect yet to be attained. Studying pathophysiology and involvement of various treatment options, such as tissue engineering, application of hydrogels, drug delivery methods, and enhancing angiogenesis, are at constantly developing stages either direct or indirect. In this review, we have gathered a wide field of information and different new therapeutic methods and targets for the scientific community, paving the way toward more settled ideas and research advances to cure diabetic wounds and manage their outcomes.
Collapse
Affiliation(s)
- Debajyoti Pal
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Pratik Das
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Prasenjit Mukherjee
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Subhasis Roy
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Shubhamitra Chaudhuri
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Shyam Sundar Kesh
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Debaki Ghosh
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Samit Kumar Nandi
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| |
Collapse
|
19
|
Xu C, Hu L, Zeng J, Wu A, Deng S, Zhao Z, Geng K, Luo J, Wang L, Zhou X, Huang W, Long Y, Song J, Zheng S, Wu J, Chen Q. Gynura divaricata (L.) DC. promotes diabetic wound healing by activating Nrf2 signaling in diabetic rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117638. [PMID: 38135237 DOI: 10.1016/j.jep.2023.117638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/08/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
THE ETHNOPHARMACOLOGICAL SIGNIFICANCE Diabetic chronic foot ulcers pose a significant therapeutic challenge as a result of the oxidative stress caused by hyperglycemia. Which impairs angiogenesis and delays wound healing, potentially leading to amputation. Gynura divaricata (L.) DC. (GD), a traditional Chinese herbal medicine with hypoglycemic effects, has been proposed as a potential therapeutic agent for diabetic wound healing. However, the underlying mechanisms of its effects remain unclear. AIM OF THE STUDY In this study, we aimed to reveal the effect and potential mechanisms of GD on accelerating diabetic wound healing in vitro and in vivo. MATERIALS AND METHODS The effects of GD on cell proliferation, apoptosis, reactive oxygen species (ROS) production, migration, mitochondrial membrane potential (MMP), and potential molecular mechanisms were investigated in high glucose (HG) stimulated human umbilical vein endothelial cells (HUVECs) using CCK-8, flow cytometry assay, wound healing assay, immunofluorescence, DCFH-DA staining, JC-1 staining, and Western blot. Full-thickness skin defects were created in STZ-induced diabetic rats, and wound healing rate was tracked by photographing them every day. HE staining, immunohistochemistry, and Western blot were employed to investigate the effect and molecular mechanism of GD on wound healing in diabetic rats. RESULTS GD significantly improved HUVEC survival, decreased apoptosis, lowered ROS production, restored MMP, improved migration ability, and raised VEGF expression. The use of Nrf2-siRNA completely abrogated these effects. Topical application of GD promoted angiogenesis and granulation tissue growth, resulting in faster healing of diabetic wounds. The expression of VEGF, CD31, and VEGFR was elevated in the skin tissue of diabetic rats after GD treatment, which upregulated HO-1, NQO-1, and Bcl-2 expression while downregulating Bax expression via activation of the Nrf2 signaling pathway. CONCLUSION The findings of this study indicate that GD has the potential to serve as a viable alternative treatment for diabetic wounds. This potential arises from its ability to mitigate the negative effects of oxidative stress on angiogenesis, which is regulated by the Nrf2 signaling pathway. The results of our study offer valuable insights into the therapeutic efficacy of GD in the treatment of diabetic wounds, emphasizing the significance of directing interventions towards the Nrf2 signaling pathway to mitigate oxidative stress and facilitate the process of angiogenesis.
Collapse
Affiliation(s)
- Caimin Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China; School of Nursing, Southwest Medical University, Luzhou, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Southwest Medical University
| | - Lixin Hu
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China; School of Nursing, Southwest Medical University, Luzhou, China
| | - Jing Zeng
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Anguo Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Shilong Deng
- Department of Nursing, The Affiliated Hospital, Southwest Medical University, Luzhou, China; Wound Healing Basic Research and Clinical Application Key Laboratory, School of Nursing, Southwest Medical University, LuZhou, China
| | - Zijuan Zhao
- Department of Nursing, The Affiliated Hospital, Southwest Medical University, Luzhou, China; Wound Healing Basic Research and Clinical Application Key Laboratory, School of Nursing, Southwest Medical University, LuZhou, China
| | - Kang Geng
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jiesi Luo
- School of Basic Medicine Sciences, Southwest Medical University, Luzhou, China
| | - Long Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaogang Zhou
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Wei Huang
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Southwest Medical University
| | - Yang Long
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Southwest Medical University
| | - Jianying Song
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China; School of Nursing, Southwest Medical University, Luzhou, China
| | - Silin Zheng
- Department of Nursing, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jianming Wu
- School of Basic Medicine Sciences, Southwest Medical University, Luzhou, China.
| | - Qi Chen
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China; School of Nursing, Southwest Medical University, Luzhou, China; Department of Nursing, The Affiliated Hospital, Southwest Medical University, Luzhou, China; Wound Healing Basic Research and Clinical Application Key Laboratory, School of Nursing, Southwest Medical University, LuZhou, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Southwest Medical University.
| |
Collapse
|
20
|
Mazio C, Mavaro I, Palladino A, Casale C, Urciuolo F, Banfi A, D'Angelo L, Netti PA, de Girolamo P, Imparato G, Attanasio C. Rapid innervation and physiological epidermal regeneration by bioengineered dermis implanted in mouse. Mater Today Bio 2024; 25:100949. [PMID: 38298559 PMCID: PMC10827562 DOI: 10.1016/j.mtbio.2024.100949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/02/2024] [Accepted: 01/06/2024] [Indexed: 02/02/2024] Open
Abstract
Tissue-engineered skin substitutes are promising tools to cover large and deep skin defects. However, the lack of a synergic and fast regeneration of the vascular network, nerves, and skin appendages limits complete skin healing and impairs functional recovery. It has been highlighted that an ideal skin substitute should mimic the structure of the native tissue to enhance clinical effectiveness. Here, we produced a pre-vascularized dermis (PVD) comprised of fibroblasts embedded in their own extracellular matrix (ECM) and a capillary-like network. Upon implantation in a mouse full-thickness skin defect model, we observed a very early innervation of the graft in 2 weeks. In addition, mouse capillaries and complete epithelialization were detectable as early as 1 week after implantation and, skin appendages developed in 2 weeks. These anatomical features underlie the interaction with the skin nerves, thus providing a further cue for reinnervation guidance. Further, the graft displays mechanical properties, collagen density, and assembly features very similar to the host tissue. Taken together our data show that the pre-existing ECM components of the PVD, physiologically organized and assembled similarly to the native tissue, support a rapid regeneration of dermal tissue. Therefore, our results suggest a promising potential for PVD in skin regeneration.
Collapse
Affiliation(s)
- Claudia Mazio
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for HealthCare@CRIB, Italy
| | - Isabella Mavaro
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for HealthCare@CRIB, Italy
- University of Naples Federico II, Department of Veterinary Medicine and Animal Production, Italy
| | - Antonio Palladino
- University of Naples Federico II, Department of Agricultural Sciences, Italy
| | - Costantino Casale
- University of Naples Federico II, Interdisciplinary Research Centre on Biomaterials (CRIB), Italy
| | - Francesco Urciuolo
- University of Naples Federico II, Department of Chemical, Materials and Industrial Production Engineering, Italy
| | - Andrea Banfi
- Basel University Hospital and University of Basel, Department of Biomedicine, Switzerland
| | - Livia D'Angelo
- University of Naples Federico II, Department of Veterinary Medicine and Animal Production, Italy
| | - Paolo A. Netti
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for HealthCare@CRIB, Italy
- University of Naples Federico II, Interdisciplinary Research Centre on Biomaterials (CRIB), Italy
- University of Naples Federico II, Department of Chemical, Materials and Industrial Production Engineering, Italy
| | - Paolo de Girolamo
- University of Naples Federico II, Department of Veterinary Medicine and Animal Production, Italy
| | - Giorgia Imparato
- Istituto Italiano di Tecnologia, Center for Advanced Biomaterials for HealthCare@CRIB, Italy
| | - Chiara Attanasio
- University of Naples Federico II, Department of Veterinary Medicine and Animal Production, Italy
| |
Collapse
|
21
|
Rimal R, Muduli S, Desai P, Marquez AB, Möller M, Platzman I, Spatz J, Singh S. Vascularized 3D Human Skin Models in the Forefront of Dermatological Research. Adv Healthc Mater 2024; 13:e2303351. [PMID: 38277705 PMCID: PMC11468127 DOI: 10.1002/adhm.202303351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/04/2023] [Indexed: 01/28/2024]
Abstract
In vitro engineered skin models are emerging as an alternative platform to reduce and replace animal testing in dermatological research. Despite the progress made in recent years, considerable challenges still exist for the inclusion of diverse cell types within skin models. Blood vessels, in particular, are essential in maintaining tissue homeostasis and are one of many primary contributors to skin disease inception and progression. Substantial efforts in the past have allowed the successful fabrication of vascularized skin models that are currently utilized for disease modeling and drugs/cosmetics testing. This review first discusses the need for vascularization within tissue-engineered skin models, highlighting their role in skin grafting and disease pathophysiology. Second, the review spotlights the milestones and recent progress in the fabrication and utilization of vascularized skin models. Additionally, advances including the use of bioreactors, organ-on-a-chip devices, and organoid systems are briefly explored. Finally, the challenges and future outlook for vascularized skin models are addressed.
Collapse
Affiliation(s)
- Rahul Rimal
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Saradaprasan Muduli
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
| | - Prachi Desai
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Andrea Bonnin Marquez
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Martin Möller
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Ilia Platzman
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 22569120HeidelbergGermany
| | - Joachim Spatz
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 22569120HeidelbergGermany
- Max Planck School Matter to LifeJahnstrasse 2969120HeidelbergGermany
| | - Smriti Singh
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
| |
Collapse
|
22
|
Peng Y, Guo Y, Ge X, Gong Y, Wang Y, Ou Z, Luo G, Zhan R, Zhang Y. Construction of programmed time-released multifunctional hydrogel with antibacterial and anti-inflammatory properties for impaired wound healing. J Nanobiotechnology 2024; 22:126. [PMID: 38519957 PMCID: PMC10960406 DOI: 10.1186/s12951-024-02390-y] [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: 12/13/2023] [Accepted: 03/08/2024] [Indexed: 03/25/2024] Open
Abstract
The successful reprogramming of impaired wound healing presents ongoing challenges due to the impaired tissue microenvironment caused by severe bacterial infection, excessive oxidative stress, as well as the inappropriate dosage timing during different stages of the healing process. Herein, a dual-layer hydrogel with sodium alginate (SA)-loaded zinc oxide (ZnO) nanoparticles and poly(N-isopropylacrylamide) (PNIPAM)-loaded Cu5.4O ultrasmall nanozymes (named programmed time-released multifunctional hydrogel, PTMH) was designed to dynamically regulate the wound inflammatory microenvironment based on different phases of wound repairing. PTMH combated bacteria at the early phase of infection by generating reactive oxygen species through ZnO under visible-light irradiation with gradual degradation of the lower layer. Subsequently, when the upper layer was in direct contact with the wound tissue, Cu5.4O ultrasmall nanozymes were released to scavenge excessive reactive oxygen species. This neutralized a range of inflammatory factors and facilitated the transition from the inflammatory phase to the proliferative phase. Furthermore, the utilization of Cu5.4O ultrasmall nanozymes enhanced angiogenesis, thereby facilitating the delivery of oxygen and nutrients to the impaired tissue. Our experimental findings indicate that PTMHs promote the healing process of diabetic wounds with bacterial infection in mice, exhibiting notable antibacterial and anti-inflammatory properties over a specific period of time.
Collapse
Affiliation(s)
- Yuan Peng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Yicheng Guo
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xin Ge
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Yali Gong
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yuhan Wang
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zelin Ou
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Gaoxing Luo
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Rixing Zhan
- Institute of Burn Research, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Yixin Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, China.
| |
Collapse
|
23
|
Pereira L, Echarte L, Romero M, Grazioli G, Pérez-Campos H, Francia A, Vicentino W, Mombrú AW, Faccio R, Álvarez I, Touriño C, Pardo H. Synthesis and characterization of a bovine collagen: GAG scaffold with Uruguayan raw material for tissue engineering. Cell Tissue Bank 2024; 25:123-142. [PMID: 34536180 DOI: 10.1007/s10561-021-09960-6] [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: 07/11/2020] [Accepted: 09/06/2021] [Indexed: 11/28/2022]
Abstract
Tissue engineering (TE) and regenerative medicine offer strategies to improve damaged tissues by using scaffolds and cells. The use of collagen-based biomaterials in the field of TE has been intensively growing over the past decades. Mesenchymal stromal cells (MSCs) and dental pulp stem cells (DPSCs) are promising cell candidates for development of clinical composites. In this study, we proposed the development of a bovine collagen type I: chondroitin-6-sulphate (CG) scaffold, obtained from Uruguayan raw material (certified as free bovine spongiform encephalopathy), with CG crosslinking enhancement using different gamma radiation doses. Structural, biomechanical and chemical characteristics of the scaffolds were assessed by Scanning Electron Microscopy, axial tensile tests, FT-IR and Raman Spectroscopy, respectively. Once we selected the most appropriate scaffold for future use as a TE product, we studied the behavior of MSCs and DPSCs cultured on the scaffold by cytotoxicity, proliferation and differentiation assays. Among the diverse porous scaffolds obtained, the one with the most adequate properties was the one exposed to 15 kGy of gamma radiation. This radiation dose contributed to the crosslinking of molecules, to the formation of new bonds and/or to the reorganization of the collagen fibers. The selected scaffold was non-cytotoxic for the tested cells and a suitable substrate for cell proliferation. Furthermore, the scaffold allowed MSCs differentiation to osteogenic, chondrogenic, and adipogenic lineages. Thus, this work shows a promising approach to the synthesis of a collagen-scaffold suitable for TE.
Collapse
Affiliation(s)
- L Pereira
- Centro NanoMat, Facultad de Química, Instituto Polo Tecnológico de Pando, UdelaR, Camino Aparicio Saravia s/n, 9100, Pando, Canelones, Uruguay
| | - L Echarte
- Área Terapia Celular y Medicina Regenerativa (ATCMR), Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - M Romero
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - G Grazioli
- Cátedra de Materiales Dentales, Facultad de Odontología, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - H Pérez-Campos
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - A Francia
- Fisiología general y bucodental, Facultad de Odontología, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - W Vicentino
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - A W Mombrú
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - R Faccio
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - I Álvarez
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - C Touriño
- Área Terapia Celular y Medicina Regenerativa (ATCMR), Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay.
| | - H Pardo
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay.
| |
Collapse
|
24
|
Jiang Y, Perez-Moreno M. Translational frontiers: insight from lymphatics in skin regeneration. Front Physiol 2024; 15:1347558. [PMID: 38487264 PMCID: PMC10937408 DOI: 10.3389/fphys.2024.1347558] [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: 12/01/2023] [Accepted: 02/01/2024] [Indexed: 03/17/2024] Open
Abstract
The remarkable regenerative ability of the skin, governed by complex molecular mechanisms, offers profound insights into the skin repair processes and the pathogenesis of various dermatological conditions. This understanding, derived from studies in human skin and various model systems, has not only deepened our knowledge of skin regeneration but also facilitated the development of skin substitutes in clinical practice. Recent research highlights the crucial role of lymphatic vessels in skin regeneration. Traditionally associated with fluid dynamics and immune modulation, these vessels are now recognized for interacting with skin stem cells and coordinating regeneration. This Mini Review provides an overview of recent advancements in basic and translational research related to skin regeneration, focusing on the dynamic interplay between lymphatic vessels and skin biology. Key highlights include the critical role of stem cell-lymphatic vessel crosstalk in orchestrating skin regeneration, emerging translational approaches, and their implications for skin diseases. Additionally, the review identifies research gaps and proposes potential future directions, underscoring the significance of this rapidly evolving research arena.
Collapse
Affiliation(s)
| | - Mirna Perez-Moreno
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Zhu J, Abaci HE. Human skin-on-a-chip for mpox pathogenesis studies and preclinical drug evaluation. Trends Pharmacol Sci 2023; 44:865-868. [PMID: 37500295 PMCID: PMC10811284 DOI: 10.1016/j.tips.2023.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/13/2023] [Accepted: 07/04/2023] [Indexed: 07/29/2023]
Abstract
Timely intervention of preventative and therapeutic measures abated a 2022 mpox global outbreak. However, the high transmissibility and unique pathological characteristics of mpox demand further investigation. Here, we discuss the potentials of human skin-on-a-chip as a valuable model for mpox disease evaluation, to achieve in-depth physiological understanding and desirable therapeutic predictive capabilities.
Collapse
Affiliation(s)
- Jia Zhu
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA; Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, USA; Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| | - Hasan E Abaci
- Department of Dermatology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
26
|
Wistner SC, Rashad L, Slaughter G. Advances in tissue engineering and biofabrication for in vitro skin modeling. BIOPRINTING (AMSTERDAM, NETHERLANDS) 2023; 35:e00306. [PMID: 38645432 PMCID: PMC11031264 DOI: 10.1016/j.bprint.2023.e00306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The global prevalence of skin disease and injury is continually increasing, yet conventional cell-based models used to study these conditions do not accurately reflect the complexity of human skin. The lack of inadequate in vitro modeling has resulted in reliance on animal-based models to test pharmaceuticals, biomedical devices, and industrial and environmental toxins to address clinical needs. These in vivo models are monetarily and morally expensive and are poor predictors of human tissue responses and clinical trial outcomes. The onset of three-dimensional (3D) culture techniques, such as cell-embedded and decellularized approaches, has offered accessible in vitro alternatives, using innovative scaffolds to improve cell-based models' structural and histological authenticity. However, these models lack adequate organizational control and complexity, resulting in variations between structures and the exclusion of physiologically relevant vascular and immunological features. Recently, biofabrication strategies, which combine biology, engineering, and manufacturing capabilities, have emerged as instrumental tools to recreate the heterogeneity of human skin precisely. Bioprinting uses computer-aided design (CAD) to yield robust and reproducible skin prototypes with unprecedented control over tissue design and assembly. As the interdisciplinary nature of biofabrication grows, we look to the promise of next-generation biofabrication technologies, such as organ-on-a-chip (OOAC) and 4D modeling, to simulate human tissue behaviors more reliably for research, pharmaceutical, and regenerative medicine purposes. This review aims to discuss the barriers to developing clinically relevant skin models, describe the evolution of skin-inspired in vitro structures, analyze the current approaches to biofabricating 3D human skin mimetics, and define the opportunities and challenges in biofabricating skin tissue for preclinical and clinical uses.
Collapse
Affiliation(s)
- Sarah C. Wistner
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Layla Rashad
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Gymama Slaughter
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
- Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, 23508, USA
| |
Collapse
|
27
|
Zielinska D, Fisch P, Moehrlen U, Finkielsztein S, Linder T, Zenobi-Wong M, Biedermann T, Klar AS. Combining bioengineered human skin with bioprinted cartilage for ear reconstruction. SCIENCE ADVANCES 2023; 9:eadh1890. [PMID: 37792948 PMCID: PMC10550230 DOI: 10.1126/sciadv.adh1890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/01/2023] [Indexed: 10/06/2023]
Abstract
Microtia is a congenital disorder that manifests as a malformation of the external ear leading to psychosocial problems in affected children. Here, we present a tissue-engineered treatment approach based on a bioprinted autologous auricular cartilage construct (EarCartilage) combined with a bioengineered human pigmented and prevascularized dermo-epidermal skin substitute (EarSkin) tested in immunocompromised rats. We confirmed that human-engineered blood capillaries of EarSkin connected to the recipient's vasculature within 1 week, enabling rapid blood perfusion and epidermal maturation. Bioengineered EarSkin displayed a stratified epidermis containing mature keratinocytes and melanocytes. The latter resided within the basal layer of the epidermis and efficiently restored the skin color. Further, in vivo tests demonstrated favorable mechanical stability of EarCartilage along with enhanced extracellular matrix deposition. In conclusion, EarCartilage combined with EarSkin represents a novel approach for the treatment of microtia with the potential to circumvent existing limitations and improve the aesthetic outcome of microtia reconstruction.
Collapse
Affiliation(s)
- Dominika Zielinska
- Tissue Biology Research Unit, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Philipp Fisch
- Tissue Engineering and Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Ueli Moehrlen
- Tissue Biology Research Unit, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | | | - Thomas Linder
- Klinik für Hals-, Nasen-, Ohren- und Gesichtschirurgie, Luzerner Kantonsspital, Luzern, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering and Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Agnes S. Klar
- Tissue Biology Research Unit, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| |
Collapse
|
28
|
Chen X, Laurent A, Liao Z, Jaccoud S, Abdel-Sayed P, Flahaut M, Scaletta C, Raffoul W, Applegate LA, Hirt-Burri N. Cutaneous Cell Therapy Manufacturing Timeframe Rationalization: Allogeneic Off-the-Freezer Fibroblasts for Dermo-Epidermal Combined Preparations (DE-FE002-SK2) in Burn Care. Pharmaceutics 2023; 15:2334. [PMID: 37765300 PMCID: PMC10536166 DOI: 10.3390/pharmaceutics15092334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/07/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Autologous cell therapy manufacturing timeframes constitute bottlenecks in clinical management pathways of severe burn patients. While effective temporary wound coverings exist for high-TBSA burns, any means to shorten the time-to-treatment with cytotherapeutic skin grafts could provide substantial therapeutic benefits. This study aimed to establish proofs-of-concept for a novel combinational cytotherapeutic construct (autologous/allogeneic DE-FE002-SK2 full dermo-epidermal graft) designed for significant cutaneous cell therapy manufacturing timeframe rationalization. Process development was based on several decades (four for autologous protocols, three for allogeneic protocols) of in-house clinical experience in cutaneous cytotherapies. Clinical grade dermal progenitor fibroblasts (standardized FE002-SK2 cell source) were used as off-the-freezer substrates in novel autologous/allogeneic dermo-epidermal bilayer sheets. Under vitamin C stimulation, FE002-SK2 primary progenitor fibroblasts rapidly produced robust allogeneic dermal templates, allowing patient keratinocyte attachment in co-culture. Notably, FE002-SK2 primary progenitor fibroblasts significantly outperformed patient fibroblasts for collagen deposition. An ex vivo de-epidermalized dermis model was used to demonstrate the efficient DE-FE002-SK2 construct bio-adhesion properties. Importantly, the presented DE-FE002-SK2 manufacturing process decreased clinical lot production timeframes from 6-8 weeks (standard autologous combined cytotherapies) to 2-3 weeks. Overall, these findings bear the potential to significantly optimize burn patient clinical pathways (for rapid wound closure and enhanced tissue healing quality) by combining extensively clinically proven cutaneous cell-based technologies.
Collapse
Affiliation(s)
- Xi Chen
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (X.C.); (A.L.); (Z.L.); (S.J.); (P.A.-S.); (M.F.); (C.S.); (W.R.)
| | - Alexis Laurent
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (X.C.); (A.L.); (Z.L.); (S.J.); (P.A.-S.); (M.F.); (C.S.); (W.R.)
- Manufacturing Department, TEC-PHARMA SA, CH-1038 Bercher, Switzerland
- Manufacturing Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland
| | - Zhifeng Liao
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (X.C.); (A.L.); (Z.L.); (S.J.); (P.A.-S.); (M.F.); (C.S.); (W.R.)
| | - Sandra Jaccoud
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (X.C.); (A.L.); (Z.L.); (S.J.); (P.A.-S.); (M.F.); (C.S.); (W.R.)
- Laboratory of Biomechanical Orthopedics, Federal Polytechnic School of Lausanne, CH-1015 Lausanne, Switzerland
| | - Philippe Abdel-Sayed
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (X.C.); (A.L.); (Z.L.); (S.J.); (P.A.-S.); (M.F.); (C.S.); (W.R.)
- STI School of Engineering, Federal Polytechnic School of Lausanne, CH-1015 Lausanne, Switzerland
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Marjorie Flahaut
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (X.C.); (A.L.); (Z.L.); (S.J.); (P.A.-S.); (M.F.); (C.S.); (W.R.)
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Corinne Scaletta
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (X.C.); (A.L.); (Z.L.); (S.J.); (P.A.-S.); (M.F.); (C.S.); (W.R.)
| | - Wassim Raffoul
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (X.C.); (A.L.); (Z.L.); (S.J.); (P.A.-S.); (M.F.); (C.S.); (W.R.)
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Lee Ann Applegate
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (X.C.); (A.L.); (Z.L.); (S.J.); (P.A.-S.); (M.F.); (C.S.); (W.R.)
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| | - Nathalie Hirt-Burri
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (X.C.); (A.L.); (Z.L.); (S.J.); (P.A.-S.); (M.F.); (C.S.); (W.R.)
| |
Collapse
|
29
|
Meuli JN, di Summa PG. Special Issue "Experimental and Clinical Advances in Skin Grafting". J Clin Med 2023; 12:jcm12103540. [PMID: 37240645 DOI: 10.3390/jcm12103540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Skin grafting is one of the oldest ways to treat soft-tissue defects [...].
Collapse
Affiliation(s)
- Joachim N Meuli
- Department of Plastic and Hand Surgery, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Pietro G di Summa
- Department of Plastic and Hand Surgery, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| |
Collapse
|
30
|
Hu Y, Zhang H, Zou Q, Liu W, Li W, Yan L, Dai H. The effect of silicon groups on the physicochemical property and bioactivity of L-phenylalanine derived poly(amide-imide). J Biomed Mater Res B Appl Biomater 2023. [PMID: 37081804 DOI: 10.1002/jbm.b.35257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/17/2023] [Accepted: 04/01/2023] [Indexed: 04/22/2023]
Abstract
Poly(amide-imide) (PAI), serving as a synthetic polymer, has been widely used in industry for excellent mechanical properties, chemical resistance and high thermal stability. However, lack of suitable cell niche and biological activity limited the further application of PAI in biomedical engineering. Herein, silicon modified L-phenylalanine derived poly(amide-imide) (PAIS) was synthesized by introducing silica to L-phenylalanine derived PAI to improve physicochemical and biological performances. The influence of silicon amount on physicochemical, immune, and angiogenic performances of PAIS were systemically studied. The results show that PAIS exerts excellent hydrophilic, mechanical, biological activity. PAIS shows no effects on the number of macrophages, but can regulate macrophage polarization and angiogenesis in a dose-dependent manner. This study advanced our understanding of silicon modification in PAI can modulate cell responses via initiating silicon concentration regulation. The acquired knowledge will provide a new strategy to design and optimize biomedical PAI in the future.
Collapse
Affiliation(s)
- Yaping Hu
- Shenzhen Research Institute of Wuhan University of Technology, Shenzhen, China
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
| | - Hongbiao Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
| | - Qiying Zou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
| | - Wenbin Liu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenqin Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
| | - Lesan Yan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
| | - Honglian Dai
- Shenzhen Research Institute of Wuhan University of Technology, Shenzhen, China
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
| |
Collapse
|
31
|
Rütsche D, Nanni M, Rüdisser S, Biedermann T, Zenobi-Wong M. Enzymatically Crosslinked Collagen as a Versatile Matrix for In Vitro and In Vivo Co-Engineering of Blood and Lymphatic Vasculature. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209476. [PMID: 36724374 DOI: 10.1002/adma.202209476] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/30/2022] [Indexed: 06/18/2023]
Abstract
Adequate vascularization is required for the successful translation of many in vitro engineered tissues. This study presents a novel collagen derivative that harbors multiple recognition peptides for orthogonal enzymatic crosslinking based on sortase A (SrtA) and Factor XIII (FXIII). SrtA-mediated crosslinking enables the rapid co-engineering of human blood and lymphatic microcapillaries and mesoscale capillaries in bulk hydrogels. Whereas tuning of gel stiffness determines the extent of neovascularization, the relative number of blood and lymphatic capillaries recapitulates the ratio of blood and lymphatic endothelial cells originally seeded into the hydrogel. Bioengineered capillaries readily form luminal structures and exhibit typical maturation markers both in vitro and in vivo. The secondary crosslinking enzyme Factor XIII is used for in situ tethering of the VEGF mimetic QK peptide to collagen. This approach supports the formation of blood and lymphatic capillaries in the absence of exogenous VEGF. Orthogonal enzymatic crosslinking is further used to bioengineer hydrogels with spatially defined polymer compositions with pro- and anti-angiogenic properties. Finally, macroporous scaffolds based on secondary crosslinking of microgels enable vascularization independent from supporting fibroblasts. Overall, this work demonstrates for the first time the co-engineering of mature micro- and meso-sized blood and lymphatic capillaries using a highly versatile collagen derivative.
Collapse
Affiliation(s)
- Dominic Rütsche
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zurich, Otto-Stern-Weg 7, Zurich, 8093, Switzerland
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, Schlieren, 8952, Switzerland
| | - Monica Nanni
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, Schlieren, 8952, Switzerland
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Leonhardstrasse 21, Zurich, 8092, Switzerland
| | - Simon Rüdisser
- Biomolecular NMR Spectroscopy Platform, Department of Biology, ETH Zurich, Hönggerbergring 64, Zurich, 8093, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, Schlieren, 8952, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zurich, Otto-Stern-Weg 7, Zurich, 8093, Switzerland
| |
Collapse
|
32
|
Deng H, Zhang J, Wu F, Wei F, Han W, Xu X, Zhang Y. Current Status of Lymphangiogenesis: Molecular Mechanism, Immune Tolerance, and Application Prospect. Cancers (Basel) 2023; 15:cancers15041169. [PMID: 36831512 PMCID: PMC9954532 DOI: 10.3390/cancers15041169] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
The lymphatic system is a channel for fluid transport and cell migration, but it has always been controversial in promoting and suppressing cancer. VEGFC/VEGFR3 signaling has long been recognized as a major molecular driver of lymphangiogenesis. However, many studies have shown that the neural network of lymphatic signaling is complex. Lymphatic vessels have been found to play an essential role in the immune regulation of tumor metastasis and cardiac repair. This review describes the effects of lipid metabolism, extracellular vesicles, and flow shear forces on lymphangiogenesis. Moreover, the pro-tumor immune tolerance function of lymphatic vessels is discussed, and the tasks of meningeal lymphatic vessels and cardiac lymphatic vessels in diseases are further discussed. Finally, the value of conversion therapy targeting the lymphatic system is introduced from the perspective of immunotherapy and pro-lymphatic biomaterials for lymphangiogenesis.
Collapse
Affiliation(s)
- Hongyang Deng
- Hepatic-Biliary-Pancreatic Institute, Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Jiaxing Zhang
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Fahong Wu
- Hepatic-Biliary-Pancreatic Institute, Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Fengxian Wei
- Hepatic-Biliary-Pancreatic Institute, Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Wei Han
- Hepatic-Biliary-Pancreatic Institute, Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Xiaodong Xu
- Hepatic-Biliary-Pancreatic Institute, Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Youcheng Zhang
- Hepatic-Biliary-Pancreatic Institute, Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730030, China
- Correspondence:
| |
Collapse
|
33
|
Nanni M, Rütsche D, Bächler C, Pontiggia L, Klar AS, Moehrlen U, Biedermann T. CD146 expression profile in human skin and pre-vascularized dermo-epidermal skin substitutes in vivo. J Biol Eng 2023; 17:9. [PMID: 36721239 PMCID: PMC9890844 DOI: 10.1186/s13036-023-00327-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/20/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND CD146 is a cell adhesion molecule whose expression profile in human skin has not yet been elucidated. Here, we characterize CD146 expression pattern in human skin, in particular in blood endothelial cells (BECs) and lymphatic endothelial cells (LECs), which constitute human dermal microvascular endothelial cells (HDMECs), as well as in perivascular cells. RESULTS We demonstrated that CD146 is a specific marker of BECs, but not of LECs. Moreover, we found CD146 expression also in human pericytes surrounding blood capillaries in human skin. In addition, we demonstrated that CD146 expression is up-regulated by the TNFα-IL-1β/NF-kB axis in both BECs and pericytes. Finally, we engineered 3D collagen hydrogels composed of HDMECs, CD146+ pericytes, and fibroblasts which developed, in vitro and in vivo, a complete microvasculature network composed of blood and lymphatic capillaries with pericytes investing blood capillaries. CONCLUSIONS Overall, our results proved that CD146 is a specific marker of BECs and pericytes, but not LECs in human skin. Further, the combination of CD146+ pericytes with HDMECs in skin substitutes allowed to bioengineer a comprehensive 3D in vitro and in vivo model of the human dermal microvasculature.
Collapse
Affiliation(s)
- Monica Nanni
- grid.412341.10000 0001 0726 4330Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, Wagistrasse 12, 8952 Zurich, Switzerland ,grid.412341.10000 0001 0726 4330Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland ,grid.5801.c0000 0001 2156 2780Department of Mechanical and Process Engineering, Institute for Mechanical Systems, ETH Zurich, Leonhardstrasse 21, 8092 Zurich, Switzerland
| | - Dominic Rütsche
- grid.412341.10000 0001 0726 4330Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, Wagistrasse 12, 8952 Zurich, Switzerland ,grid.412341.10000 0001 0726 4330Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland ,grid.5801.c0000 0001 2156 2780Department of Mechanical and Process Engineering, Institute for Mechanical Systems, ETH Zurich, Leonhardstrasse 21, 8092 Zurich, Switzerland
| | - Curdin Bächler
- grid.412341.10000 0001 0726 4330Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, Wagistrasse 12, 8952 Zurich, Switzerland ,grid.412341.10000 0001 0726 4330Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Luca Pontiggia
- grid.412341.10000 0001 0726 4330Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, Wagistrasse 12, 8952 Zurich, Switzerland ,grid.412341.10000 0001 0726 4330Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Agnes S. Klar
- grid.412341.10000 0001 0726 4330Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, Wagistrasse 12, 8952 Zurich, Switzerland ,grid.412341.10000 0001 0726 4330Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Ueli Moehrlen
- grid.412341.10000 0001 0726 4330Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, Wagistrasse 12, 8952 Zurich, Switzerland ,grid.412341.10000 0001 0726 4330Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland ,grid.412341.10000 0001 0726 4330Department of Surgery, University Children’s Hospital Zurich, Zurich, Switzerland ,grid.7400.30000 0004 1937 0650University of Zurich, Zurich, Switzerland
| | - Thomas Biedermann
- grid.412341.10000 0001 0726 4330Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, Wagistrasse 12, 8952 Zurich, Switzerland ,grid.412341.10000 0001 0726 4330Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland ,grid.7400.30000 0004 1937 0650University of Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
Kong AM, Lim SY, Palmer JA, Rixon A, Gerrand YW, Yap KK, Morrison WA, Mitchell GM. Engineering transplantable human lymphatic and blood capillary networks in a porous scaffold. J Tissue Eng 2022; 13:20417314221140979. [PMID: 36600999 PMCID: PMC9806376 DOI: 10.1177/20417314221140979] [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: 08/16/2022] [Accepted: 11/08/2022] [Indexed: 12/27/2022] Open
Abstract
Due to a relative paucity of studies on human lymphatic assembly in vitro and subsequent in vivo transplantation, capillary formation and survival of primary human lymphatic (hLEC) and blood endothelial cells (hBEC) ± primary human vascular smooth muscle cells (hvSMC) were evaluated and compared in vitro and in vivo. hLEC ± hvSMC or hBEC ± hvSMC were seeded in a 3D porous scaffold in vitro, and capillary percent vascular volume (PVV) and vascular density (VD)/mm2 assessed. Scaffolds were also transplanted into a sub-cutaneous rat wound with morphology/morphometry assessment. Initially hBEC formed a larger vessel network in vitro than hLEC, with interconnected capillaries evident at 2 days. Interconnected lymphatic capillaries were slower (3 days) to assemble. hLEC capillaries demonstrated a significant overall increase in PVV (p = 0.0083) and VD (p = 0.0039) in vitro when co-cultured with hvSMC. A similar increase did not occur for hBEC + hvSMC in vitro, but hBEC + hvSMC in vivo significantly increased PVV (p = 0.0035) and VD (p = 0.0087). Morphology/morphometry established that hLEC vessels maintained distinct cell markers, and demonstrated significantly increased individual vessel and network size, and longer survival than hBEC capillaries in vivo, and established inosculation with rat lymphatics, with evidence of lymphatic function. The porous polyurethane scaffold provided advantages to capillary network formation due to its large (300-600 μm diameter) interconnected pores, and sufficient stability to ensure successful surgical transplantation in vivo. Given their successful survival and function in vivo within the porous scaffold, in vitro assembled hLEC networks using this method are potentially applicable to clinical scenarios requiring replacement of dysfunctional or absent lymphatic networks.
Collapse
Affiliation(s)
- Anne M Kong
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
| | - Shiang Y Lim
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Department of Surgery at St Vincent’s
Hospital Melbourne, University of Melbourne, Fitzroy, VIC, Australia
- Drug Discovery Biology, Faculty of
Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, VIC,
Australia
- National Heart Research Institute
Singapore, National Heart Centre Singapore
| | - Jason A Palmer
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Centre for Eye Research Australia, East
Melbourne, VIC, Australia
| | - Amanda Rixon
- Experimental Medical and Surgical Unit,
St Vincent’s Hospital Melbourne, Fitzroy, VIC, Australia
| | - Yi-Wen Gerrand
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
| | - Kiryu K Yap
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Department of Surgery at St Vincent’s
Hospital Melbourne, University of Melbourne, Fitzroy, VIC, Australia
| | - Wayne A Morrison
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Department of Surgery at St Vincent’s
Hospital Melbourne, University of Melbourne, Fitzroy, VIC, Australia
- Faculty of Health Sciences, Australian
Catholic University, East Melbourne VIC, Australia
- Department of Plastic and
Reconstructive Surgery, St Vincent’s Hospital Melbourne, Fitzroy, VIC,
Australia
| | - Geraldine M Mitchell
- O’Brien Institute Department of St
Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Department of Surgery at St Vincent’s
Hospital Melbourne, University of Melbourne, Fitzroy, VIC, Australia
- Faculty of Health Sciences, Australian
Catholic University, East Melbourne VIC, Australia
- Geraldine M Mitchell, O’Brien Institute
Department at St Vincent’s Institute of Medical Research, 9 Princes Street,
Fitzroy, VIC 3065, Australia.
| |
Collapse
|
35
|
Nagahara AI, Homma J, Ryu B, Sekine H, Higashi Y, Shimizu T, Kawamata T. Networked lymphatic endothelial cells in a transplanted cell sheet contribute to form functional lymphatic vessels. Sci Rep 2022; 12:21698. [PMID: 36522421 PMCID: PMC9755306 DOI: 10.1038/s41598-022-26041-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
This study evaluated whether cell sheets containing a network of lymphatic endothelial cells (LECs) promoted lymphangiogenesis after transplantation in vivo. Cell sheets with a LEC network were constructed by co-culturing LECs and adipose-derived stem cells (ASCs) on temperature-responsive culture dishes. A cell ratio of 3:2 (vs. 1:4) generated networks with more branches and longer branch lengths. LEC-derived lymphatic vessels were observed 2 weeks after transplantation of a three-layered cell sheet construct onto rat gluteal muscle. Lymphatic vessel number, diameter and depth were greatest for a construct comprising two ASC sheets stacked on a LEC/ASC (3:2 ratio) sheet. Transplantation of this construct in a rat model of femoral lymphangiectomy led to the formation of functional lymphatic vessels containing both transplanted and host LECs. Further development of this technique may lead to a new method of promoting lymphangiogenesis.
Collapse
Affiliation(s)
- Ayumi Inoue Nagahara
- grid.410818.40000 0001 0720 6587Department of Neurosurgery, Graduate School of Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Jun Homma
- grid.410818.40000 0001 0720 6587Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Bikei Ryu
- grid.488555.10000 0004 1771 2637Department of Neurosurgery, Tokyo Women’s Medical University Hospital, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Hidekazu Sekine
- grid.410818.40000 0001 0720 6587Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Yuhei Higashi
- grid.410818.40000 0001 0720 6587Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan ,Tokaihit Co., Ltd., Shizuoka, Japan
| | - Tatsuya Shimizu
- grid.410818.40000 0001 0720 6587Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Takakazu Kawamata
- grid.488555.10000 0004 1771 2637Department of Neurosurgery, Tokyo Women’s Medical University Hospital, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| |
Collapse
|
36
|
Sanchez MM, Tonmoy TI, Park BH, Morgan JT. Development of a Vascularized Human Skin Equivalent with Hypodermis for Photoaging Studies. Biomolecules 2022; 12:biom12121828. [PMID: 36551256 PMCID: PMC9775308 DOI: 10.3390/biom12121828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Photoaging is an important extrinsic aging factor leading to altered skin morphology and reduced function. Prior work has revealed a connection between photoaging and loss of subcutaneous fat. Currently, primary models for studying this are in vivo (human samples or animal models) or in vitro models, including human skin equivalents (HSEs). In vivo models are limited by accessibility and cost, while HSEs typically do not include a subcutaneous adipose component. To address this, we developed an "adipose-vascular" HSE (AVHSE) culture method, which includes both hypodermal adipose and vascular cells. Furthermore, we tested AVHSE as a potential model for hypodermal adipose aging via exposure to 0.45 ± 0.15 mW/cm2 385 nm light (UVA). One week of 2 h daily UVA exposure had limited impact on epidermal and vascular components of the AVHSE, but significantly reduced adiposity by approximately 50%. Overall, we have developed a novel method for generating HSE that include vascular and adipose components and demonstrated potential as an aging model using photoaging as an example.
Collapse
|
37
|
Jiang L, Yang X, Zhang Y, He D, Gao Y, Lu K, Hao Y, Gao Y, Lu D, Jin X, Li C. Novel ROS-scavenging hydrogel with enhanced anti-inflammation and angiogenic properties for promoting diabetic wound healing. BIOMATERIALS ADVANCES 2022; 144:213226. [PMID: 36481518 DOI: 10.1016/j.bioadv.2022.213226] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/08/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022]
Abstract
Accelerating angiogenesis of diabetic wounds is crucial to promoting wound healing. Currently, vascular endothelial growth factor (VEGF), an angiogenesis-related bioactive molecule, is widely used in clinic to enhance wound angiogenesis, but it faces problems of inactivation and low utilization due to harsh microenvironment. Here, we developed a novel reactive oxygen species (ROS)-scavenging hydrogel aimed to polarize macrophages toward an anti-inflammatory phenotype, inducing efficient angiogenesis in diabetic wounds. This composite hydrogel with good biosafety and mechanical properties showed sustainable release of bioactive VEGF. Importantly, it could significantly reduce ROS level and rapidly improve wound microenvironment, which ensured the activity of VEGF in vitro and in vivo and successful healing eventually. At the same time, the composite hydrogel exhibited excellent antibacterial properties. In vivo results confirmed good anti-inflammatory, stimulated vascularization and accelerated wound healing attributed to the novel ROS-scavenging hydrogel, which might serve as a promising wound dressing in diabetic wound healing.
Collapse
Affiliation(s)
- Lianghua Jiang
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, People's Republic of China
| | - Xufeng Yang
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, People's Republic of China
| | - Yijian Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, People's Republic of China
| | - Dawei He
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, People's Republic of China
| | - Yijun Gao
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, People's Republic of China
| | - Ke Lu
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, People's Republic of China
| | - Yanming Hao
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, People's Republic of China
| | - Yan Gao
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, People's Republic of China
| | - Daming Lu
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, People's Republic of China.
| | - Xiangyun Jin
- Department of Orthopaedics, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, People's Republic of China.
| | - Chong Li
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, People's Republic of China.
| |
Collapse
|
38
|
Sanchez MM, Bagdasarian IA, Darch W, Morgan JT. Organotypic cultures as aging associated disease models. Aging (Albany NY) 2022; 14:9338-9383. [PMID: 36435511 PMCID: PMC9740367 DOI: 10.18632/aging.204361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/21/2022] [Indexed: 11/24/2022]
Abstract
Aging remains a primary risk factor for a host of diseases, including leading causes of death. Aging and associated diseases are inherently multifactorial, with numerous contributing factors and phenotypes at the molecular, cellular, tissue, and organismal scales. Despite the complexity of aging phenomena, models currently used in aging research possess limitations. Frequently used in vivo models often have important physiological differences, age at different rates, or are genetically engineered to match late disease phenotypes rather than early causes. Conversely, routinely used in vitro models lack the complex tissue-scale and systemic cues that are disrupted in aging. To fill in gaps between in vivo and traditional in vitro models, researchers have increasingly been turning to organotypic models, which provide increased physiological relevance with the accessibility and control of in vitro context. While powerful tools, the development of these models is a field of its own, and many aging researchers may be unaware of recent progress in organotypic models, or hesitant to include these models in their own work. In this review, we describe recent progress in tissue engineering applied to organotypic models, highlighting examples explicitly linked to aging and associated disease, as well as examples of models that are relevant to aging. We specifically highlight progress made in skin, gut, and skeletal muscle, and describe how recently demonstrated models have been used for aging studies or similar phenotypes. Throughout, this review emphasizes the accessibility of these models and aims to provide a resource for researchers seeking to leverage these powerful tools.
Collapse
Affiliation(s)
- Martina M. Sanchez
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | | | - William Darch
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | - Joshua T. Morgan
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| |
Collapse
|
39
|
Freitas-Ribeiro S, Diogo GS, Oliveira C, Martins A, Silva TH, Jarnalo M, Horta R, Reis RL, Pirraco RP. Growth Factor-Free Vascularization of Marine-Origin Collagen Sponges Using Cryopreserved Stromal Vascular Fractions from Human Adipose Tissue. Mar Drugs 2022; 20:md20100623. [PMID: 36286447 PMCID: PMC9604698 DOI: 10.3390/md20100623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
The successful integration of transplanted three-dimensional tissue engineering (TE) constructs depends greatly on their rapid vascularization. Therefore, it is essential to address this vascularization issue in the initial design of constructs for perfused tissues. Two of the most important variables in this regard are scaffold composition and cell sourcing. Collagens with marine origins overcome some issues associated with mammal-derived collagen while maintaining their advantages in terms of biocompatibility. Concurrently, the freshly isolated stromal vascular fraction (SVF) of adipose tissue has been proposed as an advantageous cell fraction for vascularization purposes due to its highly angiogenic properties, allowing extrinsic angiogenic growth factor-free vascularization strategies for TE applications. In this study, we aimed at understanding whether marine collagen 3D matrices could support cryopreserved human SVF in maintaining intrinsic angiogenic properties observed for fresh SVF. For this, cryopreserved human SVF was seeded on blue shark collagen sponges and cultured up to 7 days in a basal medium. The secretome profile of several angiogenesis-related factors was studied throughout culture times and correlated with the expression pattern of CD31 and CD146, which showed the formation of a prevascular network. Upon in ovo implantation, increased vessel recruitment was observed in prevascularized sponges when compared with sponges without SVF cells. Immunohistochemistry for CD31 demonstrated the improved integration of prevascularized sponges within chick chorioalantoic membrane (CAM) tissues, while in situ hybridization showed human cells lining blood vessels. These results demonstrate the potential of using cryopreserved SVF combined with marine collagen as a streamlined approach to improve the vascularization of TE constructs.
Collapse
Affiliation(s)
- Sara Freitas-Ribeiro
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Gabriela S. Diogo
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Catarina Oliveira
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Albino Martins
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Tiago H. Silva
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Mariana Jarnalo
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, 4200-319 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Ricardo Horta
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, 4200-319 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Rui L. Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Rogério P. Pirraco
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
- Correspondence:
| |
Collapse
|
40
|
Sun S, Jin L, Zheng Y, Zhu J. Modeling human HSV infection via a vascularized immune-competent skin-on-chip platform. Nat Commun 2022; 13:5481. [PMID: 36123328 PMCID: PMC9485166 DOI: 10.1038/s41467-022-33114-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 09/02/2022] [Indexed: 02/05/2023] Open
Abstract
Herpes simplex virus (HSV) naturally infects skin and mucosal surfaces, causing lifelong recurrent disease worldwide, with no cure or vaccine. Biomimetic human tissue and organ platforms provide attractive alternatives over animal models to recapitulate human diseases. Combining prevascularization and microfluidic approaches, we present a vascularized, three-dimensional skin-on-chip that mimics human skin architecture and is competent to immune-cell and drug perfusion. The endothelialized microvasculature embedded in a fibroblast-containing dermis responds to biological stimulation, while the cornified epidermis functions as a protective barrier. HSV infection of the skin-on-chip displays tissue-level key morphological and pathophysiological features typical of genital herpes infection in humans, including the production of proinflammatory cytokine IL-8, which triggers rapid neutrophil trans-endothelial extravasation and directional migration. Importantly, perfusion with the antiviral drug acyclovir inhibits HSV infection in a dose-dependent and time-sensitive manner. Thus, our vascularized skin-on-chip represents a promising platform for human HSV disease modeling and preclinical therapeutic evaluation.
Collapse
Affiliation(s)
- Sijie Sun
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
| | - Lei Jin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA
| | - Ying Zheng
- Department of Bioengineering, University of Washington, Seattle, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA
| | - Jia Zhu
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, USA.
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, USA.
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA.
| |
Collapse
|
41
|
Roth‐Carter QR, Koetsier JL, Broussard JA, Green KJ. Organotypic Human Skin Cultures Incorporating Primary Melanocytes. Curr Protoc 2022; 2:e536. [PMID: 36165649 PMCID: PMC9796167 DOI: 10.1002/cpz1.536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Three-dimensional (3D) human organotypic skin cultures provide a physiologically relevant model that recapitulates in vivo skin features. Most commonly, organotypic skin cultures are created by seeding isolated epidermal keratinocytes onto a collagen/fibroblast plug and lifting to an air liquid interface. These conditions are sufficient to drive stratification and differentiation of the keratinocytes to form an epidermal-like sheet with remarkable similarities to human epidermis. Coupled with genetic or pharmacological treatments, these cultures provide a powerful tool for elucidating keratinocyte biology. Recent focus has been placed on increasing the utility of organotypic skin cultures by incorporating other cell types that are present in the skin, such as melanocytes, immune cells, and other cells. Here we describe a step-by-step protocol for the isolation of neonatal human epidermal keratinocytes and melanocytes from foreskins, and the creation of organotypic skin cultures that include both cell types. We also describe methods that can be used to assess melanocyte behavior in these organotypic cultures, including methods for whole mount staining, measurement of melanocyte dendricity, staining for pigment, and 5-bromo-2'-deoxyuridine (BrdU) labeling for identification of proliferating cells. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Isolation of primary cells Alternate Protocol: Isolation of primary cells using differential trypsinization Basic Protocol 2: Organotypic culture protocol Support Protocol 1: Culture and maintenance of NHEKs and melanocytes Support Protocol 2: Lentiviral transduction of melanocytes Support Protocol 3: Retroviral transduction of NHEKs Support Protocol 4: Whole mount immunostaining protocol Support Protocol 5: Measuring melanocyte dendricity Support Protocol 6: Fontana-Masson staining protocol Support Protocol 7: BrdU labeling and staining.
Collapse
Affiliation(s)
- Quinn R. Roth‐Carter
- Department of Pathology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinois
| | - Jennifer L. Koetsier
- Department of Pathology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinois
| | - Joshua A. Broussard
- Department of Pathology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinois
- Department of Dermatology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinois
- Robert H. Lurie Comprehensive Cancer CenterNorthwestern UniversityChicagoIllinois
| | - Kathleen J. Green
- Department of Pathology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinois
- Department of Dermatology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinois
- Robert H. Lurie Comprehensive Cancer CenterNorthwestern UniversityChicagoIllinois
| |
Collapse
|
42
|
Jeong DP, Hall E, Neu E, Hanjaya-Putra D. Podoplanin is Responsible for the Distinct Blood and Lymphatic Capillaries. Cell Mol Bioeng 2022; 15:467-478. [PMID: 36444348 PMCID: PMC9700554 DOI: 10.1007/s12195-022-00730-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022] Open
Abstract
Abstract
Introduction
Controlling the formation of blood and lymphatic vasculatures is crucial for engineered tissues. Although the lymphatic vessels originate from embryonic blood vessels, the two retain functional and physiological differences even as they develop in the vicinity of each other. This suggests that there is a previously unknown molecular mechanism by which blood (BECs) and lymphatic endothelial cells (LECs) recognize each other and coordinate to generate distinct capillary networks.
Methods
We utilized Matrigel and fibrin assays to determine how cord-like structures (CLS) can be controlled by altering LEC and BEC identity through podoplanin (PDPN) and folliculin (FLCN) expressions. We generated BECΔFLCN and LECΔPDPN, and observed cell migration to characterize loss lymphatic and blood characteristics due to respective knockouts.
Results
We observed that LECs and BECs form distinct CLS in Matrigel and fibrin gels despite being cultured in close proximity with each other. We confirmed that the LECs and BECs do not recognize each other through paracrine signaling, as proliferation and migration of both cells were unaffected by paracrine signals. On the other hand, we found PDPN to be the key surface protein that is responsible for LEC-BEC recognition, and LECs lacking PDPN became pseudo-BECs and vice versa. We also found that FLCN maintains BEC identity through downregulation of PDPN.
Conclusions
Overall, these observations reveal a new molecular pathway through which LECs and BECs form distinct CLS through physical contact by PDPN which in turn is regulated by FLCN, which has important implications toward designing functional engineered tissues.
Collapse
|
43
|
Vahav I, Thon M, van den Broek LJ, Spiekstra SW, Atac B, Lindner G, Schimek K, Marx U, Gibbs S. Proof-of-Concept Organ-on-Chip Study: Topical Cinnamaldehyde Exposure of Reconstructed Human Skin with Integrated Neopapillae Cultured under Dynamic Flow. Pharmaceutics 2022; 14:pharmaceutics14081529. [PMID: 35893784 PMCID: PMC9330995 DOI: 10.3390/pharmaceutics14081529] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 02/01/2023] Open
Abstract
Pharmaceutical and personal care industries require human representative models for testing to ensure the safety of their products. A major route of penetration into our body after substance exposure is via the skin. Our aim was to generate robust culture conditions for a next generation human skin-on-chip model containing neopapillae and to establish proof-of-concept testing with the sensitizer, cinnamaldehyde. Reconstructed human skin consisting of a stratified and differentiated epidermis on a fibroblast populated hydrogel containing neopapillae spheroids (RhS-NP), were cultured air-exposed and under dynamic flow for 10 days. The robustness of three independent experiments, each with up to 21 intra-experiment replicates, was investigated. The epidermis was seen to invaginate into the hydrogel towards the neopapille spheroids. Daily measurements of lactate dehydrogenase (LDH) and glucose levels within the culture medium demonstrated high viability and stable metabolic activity throughout the culture period in all three independent experiments and in the replicates within an experiment. Topical cinnamaldehyde exposure to RhS-NP resulted in dose-dependent cytotoxicity (increased LDH release) and elevated cytokine secretion of contact sensitizer specific IL-18, pro-inflammatory IL-1β, inflammatory IL-23 and IFN-γ, as well as anti-inflammatory IL-10 and IL-12p70. This study demonstrates the robustness and feasibility of complex next generation skin models for investigating skin immunotoxicity.
Collapse
Affiliation(s)
- Irit Vahav
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- TissUse GmbH, Oudenarder Str. 16, 13347 Berlin, Germany
- Amsterdam Movement Sciences, Tissue Function & Regeneration, 1081 HV Amsterdam, The Netherlands
| | - Maria Thon
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, 1081 HV Amsterdam, The Netherlands
| | - Lenie J. van den Broek
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Sander W. Spiekstra
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, 1081 HV Amsterdam, The Netherlands
| | - Beren Atac
- TissUse GmbH, Oudenarder Str. 16, 13347 Berlin, Germany
- Department of Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Gerd Lindner
- TissUse GmbH, Oudenarder Str. 16, 13347 Berlin, Germany
- Provio GmbH, Oranienburger Chaussee 2, 16548 Glienicke/Nordbahn, Germany
| | | | - Uwe Marx
- TissUse GmbH, Oudenarder Str. 16, 13347 Berlin, Germany
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, 1081 HV Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands
- Correspondence:
| |
Collapse
|
44
|
Shin GJE, Abaci HE, Smith MC. Cellular Pathogenesis of Chemotherapy-Induced Peripheral Neuropathy: Insights From Drosophila and Human-Engineered Skin Models. FRONTIERS IN PAIN RESEARCH 2022; 3:912977. [PMID: 35875478 PMCID: PMC9304629 DOI: 10.3389/fpain.2022.912977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a highly prevalent and complex condition arising from chemotherapy cancer treatments. Currently, there are no treatment or prevention options in the clinic. CIPN accompanies pain-related sensory functions starting from the hands and feet. Studies focusing on neurons in vitro and in vivo models significantly advanced our understanding of CIPN pathological mechanisms. However, given the direct toxicity shown in both neurons and non-neuronal cells, effective in vivo or in vitro models that allow the investigation of neurons in their local environment are required. No single model can provide a complete solution for the required investigation, therefore, utilizing a multi-model approach would allow complementary advantages of different models and robustly validate findings before further translation. This review aims first to summarize approaches and insights from CIPN in vivo models utilizing small model organisms. We will focus on Drosophila melanogaster CIPN models that are genetically amenable and accessible to study neuronal interactions with the local environment in vivo. Second, we will discuss how these findings could be tested in physiologically relevant vertebrate models. We will focus on in vitro approaches using human cells and summarize the current understanding of engineering approaches that may allow the investigation of pathological changes in neurons and the skin environment.
Collapse
Affiliation(s)
- Grace Ji-eun Shin
- Zuckerman Mind Brain and Behavior Institute, Jerome L. Greene Science Center, Columbia University, New York, NY, United States
- *Correspondence: Grace Ji-eun Shin
| | - Hasan Erbil Abaci
- Department of Dermatology, Columbia University Medical Center, Saint Nicholas Avenue, New York, NY, United States
| | - Madison Christine Smith
- Zuckerman Mind Brain and Behavior Institute, Jerome L. Greene Science Center, Columbia University, New York, NY, United States
| |
Collapse
|
45
|
Abstract
The lymphatic system, composed of initial and collecting lymphatic vessels as well as lymph nodes that are present in almost every tissue of the human body, acts as an essential transport system for fluids, biomolecules and cells between peripheral tissues and the central circulation. Consequently, it is required for normal body physiology but is also involved in the pathogenesis of various diseases, most notably cancer. The important role of tumor-associated lymphatic vessels and lymphangiogenesis in the formation of lymph node metastasis has been elucidated during the last two decades, whereas the underlying mechanisms and the relation between lymphatic and peripheral organ dissemination of cancer cells are incompletely understood. Lymphatic vessels are also important for tumor-host communication, relaying molecular information from a primary or metastatic tumor to regional lymph nodes and the circulatory system. Beyond antigen transport, lymphatic endothelial cells, particularly those residing in lymph node sinuses, have recently been recognized as direct regulators of tumor immunity and immunotherapy responsiveness, presenting tumor antigens and expressing several immune-modulatory signals including PD-L1. In this review, we summarize recent discoveries in this rapidly evolving field and highlight strategies and challenges of therapeutic targeting of lymphatic vessels or specific lymphatic functions in cancer patients.
Collapse
Affiliation(s)
- Lothar C Dieterich
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Carlotta Tacconi
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.,Department of Biosciences, University of Milan, Milan, Italy
| | - Luca Ducoli
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| |
Collapse
|
46
|
Kaur G, Narayanan G, Garg D, Sachdev A, Matai I. Biomaterials-Based Regenerative Strategies for Skin Tissue Wound Healing. ACS APPLIED BIO MATERIALS 2022; 5:2069-2106. [PMID: 35451829 DOI: 10.1021/acsabm.2c00035] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Skin tissue wound healing proceeds through four major stages, including hematoma formation, inflammation, and neo-tissue formation, and culminates with tissue remodeling. These four steps significantly overlap with each other and are aided by various factors such as cells, cytokines (both anti- and pro-inflammatory), and growth factors that aid in the neo-tissue formation. In all these stages, advanced biomaterials provide several functional advantages, such as removing wound exudates, providing cover, transporting oxygen to the wound site, and preventing infection from microbes. In addition, advanced biomaterials serve as vehicles to carry proteins/drug molecules/growth factors and/or antimicrobial agents to the target wound site. In this review, we report recent advancements in biomaterials-based regenerative strategies that augment the skin tissue wound healing process. In conjunction with other medical sciences, designing nanoengineered biomaterials is gaining significant attention for providing numerous functionalities to trigger wound repair. In this regard, we highlight the advent of nanomaterial-based constructs for wound healing, especially those that are being evaluated in clinical settings. Herein, we also emphasize the competence and versatility of the three-dimensional (3D) bioprinting technique for advanced wound management. Finally, we discuss the challenges and clinical perspective of various biomaterial-based wound dressings, along with prospective future directions. With regenerative strategies that utilize a cocktail of cell sources, antimicrobial agents, drugs, and/or growth factors, it is expected that significant patient-specific strategies will be developed in the near future, resulting in complete wound healing with no scar tissue formation.
Collapse
Affiliation(s)
- Gurvinder Kaur
- Materials Science and Sensor Applications, Central Scientific Instruments Organization, Chandigarh 160030, India
| | - Ganesh Narayanan
- Fiber and Polymer Science Program, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Deepa Garg
- Materials Science and Sensor Applications, Central Scientific Instruments Organization, Chandigarh 160030, India
| | - Abhay Sachdev
- Materials Science and Sensor Applications, Central Scientific Instruments Organization, Chandigarh 160030, India
| | - Ishita Matai
- Department of Biotechnology, School of Biological Sciences, Amity University Punjab, Mohali 140306, India
| |
Collapse
|
47
|
Hooks JST, Bernard FC, Cruz-Acuña R, Nepiyushchikh Z, Gonzalez-Vargas Y, García AJ, Dixon JB. Synthetic hydrogels engineered to promote collecting lymphatic vessel sprouting. Biomaterials 2022; 284:121483. [PMID: 35428014 PMCID: PMC9134840 DOI: 10.1016/j.biomaterials.2022.121483] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 12/16/2022]
Abstract
The lymphatic vasculature is an essential component of the body's circulation providing a network of vessels to return fluid and proteins from the tissue space to the blood, to facilitate immune ce-ll and antigen transport to lymph nodes, and to take up dietary lipid from the intestine. The development of biomaterial-based strategies to facilitate the growth of lymphatics either for regenerative purposes or as model system to study lymphatic biology is still in its nascent stages. In particular, platforms that encourage the sprouting and formation of lymphatic networks from collecting vessels are particularly underdeveloped. Through implementation of a modular, poly(ethylene glycol) (PEG)-based hydrogel, we explored the independent contributions of matrix elasticity, degradability, and adhesive peptide presentation on sprouting of implanted segments of rat lymphatic collecting vessels. An engineered hydrogel with 680 Pa elasticity, 2.0 mM RGD adhesive peptide, and full susceptibility to protease degradability produced the highest levels of sprouting relative to other physicochemical matrix properties. This engineered hydrogel was then utilized as a scaffold to facilitate the implantation of a donor vessel that functionally grafted into the host vasculature. This hydrogel provides a promising platform for facilitating lymphangiogenesis in vivo or as a means to understand the cellular mechanisms involved in the sprout process during collecting lymphatic vessel collateralization.
Collapse
Affiliation(s)
- Joshua S T Hooks
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. Atlanta, GA, 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Dr. Atlanta, GA, 30313, USA
| | - Fabrice C Bernard
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. Atlanta, GA, 30332, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Ricardo Cruz-Acuña
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. Atlanta, GA, 30332, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Zhanna Nepiyushchikh
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. Atlanta, GA, 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Dr. Atlanta, GA, 30313, USA
| | - Yarelis Gonzalez-Vargas
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. Atlanta, GA, 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Dr. Atlanta, GA, 30313, USA
| | - Andrés J García
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. Atlanta, GA, 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Dr. Atlanta, GA, 30313, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - J Brandon Dixon
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. Atlanta, GA, 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Dr. Atlanta, GA, 30313, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr NW, Atlanta, GA, 30332, USA.
| |
Collapse
|
48
|
Li J, Shen J, Zhuang B, Wei M, Liu Y, Liu D, Yan W, Jia X, Jin Y. Light-triggered on-site rapid formation of antibacterial hydrogel dressings for accelerated healing of infected wounds. BIOMATERIALS ADVANCES 2022; 136:212784. [PMID: 35929299 DOI: 10.1016/j.bioadv.2022.212784] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 06/15/2023]
Abstract
An optimal wound dressing can seal variously shaped wounds and provide a complete barrier to resist bacterial invasion; more importantly, the dressing can be stretched or compressed when the wounds are subjected to external forces and quickly return to its original state after the forces are withdrawn. Here, we designed dressings with light-triggered on-site rapid formation of antibacterial hydrogel for the accelerated healing of infected wounds. The pro-hydrogel, composed of acrylamide (AM) and dopamine-hyaluronic acid-ε-poly-l-lysine (DA-HA-EPL), was filled into the Vibrio vulnificus-infected wound. A 405-nm blue light was exerted on the wound to rapidly photopolymerize AM to its polymer, i.e., polyacrylamide (PAM). A hydrogel network of PAM/DA-HA-EPL immediately formed on site within several seconds to insulate the wound. PAM/DA-HA-EPL possessed adhesion performance to adapt to changes in wound morphologies due to external forces. Moreover, it presented high antibacterial ability due to the presence of EPL, in vitro biocompatibility and the ability to promote cell migration. Vibrio vulnificus-infected wounds were established on full-thickness mouse skin, and the hydrogel dressing exhibited high healing efficiency in terms of skin tissue regeneration, collagen deposition, and angiogenesis. PAM/DA-HA-EPL is a promising hydrogel dressing for the accelerated healing of infected wounds.
Collapse
Affiliation(s)
- Jingfei Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jintao Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bo Zhuang
- Department of Chemical Defense, Institute of NBC Defense, Beijing 102205, China
| | - Meng Wei
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yan Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Dongdong Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wenrui Yan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xueli Jia
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
49
|
Pontiggia L, Van Hengel IAJ, Klar A, Rütsche D, Nanni M, Scheidegger A, Figi S, Reichmann E, Moehrlen U, Biedermann T. Bioprinting and plastic compression of large pigmented and vascularized human dermo-epidermal skin substitutes by means of a new robotic platform. J Tissue Eng 2022; 13:20417314221088513. [PMID: 35495096 PMCID: PMC9044789 DOI: 10.1177/20417314221088513] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Indexed: 12/19/2022] Open
Abstract
Extensive availability of engineered autologous dermo-epidermal skin substitutes (DESS) with functional and structural properties of normal human skin represents a goal for the treatment of large skin defects such as severe burns. Recently, a clinical phase I trial with this type of DESS was successfully completed, which included patients own keratinocytes and fibroblasts. Yet, two important features of natural skin were missing: pigmentation and vascularization. The first has important physiological and psychological implications for the patient, the second impacts survival and quality of the graft. Additionally, accurate reproduction of large amounts of patient’s skin in an automated way is essential for upscaling DESS production. Therefore, in the present study, we implemented a new robotic unit (called SkinFactory) for 3D bioprinting of pigmented and pre-vascularized DESS using normal human skin derived fibroblasts, blood- and lymphatic endothelial cells, keratinocytes, and melanocytes. We show the feasibility of our approach by demonstrating the viability of all the cells after printing in vitro, the integrity of the reconstituted capillary network in vivo after transplantation to immunodeficient rats and the anastomosis to the vascular plexus of the host. Our work has to be considered as a proof of concept in view of the implementation of an extended platform, which fully automatize the process of skin substitution: this would be a considerable improvement of the treatment of burn victims and patients with severe skin lesions based on patients own skin derived cells.
Collapse
Affiliation(s)
- Luca Pontiggia
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Ingmar AJ Van Hengel
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Agnes Klar
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Dominic Rütsche
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Monica Nanni
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | | | | | - Ernst Reichmann
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Ueli Moehrlen
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Pediatric Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Zurich Center for Fetal Diagnosis and Treatment, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
50
|
Lymphatic Tissue Bioengineering for the Treatment of Postsurgical Lymphedema. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9040162. [PMID: 35447722 PMCID: PMC9025804 DOI: 10.3390/bioengineering9040162] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/17/2022] [Accepted: 03/20/2022] [Indexed: 01/28/2023]
Abstract
Lymphedema is characterized by progressive and chronic tissue swelling and inflammation from local accumulation of interstitial fluid due to lymphatic injury or dysfunction. It is a debilitating condition that significantly impacts a patient's quality of life, and has limited treatment options. With better understanding of the molecular mechanisms and pathophysiology of lymphedema and advances in tissue engineering technologies, lymphatic tissue bioengineering and regeneration have emerged as a potential therapeutic option for postsurgical lymphedema. Various strategies involving stem cells, lymphangiogenic factors, bioengineered matrices and mechanical stimuli allow more precisely controlled regeneration of lymphatic tissue at the site of lymphedema without subjecting patients to complications or iatrogenic injuries associated with surgeries. This review provides an overview of current innovative approaches of lymphatic tissue bioengineering that represent a promising treatment option for postsurgical lymphedema.
Collapse
|