1
|
Botelho CM, Rodríguez-Cabello JC, Barbosa MA. Harnessing Thrombospondin-1-Enabled Decellularized Nucleus Pulposus Matrices and Elastin-Like Recombinamers to Rebuild an Avascular Analogue of the Intervertebral Disc. J Biomed Mater Res A 2025; 113:e37911. [PMID: 40269577 DOI: 10.1002/jbm.a.37911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/21/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025]
Abstract
With the degeneration of the intervertebral disc (IVD), the ingrowth of vascular and neural structures occurs. Both nerves and blood vessels engage in the development of inflammation and the onset of discogenic pain. The present study aimed to produce a hierarchical biomaterial capable of inhibiting angiogenesis by emulating the microenvironment of non-degenerated IVDs. To this end, we have incorporated an angiogenesis modulator-thrombospondin-1 (TSP-1) into a three-dimensional (3D) hydrogel network containing decellularized nucleus pulposus (dNPs) and azide-cyclooctyne modified elastin-like recombinamers (ELRs). Following the decellularization of nucleus pulposus (NPs) isolated from bovine tissues, pre-gels (pGs) were assembled based on the acid-pepsin extraction of soluble collagens found in the dNPs. Given the inherent affinity of these macromolecules to TSP-1, which was corroborated by immunohistochemical analysis and FT-IR spectroscopy, the pGs were supplemented with two concentrations of TSP-1. Angiogenesis was evaluated using the chick chorioallantoic membrane (CAM) in vivo model. Conjugation of TSP-1 with the pGs resulted in a synergistic suppression of blood vessel formation. Complexation with the ELRs improved the viscoelastic moduli and the structural stability of the hydrogels, which maintained their hydration and osmolarity properties due to the presence of the dNPs. When placed in direct contact with human primary fibroblasts, the materials displayed high cytocompatibility and tunable degradation rates. Our findings indicate that TSP-1-enabled dNP-derived pGs inhibit angiogenesis in vivo, while the presence of the ELRs aids in improving the mechanical properties of the hydrogels, thus providing a platform for rebuilding an avascular analogue of the healthy IVD.
Collapse
Affiliation(s)
- Carlos Marinho Botelho
- i3S - Instituto de Investigação e Inovação Em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - José Carlos Rodríguez-Cabello
- BIOFORGE - Grupo de Materiales Avanzados y Nanobiotecnología, Universidad de Valladolid, L.A.D.I.S., CIBER-BBN, Valladolid, Spain
| | - Mário Adolfo Barbosa
- i3S - Instituto de Investigação e Inovação Em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
2
|
Casademont-Roca A, Xing Z, Bernardi M, Rookmaker M, de Kort L, de Graaf P. A novel vascularized urethra-on-a-chip model. Sci Rep 2025; 15:8062. [PMID: 40055501 DOI: 10.1038/s41598-025-92485-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 02/27/2025] [Indexed: 03/12/2025] Open
Abstract
The male urethra transports urine and semen. Any disease of the male urethra, hindering normal voiding or ejaculation, has a major impact on quality of life. Urethral stricture disease is common and molecular research into urethral strictures is hampered by the lack of reliable models of the human urethra. The aim of this project is to develop an in vitro model system of the human urethra. We hypothesized that by using the organ-on-a-chip technology we would be able to recapitulate physiology, functionality and the biomechanical cues of the native urethra and its surrounding vascular bed. Our approach consisted in using the F300R microfluidic device in combination with a rocking system to develop a potential urethra-on-a-chip. Urethral epithelial cells were used to mimic the native urethral epithelium. Gelatin-based hydrogels were tested for vasculogenic properties by placing the gel on the chick chorioallantoic membrane (CAM). Furthermore, the same gels were used for the formation of a micro vascular bed. Microvessel-like structures were formed in the gelatin-based hydrogels. Furthermore, these gels supported penetration, survival and proliferation of chicken endothelial cells when placed on the CAM. While we could only recapitulate a low fluidic shear stress (FSS) of 0.049 dyne/cm2, this was enough to form a confluent monolayer during dynamic conditions. This was not accomplished during static conditions. This project holds promise in mimicking the native layers of the urethra: epithelium and surrounding vascular tissue, under dynamic conditions. This new approach could provide a valuable platform to study the pathogenesis of urethral diseases and verify the effectiveness of drug treatment.
Collapse
Affiliation(s)
- Aina Casademont-Roca
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Zhentao Xing
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Murillo Bernardi
- Department of Nephrology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maarten Rookmaker
- Department of Nephrology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Laetitia de Kort
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Petra de Graaf
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
3
|
Heimes D, Wiesmann-Imilowski N, Heidebrecht T, Blatt S, Pabst A, Becker P, Fuest S, Brieger J, Smeets R, Kämmerer PW. Biofunctionalization of silk fibroin scaffolds with enamel matrix protein and injectable platelet rich fibrin for soft tissue augmentation: an in-ovo study. Int J Implant Dent 2025; 11:13. [PMID: 39976848 PMCID: PMC11842663 DOI: 10.1186/s40729-025-00601-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/12/2025] [Indexed: 02/23/2025] Open
Abstract
PURPOSE Silk fibroin (SF) is a biomaterial derived from the cocoon of the mulberry silkworm. This study aimed to assess the capacity of SF matrices biologized with injectable platelet-rich fibrin (iPRF) or enamel matrix protein (EMP) to modulate angiogenesis and immune response in the chorioallantoic membrane (CAM) assay. METHODS 300 eggs were divided into the following groups: CM + NaCl, CM + iPRF, CM + EMP, SF + NaCl, SF + iPRF, and SF + EMP. Matrices were applied to the CAM on embryonic development day (EDD) 7 after rehydration. Angiogenesis, represented by vascularized area, vessel density, and vessel junctions, was evaluated on EDD 10, 12, and 14. Additionally, gene expression of HIF-1ɑ, VEGF, MMP-13, and NOS2 was assessed via quantitative polymerase chain reaction (qPCR) on EDD 11 and 14. RESULTS The number of vascularized specimens was notably higher in SF matrices regardless of the treatment applied, while in the CM group, only matrices biofunctionalized with iPRF demonstrated vascularization. On EDD 14, the CM + iPRF group exhibited the highest values for total vascularized area (CM + iPRF: 57.52%, SF + iPRF: 21.87%, p < 0.001), vessel density (CM + iPRF: 0.0067 μm/µm2, SF + iPRF: 0.0032 μm/µm2, p = 0.002), number of vessel junctions (CM + iPRF: 14.45, SF + iPRF: 4.82, p = 0.001). Gene expressions displayed high data variability and no significant differences between the groups. CONCLUSIONS Biofunctionalization with iPRF in CM leads to a high vascularization rate probably through their capability of retaining higher liquid volumes, suggesting improved intraoral wound healing after guided tissue regeneration (GTR). Despite biofunctionalization, SF matrices exhibit a high vascularization, indicating SF as a promising material for GTR.
Collapse
Affiliation(s)
- Diana Heimes
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131, Mainz, Germany.
| | - Nadine Wiesmann-Imilowski
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131, Mainz, Germany
- Department of Otorhinolaryngology, University Medical Center Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Timpe Heidebrecht
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131, Mainz, Germany
| | - Sebastian Blatt
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131, Mainz, Germany
| | - Andreas Pabst
- Department of Oral and Maxillofacial Surgery, Federal Armed Forces Hospital, Rübenacherstr. 170, 56072, Koblenz, Germany
| | - Philipp Becker
- Department of Oral and Maxillofacial Surgery, Federal Armed Forces Hospital, Rübenacherstr. 170, 56072, Koblenz, Germany
| | - Sandra Fuest
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg- Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Jürgen Brieger
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131, Mainz, Germany
- Department of Otorhinolaryngology, University Medical Center Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg- Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Peer W Kämmerer
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131, Mainz, Germany
| |
Collapse
|
4
|
Phan CM, Luu CH, Murugesan M, Nguyen TNQ, Ha NYN, Ngo HL, Nguyen NDH, Pan Z, Phan VHG, Li Y, Thambi T. Injectable gelatin-pectin hydrogel for dental tissue engineering: Enhanced angiogenesis and antibacterial efficacy for pulpitis therapy. Int J Biol Macromol 2025; 284:137939. [PMID: 39592046 DOI: 10.1016/j.ijbiomac.2024.137939] [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: 10/07/2024] [Revised: 11/01/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024]
Abstract
Pulpitis is inflammation of the dental pulp, often caused by bacterial infection from untreated cavities, leading to pain. The main challenge in treatment is eliminating infection while preserving tooth vitality. This study aims to address this challenge by developing a hydrogel for convenient insertion into the root canal system, securely attaching to dentin walls. An injectable hydrogel system is developed by chemically cross-linking natural polysaccharide pectin with gelatin (GPG) through reversible Schiff base reaction. The GPG system was then used to encapsulate and release drugs, such as ciprofloxacin (CIP) for infection prevention and deferoxamine (DFO) for promoting blood vessel proliferation and reducing inflammatory reactions. The GPGs absorbed significant amounts of CIP and DFO, enabling sustained release over a nearly ten-day period. When subcutaneously implanted, the GPGs formed stable gel depots, with only 50 % of the gels degrading after 3 weeks, indicating a sustained biodegradation pattern. Additionally, the GPG system demonstrated excellent antibacterial activity against both gram-negative and gram-positive bacteria. Results from in vitro scratch healing tests and in ovo chorioallantoic membrane chick model tests showed promising biocompatibility and promotion of vascular proliferation by the GPG. This study heralds a novel frontier in endodontic therapeutics, poised to potentially enable dental pulp regeneration.
Collapse
Affiliation(s)
- Chau My Phan
- College of Materials and Textile Engineering & Nanotechnology Research Institute, Jiaxing University, Jiaxing 314001, Zhejiang Province, PR China; Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Cuong Hung Luu
- School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia; Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD 4111, Australia
| | - Mohanapriya Murugesan
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin si, Gyeonggi do 17104, Republic of Korea
| | - Thi-Nhu-Quynh Nguyen
- Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Nhu-Y Ngoc Ha
- Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Huong Lan Ngo
- Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Ngoc-Dan Ho Nguyen
- Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Zhouyi Pan
- College of Materials and Textile Engineering & Nanotechnology Research Institute, Jiaxing University, Jiaxing 314001, Zhejiang Province, PR China
| | - V H Giang Phan
- Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam.
| | - Yi Li
- College of Materials and Textile Engineering & Nanotechnology Research Institute, Jiaxing University, Jiaxing 314001, Zhejiang Province, PR China.
| | - Thavasyappan Thambi
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin si, Gyeonggi do 17104, Republic of Korea.
| |
Collapse
|
5
|
Sasan S, Molavi AM, Moqadam KH, Farrokhi N, Oroojalian F. Enhanced wound healing properties of biodegradable PCL/alginate core-shell nanofibers containing Salvia abrotanoides essential oil and ZnO nanoparticles. Int J Biol Macromol 2024; 279:135152. [PMID: 39214210 DOI: 10.1016/j.ijbiomac.2024.135152] [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: 01/11/2024] [Revised: 07/26/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Electrospun nanofibrous membranes, with their unique structural features, can potentially enhance wound healing through controlled delivery of active agents. Here, an innovative porous nanofibrous membrane was developed as a dressing patch with antibacterial and anti-inflammatory functionalities for cutaneous wound healing. Zinc oxide nanoparticles (ZnO NPs) and Salvia abrotanoides essential oil (SAEO) were incorporated into sodium alginate, which served as the shell. Poly(ε-caprolactone) was used as the core of coaxial electrospun wound dressing nanofibers (PCL/SA@ZnO/SAEO). With the addition of ZnO NPs and SAEO, the average diameter of nanofibers was 187 ± 51 nm, with improved tensile strength (4.7 ± 0.4 MPa), elongation at break (32.9 ± 2.1), and elastic modulus (21.4 ± 2.0). Concurrent application of ZnO NPs and SAEO increased antimicrobial activity against Staphylococcus aureus and Escherichia coli and promoted the proliferation, attachment, and viability (>90 %) of L929 cells. The PCL/SA@ZnO/SAEO scaffold accelerated the healing time with total wound healing over 14 days in mouse models carrying full-thickness wounds compared to the nanofibrous scaffold without additives. Histopathological examinations demonstrated better tissue regeneration, i.e., enhanced collagen deposition, improved re-epithelialization, and neovascularization, and increased quantity of hair follicles. Moreover, the chicken chorioallantoic membrane assay confirmed the synergistic angiogenic effects of SAEO and ZnO NPs. Finally, the in vitro and in vivo results proposed the bioactive core-shell nanofibers synthesized as encouraging wound dressing materials for hastening the healing of cutaneous wounds.
Collapse
Affiliation(s)
- Samira Sasan
- Department of Cell & Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Amir Mahdi Molavi
- Department of Materials Research, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | | | - Naser Farrokhi
- Department of Cell & Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Fatemeh Oroojalian
- Natural Products & Medicinal Plants Research Center, North Khorasan University of Medical Science, Bojnurd 74877-94149, Iran; Department of Medical Nanotechnology, School of Medicine, North Khorasan University of Medical Science, Bojnurd, Iran.
| |
Collapse
|
6
|
Oliveira M, Sarker PP, Skovorodkin I, Kalantarifard A, Haskavuk T, Mac Intyre J, Nallukunnel Raju E, Nooranian S, Shioda H, Nishikawa M, Sakai Y, Vainio SJ, Elbuken C, Raykhel I. From ex ovo to in vitro: xenotransplantation and vascularization of mouse embryonic kidneys in a microfluidic chip. LAB ON A CHIP 2024; 24:4816-4826. [PMID: 39290081 PMCID: PMC11408908 DOI: 10.1039/d4lc00547c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024]
Abstract
Organoids are emerging as a powerful tool to investigate complex biological structures in vitro. Vascularization of organoids is crucial to recapitulate the morphology and function of the represented human organ, especially in the case of the kidney, whose primary function of blood filtration is closely associated with blood circulation. Current in vitro microfluidic approaches have only provided initial vascularization of kidney organoids, whereas in vivo transplantation to animal models is problematic due to ethical problems, with the exception of xenotransplantation onto a chicken chorioallantoic membrane (CAM). Although CAM can serve as a good environment for vascularization, it can only be used for a fixed length of time, limited by development of the embryo. Here, we propose a novel lab on a chip design that allows organoids of different origin to be cultured and vascularized on a CAM, as well as to be transferred to in vitro conditions when required. Mouse embryonic kidneys cultured on the CAM showed enhanced vascularization by intrinsic endothelial cells, and made connections with the chicken vasculature, as evidenced by blood flowing through them. After the chips were transferred to in vitro conditions, the vasculature inside the organoids was successfully maintained. To our knowledge, this is the first demonstration of the combination of in vivo and in vitro approaches applied to microfluidic chip design.
Collapse
Affiliation(s)
- Micaela Oliveira
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Partha Protim Sarker
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| | - Ilya Skovorodkin
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| | - Ali Kalantarifard
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Tugce Haskavuk
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| | - Jonatan Mac Intyre
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Elizabath Nallukunnel Raju
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Samin Nooranian
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Hiroki Shioda
- Laboratory of Organs and Biosystems Engineering, Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Masaki Nishikawa
- Laboratory of Organs and Biosystems Engineering, Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Yasuyuki Sakai
- Laboratory of Organs and Biosystems Engineering, Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Seppo J Vainio
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
- Infotech Oulu, University of Oulu, Oulu, Finland
- Kvantum Institute, University of Oulu, Oulu, Finland
| | - Caglar Elbuken
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
- VTT Technical Research Centre of Finland Ltd., Finland
| | - Irina Raykhel
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
- Laboratory of Organs and Biosystems Engineering, Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Shekatkar M, Kheur S, Deshpande S, Sakhare S, Sanap A, Kheur M, Bhonde R. Critical appraisal of the chorioallantoic membrane model for studying angiogenesis in preclinical research. Mol Biol Rep 2024; 51:1026. [PMID: 39340708 DOI: 10.1007/s11033-024-09956-x] [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: 03/01/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Angiogenesis, the biological mechanism by which new blood vessels are generated from existing ones, plays a vital role in growth and development. Effective preclinical screening is necessary for the development of medications that may enhance or inhibit angiogenesis in the setting of different disorders. Traditional in vitro and, in vivo models of angiogenesis are laborious and time-consuming, necessitating advanced infrastructure for embryo culture. MAIN BODY A challenge encountered by researchers studying angiogenesis is the lack of appropriate techniques to evaluate the impact of regulators on the angiogenic response. An ideal test should possess reliability, technical simplicity, easy quantifiability, and, most importantly, physiological relevance. The CAM model, leveraging the extraembryonic membrane of the chicken embryo, offers a unique combination of accessibility, low cost, and rapid development, making it an attractive option for angiogenesis assays. This review evaluates the strengths and limitations of the CAM model in the context of its anatomical and physiological properties, and its relevance to human pathophysiological conditions. Its abundant capillary network makes it a common choice for studying angiogenesis. The CAM assay serves as a substitute for animal models and offers a natural setting for developing blood vessels and the many elements involved in the intricate interaction with the host. Despite its advantages, the CAM model's limitations are notable. These include species-specific responses that may not always extrapolate to humans and the ethical considerations of using avian embryos. We discuss methodological adaptations that can mitigate some of these limitations and propose future directions to enhance the translational relevance of this model. This review underscores the CAM model's valuable role in angiogenesis research and aims to guide researchers in optimizing its use for more predictive and robust preclinical studies. CONCLUSION The highly vascularized chorioallantoic membrane (CAM) of fertilized chicken eggs is a cost-effective and easily available method for screening angiogenesis, in comparison to other animal models.
Collapse
Affiliation(s)
- Madhura Shekatkar
- Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| | - Supriya Kheur
- Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India.
| | - Shantanu Deshpande
- Department of Pediatric and Preventive Dentistry, Bharati Vidyapeeth (Deemed to be University), Dental College and Hospital, Navi Mumbai, India
| | - Swapnali Sakhare
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Avinash Sanap
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Mohit Kheur
- Department of Prosthodontics, M.A. Rangoonwala College of Dental Sciences and Research Centre, Pune, Maharashtra, India
| | - Ramesh Bhonde
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| |
Collapse
|
8
|
Abbasi-Malati Z, Khanicheragh P, Narmi MT, Mardi N, Khosrowshahi ND, Hiradfar A, Rezabakhsh A, Sadeghsoltani F, Rashidi S, Chegeni SA, Roozbahani G, Rahbarghazi R. Tumoroids, a valid preclinical screening platform for monitoring cancer angiogenesis. Stem Cell Res Ther 2024; 15:267. [PMID: 39183337 PMCID: PMC11346257 DOI: 10.1186/s13287-024-03880-4] [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: 02/25/2024] [Accepted: 08/06/2024] [Indexed: 08/27/2024] Open
Abstract
In recent years, biologists and clinicians have witnessed prominent advances in in vitro 3D culture techniques related to biomimetic human/animal tissue analogs. Numerous data have confirmed that unicellular and multicellular (tumoroids) tumor spheroids with dense native cells in certain matrices are sensitive and valid analytical tools for drug screening, cancer cell dynamic growth, behavior, etc. in laboratory settings. Angiogenesis/vascularization is a very critical biological phenomenon to support oxygen and nutrients to tumor cells within the deep layer of solid masses. It has been shown that endothelial cell (EC)-incorporated or -free spheroid/tumoroid systems provide a relatively reliable biological platform for monitoring the formation of nascent blood vessels in micron/micrometer scales. Besides, the paracrine angiogenic activity of cells within the spheroid/tumoroid systems can be monitored after being treated with different therapeutic approaches. Here, we aimed to collect recent advances and findings related to the monitoring of cancer angiogenesis using unicellular and multicellular tumor spheroids. Vascularized spheroids/tumoroids can help us in the elucidation of mechanisms related to cancer formation, development, and metastasis by monitoring the main influencing factors.
Collapse
Affiliation(s)
- Zahra Abbasi-Malati
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Khanicheragh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Narges Mardi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nafiseh Didar Khosrowshahi
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Amirataollah Hiradfar
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysa Rezabakhsh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Somayyeh Rashidi
- Department of Medical Biotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Golbarg Roozbahani
- Department of Plant, Cell and Molecular Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
9
|
Simińska-Stanny J, Nicolas L, Chafai A, Jafari H, Hajiabbas M, Dodi G, Gardikiotis I, Delporte C, Nie L, Podstawczyk D, Shavandi A. Advanced PEG-tyramine biomaterial ink for precision engineering of perfusable and flexible small-diameter vascular constructs via coaxial printing. Bioact Mater 2024; 36:168-184. [PMID: 38463551 PMCID: PMC10924180 DOI: 10.1016/j.bioactmat.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/09/2024] [Accepted: 02/16/2024] [Indexed: 03/12/2024] Open
Abstract
Vascularization is crucial for providing nutrients and oxygen to cells while removing waste. Despite advances in 3D-bioprinting, the fabrication of structures with void spaces and channels remains challenging. This study presents a novel approach to create robust yet flexible and permeable small (600-1300 μm) artificial vessels in a single processing step using 3D coaxial extrusion printing of a biomaterial ink, based on tyramine-modified polyethylene glycol (PEG-Tyr). We combined the gelatin biocompatibility/activity, robustness of PEG-Tyr and alginate with the shear-thinning properties of methylcellulose (MC) in a new biomaterial ink for the fabrication of bioinspired vessels. Chemical characterization using NMR and FTIR spectroscopy confirmed the successful modification of PEG with Tyr and rheological characterization indicated that the addition of PEG-Tyr decreased the viscosity of the ink. Enzyme-mediated crosslinking of PEG-Tyr allowed the formation of covalent crosslinks within the hydrogel chains, ensuring its stability. PEG-Tyr units improved the mechanical properties of the material, resulting in stretchable and elastic constructs without compromising cell viability and adhesion. The printed vessel structures displayed uniform wall thickness, shape retention, improved elasticity, permeability, and colonization by endothelial-derived - EA.hy926 cells. The chorioallantoic membrane (CAM) and in vivo assays demonstrated the hydrogel's ability to support neoangiogenesis. The hydrogel material with PEG-Tyr modification holds promise for vascular tissue engineering applications, providing a flexible, biocompatible, and functional platform for the fabrication of vascular structures.
Collapse
Affiliation(s)
- Julia Simińska-Stanny
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Lise Nicolas
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
- European School of Materials Science and Engineering, University of Lorraine, Nancy, France
| | - Adam Chafai
- Université Libre de Bruxelles (ULB), Micro-milli Platform, Avenue F.D. Roosevelt, 50 - CP 165/67, 1050, Brussels, Belgium
| | - Hafez Jafari
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Maryam Hajiabbas
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
- Université Libre de Bruxelles (ULB), Faculté de Médecine, Campus Erasme - CP 611, Laboratory of Pathophysiological and Nutritional Biochemistry, Route de Lennik, 808, 1070, Bruxelles, Belgium
| | - Gianina Dodi
- Faculty of Medical Bioengineering, Grigore T. Popa, University of Medicine and Pharmacy of Iasi, Romania
| | - Ioannis Gardikiotis
- Advanced Research and Development Center for Experimental Medicine, Grigore T. Popa, University of Medicine and Pharmacy of Iasi, Romania
| | - Christine Delporte
- Université Libre de Bruxelles (ULB), Faculté de Médecine, Campus Erasme - CP 611, Laboratory of Pathophysiological and Nutritional Biochemistry, Route de Lennik, 808, 1070, Bruxelles, Belgium
| | - Lei Nie
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Daria Podstawczyk
- Department of Process Engineering and Technology of Polymer and Carbon Materials, Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, 50-373, Wroclaw, Poland
| | - Amin Shavandi
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| |
Collapse
|
10
|
Li D, Li Q, Xu T, Guo X, Tang H, Wang W, Zhang W, Zhang Y. Pro-vasculogenic Fibers by PDA-Mediated Surface Functionalization Using Cell-Free Fat Extract (CEFFE). Biomacromolecules 2024; 25:1550-1562. [PMID: 38411008 DOI: 10.1021/acs.biomac.3c01124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Formation of adequate vascular network within engineered three-dimensional (3D) tissue substitutes postimplantation remains a major challenge for the success of biomaterials-based tissue regeneration. To better mimic the in vivo angiogenic and vasculogenic processes, nowadays increasing attention is given to the strategy of functionalizing biomaterial scaffolds with multiple bioactive agents. Aimed at engineering electrospun biomimicking fibers with pro-vasculogenic capability, this study was proposed to functionalize electrospun fibers of polycaprolactone/gelatin (PCL/GT) by cell-free fat extract (CEFFE or FE), a newly emerging natural "cocktail" of cytokines and growth factors extracted from human adipose tissue. This was achieved by having the electrospun PCL/GT fiber surface coated with polydopamine (PDA) followed by PDA-mediated immobilization of FE to generate the pro-vasculogenic fibers of FE-PDA@PCL/GT. It was found that the PDA-coated fibrous mat of PCL/GT exhibited a high FE-loading efficiency (∼90%) and enabled the FE to be released in a highly sustained manner. The engineered FE-PDA@PCL/GT fibers possess improved cytocompatibility, as evidenced by the enhanced cellular proliferation, migration, and RNA and protein expressions (e.g., CD31, vWF, VE-cadherin) in the human umbilical vein endothelial cells (huvECs) used. Most importantly, the FE-PDA@PCL/GT fibrous scaffolds were found to enormously stimulate tube formation in vitro, microvascular development in the in ovo chick chorioallantoic membrane (CAM) assay, and vascularization of 3D construct in a rat subcutaneous embedding model. This study highlights the potential of currently engineered pro-vasculogenic fibers as a versatile platform for engineering vascularized biomaterial constructs for functional tissue regeneration.
Collapse
Affiliation(s)
- Donghong Li
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Qinglin Li
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Tingting Xu
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Xuran Guo
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Han Tang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Wenbo Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yanzhong Zhang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, China
| |
Collapse
|
11
|
Conci C, Sironi L, Jacchetti E, Panzeri D, Inverso D, Martínez Vázquez R, Osellame R, Collini M, Cerullo G, Chirico G, Raimondi MT. In vivo label-free tissue histology through a microstructured imaging window. APL Bioeng 2024; 8:016102. [PMID: 38222895 PMCID: PMC10787586 DOI: 10.1063/5.0165411] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/06/2023] [Indexed: 01/16/2024] Open
Abstract
Tissue histopathology, based on hematoxylin and eosin (H&E) staining of thin tissue slices, is the gold standard for the evaluation of the immune reaction to the implant of a biomaterial. It is based on lengthy and costly procedures that do not allow longitudinal studies. The use of non-linear excitation microscopy in vivo, largely label-free, has the potential to overcome these limitations. With this purpose, we develop and validate an implantable microstructured device for the non-linear excitation microscopy assessment of the immune reaction to an implanted biomaterial label-free. The microstructured device, shaped as a matrix of regular 3D lattices, is obtained by two-photon laser polymerization. It is subsequently implanted in the chorioallantoic membrane (CAM) of embryonated chicken eggs for 7 days to act as an intrinsic 3D reference frame for cell counting and identification. The histological analysis based on H&E images of the tissue sections sampled around the implanted microstructures is compared to non-linear excitation and confocal images to build a cell atlas that correlates the histological observations to the label-free images. In this way, we can quantify the number of cells recruited in the tissue reconstituted in the microstructures and identify granulocytes on label-free images within and outside the microstructures. Collagen and microvessels are also identified by means of second-harmonic generation and autofluorescence imaging. The analysis indicates that the tissue reaction to implanted microstructures is like the one typical of CAM healing after injury, without a massive foreign body reaction. This opens the path to the use of similar microstructures coupled to a biomaterial, to image in vivo the regenerating interface between a tissue and a biomaterial with label-free non-linear excitation microscopy. This promises to be a transformative approach, alternative to conventional histopathology, for the bioengineering and the validation of biomaterials in in vivo longitudinal studies.
Collapse
Affiliation(s)
- Claudio Conci
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Laura Sironi
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Emanuela Jacchetti
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Davide Panzeri
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Donato Inverso
- Division of Immunology, Transplantation and Infectious Diseases IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Rebeca Martínez Vázquez
- Institute for Photonics and Nanotechnologies (IFN), CNR and Department of Physics, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Roberto Osellame
- Institute for Photonics and Nanotechnologies (IFN), CNR and Department of Physics, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Maddalena Collini
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Giulio Cerullo
- Institute for Photonics and Nanotechnologies (IFN), CNR and Department of Physics, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Giuseppe Chirico
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| |
Collapse
|
12
|
Salah RA, El-Derby AM, El-Gammal Z, Wadie B, Ahmed SM, Elshenawy SE, Magdy S, Salah A, Gabr M, Mohamed I, El-Badri N. Hepatocellular carcinoma patients serum modulates the regenerative capacities of adipose mesenchymal stromal cells. Heliyon 2024; 10:e24794. [PMID: 38333871 PMCID: PMC10850426 DOI: 10.1016/j.heliyon.2024.e24794] [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: 04/22/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent cancers causing the highest mortality rate worldwide. Treatment options of surgery, radiation, cytotoxic drugs and liver transplantation suffer significant side effects and a high frequency of relapse. Stem cell therapy has been proposed as a new effective therapy, however, controversial reports are emerging on the role of mesenchymal stem cells in cancer. In this work, we aimed to assess the regenerative capacities of adipose mesenchymal stem cells when exposed to serum from HCC patients, by assessing the effect of the sera on modulating the regenerative capacities of h-AMSCs and the cancer properties in HCC cells. This will pave the way for maximizing the efficacy of MSCs in cancer therapy. Our data show that HCC serum-treated hA-MSCs suffered oncogene-induced senescence as shown by their altered morphology and ameliorated proliferation and differentiation. The cells were enlarged with small irregular nuclei, swollen rough endoplasmic reticulum cisternae, and aging lysosomes typified by dark residual bodies. HCC serum-treated Huh-7 cancer cells on the other hand displayed higher tumor aggressiveness as depicted by altered morphology, increased cellular proliferation and migration, and decreased percentage of early and late apoptotic cells. Our findings provide evidence that exposure of hA-MSCs to the serum of HCC patients decreases their regenerative capacities and should be considered when employed as a potential therapy in HCC patients.
Collapse
Affiliation(s)
- Radwa Ayman Salah
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Azza M. El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Zaynab El-Gammal
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
- Stem Cells and Regenerative Medicine Department, Egypt Center for Research and Regenerative Medicine (ECRRM), Giza, 12578, Egypt
| | - Bishoy Wadie
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Sara M. Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Shimaa E. Elshenawy
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
- Stem Cells and Regenerative Medicine Department, Egypt Center for Research and Regenerative Medicine (ECRRM), Giza, 12578, Egypt
| | - Shireen Magdy
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Ayman Salah
- Department of Hepatogastroenterology, Kasr El-Aini Cairo University, Cairo, Egypt
| | - Mahmoud Gabr
- Urology and Nephrology Center, Mansoura, 35516, Egypt
| | - Ihab Mohamed
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| |
Collapse
|
13
|
Krattiger LA, Moser LO, Odabasi R, Odriozola A, Simona BR, Djonov V, Tibbitt MW, Ehrbar M. Recovery of Therapeutically Ablated Engineered Blood-Vessel Networks on a Plug-and-Play Platform. Adv Healthc Mater 2024; 13:e2301142. [PMID: 37946678 DOI: 10.1002/adhm.202301142] [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: 06/20/2023] [Revised: 10/13/2023] [Indexed: 11/12/2023]
Abstract
Limiting the availability of key angiogenesis-promoting factors is a successful strategy to ablate tumor-supplying blood vessels or to reduce excessive vasculature in diabetic retinopathy. However, the efficacy of such anti-angiogenic therapies (AATs) varies with tumor type, and regrowth of vessels is observed upon termination of treatment. The ability to understand and develop AATs remains limited by a lack of robust in vitro systems for modeling the recovery of vascular networks. Here, complex 3D micro-capillary networks are engineered by sequentially seeding human bone marrow-derived mesenchymal stromal cells and human umbilical vein endothelial cells (ECs) on a previously established, synthetic plug-and-play hydrogel platform. In the tightly interconnected vascular networks that form this way, the two cell types share a basement membrane-like layer and can be maintained for several days of co-culture. Pre-formed networks degrade in the presence of bevacizumab. Upon treatment termination, vessel structures grow back to their original positions after replenishment with new ECs, which also integrate into unperturbed established networks. The data suggest that this plug-and-play platform enables the screening of drugs with blood-vessel inhibiting functions. It is believed that this platform could be of particular interest in studying resistance or recovery mechanisms to AAT treatment.
Collapse
Affiliation(s)
- Lisa A Krattiger
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, Zurich, 8091, Switzerland
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, Zürich, 8092, Switzerland
| | - Lukas O Moser
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, Zurich, 8091, Switzerland
| | - Rodi Odabasi
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, Zurich, 8091, Switzerland
| | - Adolfo Odriozola
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, Bern, 3012, Switzerland
| | - Benjamin R Simona
- Ectica Technologies AG, Raeffelstrasse 24, Zurich, 8045, Switzerland
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, Bern, 3012, Switzerland
| | - Mark W Tibbitt
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, Zürich, 8092, Switzerland
| | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, Zurich, 8091, Switzerland
| |
Collapse
|
14
|
Aadinath W, Muthuvijayan V. Antibacterial and angiogenic potential of iron oxide nanoparticles-stabilized acrylate-based scaffolds for bone tissue engineering applications. Colloids Surf B Biointerfaces 2023; 231:113572. [PMID: 37797467 DOI: 10.1016/j.colsurfb.2023.113572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/07/2023]
Abstract
Pickering emulsion polymerization, stabilized by inorganic nanoparticles such as iron oxide nanoparticles (IONPs), can be used to fabricate scaffolds with the desired porosity and pore size. These nanoparticles create stable emulsions that can be processed under harsh polymerization conditions. IONPs, apart from serving as an emulsifier, impart beneficial bioactivities such as antibacterial and pro-angiogenic activity. Here, we coated IONPs with three different weights of oleic acid (5.0 g, 7.5 g, and 10.0 g) to synthesize oleic acid-IONPs (OA-IONPs) that possess the desired hydrophobicity (contact angle > 100°). Next, glycidyl methacrylate and trimethylolpropane triacrylate were polymerized using the Pickering emulsion polymerization technique stabilized by the OA-IONPs. The physicochemical properties of the resulting porous scaffolds were thoroughly characterized using scanning electron microscopy (SEM), thermogravimetric analysis (TGA), Fourier transform infrared spectroscopy (FT-IR), vibrating sample magnetometry (VSM), and a universal testing machine (UTM). The SEM images confirmed the formation of a porous scaffold. The IONPs content, measured using inductively coupled plasma mass spectrometry (ICP-MS), was in the range of 22-26 µg/mg of the scaffold. The mechanical strengths of the scaffolds were in the range of cancellous bone. The degradation profile of the scaffolds varied between 29% and 41% degradation over 30 days. In vitro cytotoxicity studies conducted using the fibroblast (L929) and osteosarcoma (MG-63) cell lines proved that these scaffolds were non-toxic. SEM images showed that the MG-63 cells adhered firmly to the scaffolds and exhibited a well-spread morphology. The antibacterial activity was confirmed by percentage inhibition studies, SEM analysis of bacterial membrane distortion, and reactive oxygen species (ROS) generation in the bacteria. Chick chorioallantoic membrane assay showed that the total vessel length and branch points were significantly increased in the presence of the scaffolds. These results confirm the pro-angiogenic potential of the fabricated scaffolds. The physicochemical, mechanical, and biological properties of the material suggest that the developed scaffolds would be suitable for bone tissue engineering applications.
Collapse
Affiliation(s)
- W Aadinath
- Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India
| | - Vignesh Muthuvijayan
- Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India.
| |
Collapse
|
15
|
Chen L, Wang Z, Fu X, Wang S, Feng Y, Coudyzer W, Wu S, Zhang H, Chai Z, Li Y, Ni Y. Dynamic 3D morphology of chick embryos and allantois depicted nondestructively by 3.0T clinical magnetic resonance imaging. Poult Sci 2023; 102:102902. [PMID: 37429051 PMCID: PMC10466300 DOI: 10.1016/j.psj.2023.102902] [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: 02/14/2023] [Revised: 06/19/2023] [Accepted: 06/24/2023] [Indexed: 07/12/2023] Open
Abstract
Driven by a global trend of applying replace-reduce-refine or 3Rs' guidance for experimental animals in life sciences, chick embryo and particularly allantois with its chorioallantoic membrane have been increasingly utilized to substitute laboratory animals, which call for more extensive and updated knowledge about this novel experimental setup. In this study, being noninvasive, nonionizing, and super-contrasting with high spatiotemporal resolutions, magnetic resonance imaging (MRI) was chosen as an imaging modality for in ovo monitoring morphologic evolution of the chick embryo, allantois, and chorioallantoic membrane longitudinally throughout embryonic day (ED) 1 until ED20. Cooled in 0°C ice bath for 60 min to reduce MRI motion artifacts, 3 chick embryos (n = 60 in total) on each ED were scanned by a clinical 3.0T MRI scanner to demonstrate 3D images of both T2- and T1-weighted imaging (T2WI, T1WI) sequences at axial, sagittal, and coronal slices. The volumes of both the entire chick embryo and allantois were semi-automatically segmented based on intensity-based thresholding and region-growing algorithms. The morphometries or quantified 3D structures were achieved by refined segmentation, and confirmed by histological analyses (one for each ED). After MRI, the rest of chick embryos (n = 40) continued for incubation. The images from ED2 to ED4 could demonstrate the structural changes of latebra, suggesting its transition into a nutrient supplying channel of yolk sac. The allantois could be recognized by MRI, and its relative volumes on each ED revealed an evolving profile peaked on ED12, with a statistically significant difference from those of earlier and later EDs (P < 0.01). The hypointensity of the yolk due to the susceptibility effect of its enriched iron content overshadowed the otherwise hyperintensity of its lipid components. The chick embryos survived prior cooling and MRI till hatching on ED21. The results could be further developed into a 3D MRI atlas of chick embryo. Clinical 3.0T MRI proved effective as a noninvasive approach to study in ovo 3D embryonic development across the full period (ED1-ED20), which can complement the present knowhow for poultry industry and biomedical science.
Collapse
Affiliation(s)
- Lei Chen
- KU Leuven, Campus Gasthuisberg, Leuven 3000, Belgium
| | | | - Xubin Fu
- Tianjin Ringpu Bio-Technology Co., Ltd., Tianjin, China
| | - Shuncong Wang
- KU Leuven, Campus Gasthuisberg, Leuven 3000, Belgium
| | - Yuanbo Feng
- KU Leuven, Campus Gasthuisberg, Leuven 3000, Belgium
| | | | - Shugeng Wu
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture & Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Haijun Zhang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture & Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhihong Chai
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yue Li
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yicheng Ni
- Department of Radiology, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
16
|
Hu X, Ma Z, Xu B, Li S, Yao Z, Liang B, Wang J, Liao W, Lin L, Wang C, Zheng S, Wu Q, Huang Q, Yu L, Wang F, Shi M. Glutamine metabolic microenvironment drives M2 macrophage polarization to mediate trastuzumab resistance in HER2-positive gastric cancer. Cancer Commun (Lond) 2023; 43:909-937. [PMID: 37434399 PMCID: PMC10397568 DOI: 10.1002/cac2.12459] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 01/04/2023] [Accepted: 06/21/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND Trastuzumab is a first-line targeted therapy for human epidermal growth factor receptor-2 (HER2)-positive gastric cancer. However, the inevitable occurrence of acquired trastuzumab resistance limits the drug benefit, and there is currently no effective reversal measure. Existing researches on the mechanism of trastuzumab resistance mainly focused on tumor cells themselves, while the understanding of the mechanisms of environment-mediated drug resistance is relatively lacking. This study aimed to further explore the mechanisms of trastuzumab resistance to identify strategies to promote survival in these patients. METHODS Trastuzumab-sensitive and trastuzumab-resistant HER2-positive tumor tissues and cells were collected for transcriptome sequencing. Bioinformatics were used to analyze cell subtypes, metabolic pathways, and molecular signaling pathways. Changes in microenvironmental indicators (such as macrophage, angiogenesis, and metabolism) were verified by immunofluorescence (IF) and immunohistochemical (IHC) analyses. Finally, a multi-scale agent-based model (ABM) was constructed. The effects of combination treatment were further validated in nude mice to verify these effects predicted by the ABM. RESULTS Based on transcriptome sequencing, molecular biology, and in vivo experiments, we found that the level of glutamine metabolism in trastuzumab-resistant HER2-positive cells was increased, and glutaminase 1 (GLS1) was significantly overexpressed. Meanwhile, tumor-derived GLS1 microvesicles drove M2 macrophage polarization. Furthermore, angiogenesis promoted trastuzumab resistance. IHC showed high glutamine metabolism, M2 macrophage polarization, and angiogenesis in trastuzumab-resistant HER2-positive tumor tissues from patients and nude mice. Mechanistically, the cell division cycle 42 (CDC42) promoted GLS1 expression in tumor cells by activating nuclear factor kappa-B (NF-κB) p65 and drove GLS1 microvesicle secretion through IQ motif-containing GTPase-activating protein 1 (IQGAP1). Based on the ABM and in vivo experiments, we confirmed that the combination of anti-glutamine metabolism, anti-angiogenesis, and pro-M1 polarization therapy had the best effect in reversing trastuzumab resistance in HER2-positive gastric cancer. CONCLUSIONS This study revealed that tumor cells secrete GLS1 microvesicles via CDC42 to promote glutamine metabolism, M2 macrophage polarization, and pro-angiogenic function of macrophages, leading to acquired trastuzumab resistance in HER2-positive gastric cancer. A combination of anti-glutamine metabolism, anti-angiogenesis, and pro-M1 polarization therapy may provide a new insight into reversing trastuzumab resistance.
Collapse
Affiliation(s)
- Xingbin Hu
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Zhenfeng Ma
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Beibei Xu
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Shulong Li
- School of Biomedical EngineeringSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Zhiqi Yao
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Bishan Liang
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Jiao Wang
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Wangjun Liao
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Li Lin
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Chunling Wang
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Siting Zheng
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Qijing Wu
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Qiong Huang
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Le Yu
- School of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Fenghua Wang
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdongP. R. China
| | - Min Shi
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| |
Collapse
|
17
|
Simińska-Stanny J, Hachemi F, Dodi G, Cojocaru FD, Gardikiotis I, Podstawczyk D, Delporte C, Jiang G, Nie L, Shavandi A. Optimizing phenol-modified hyaluronic acid for designing shape-maintaining biofabricated hydrogel scaffolds in soft tissue engineering. Int J Biol Macromol 2023:125201. [PMID: 37270140 DOI: 10.1016/j.ijbiomac.2023.125201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/18/2023] [Accepted: 05/31/2023] [Indexed: 06/05/2023]
Abstract
In this study, we developed a well-printable biomaterial ink for 3D printing of shape-maintaining hydrogel scaffolds. The hydrogel base comprised tyramine-modified hyaluronic acid (HA-Tyr) and gelatin methacrylate (GelMA) and was dually cross-linked. Using the Box-Behnken design, we explored how varying the ink composition affected fiber formation and shape preservation. By adjusting the polymer ratios, we produced a stable hydrogel with varying responses, from a viscous liquid to a thick gel, and optimized 3D scaffolds that were structurally stable both during and after printing, offering precision and flexibility. Our ink exhibited shear-thinning behavior and high swelling capacity, as well as ECM-like characteristics and biocompatibility, making it an ideal candidate for soft tissues matrices with storage modulus of around 300 Pa. Animal trials and CAM assays confirmed its biocompatibility and integration with host tissue.
Collapse
Affiliation(s)
- Julia Simińska-Stanny
- Université libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium; Department of Process Engineering and Technology of Polymer and Carbon Materials, Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, 50-373 Wroclaw, Poland
| | - Feza Hachemi
- Université libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium; Université Paris Saclay, Polytech Paris Saclay, Rue Louis de Broglie, 91400 Orsay, France
| | - Gianina Dodi
- Faculty of Medical Bioengineering, Grigore T. Popa, University of Medicine and Pharmacy of Iasi, Romania
| | - Florina D Cojocaru
- Faculty of Medical Bioengineering, Grigore T. Popa, University of Medicine and Pharmacy of Iasi, Romania
| | - Ioannis Gardikiotis
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa, University of Medicine and Pharmacy of Iasi, Romania
| | - Daria Podstawczyk
- Department of Process Engineering and Technology of Polymer and Carbon Materials, Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, 50-373 Wroclaw, Poland
| | - Christine Delporte
- Université libre de Bruxelles (ULB), Faculté de Médecine, Campus Erasme - CP 611, Laboratory of Pathophysiological and Nutritional Biochemistry, Route de Lennik, 808, 1070 Bruxelles, Belgium
| | - Guohua Jiang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China; International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers, Zhejiang Sci-Tech University, Hangzhou, China
| | - Lei Nie
- Université libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium; College of Life Science, Xinyang Normal University, Xinyang, China
| | - Amin Shavandi
- Université libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium.
| |
Collapse
|
18
|
Subbaraj GK, Masoodi T, Yasam SK, Chandrashekar K, Kulanthaivel L, Shaik NA, Hashem S, Alshabeeb Akil AS, Bhat AA. Anti-angiogenic effect of nano-formulated water soluble kaempferol and combretastatin in an in vivo chick chorioallantoic membrane model and HUVEC cells. Biomed Pharmacother 2023; 163:114820. [PMID: 37141736 DOI: 10.1016/j.biopha.2023.114820] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/19/2023] [Accepted: 04/30/2023] [Indexed: 05/06/2023] Open
Abstract
The present study evaluated the efficacy of nano-formulated water-soluble kaempferol and combretastatin alone and combined against the native kaempferol and combretastatin on angiogenesis. The solvent evaporation method was used to synthesize the nano-formulated water-soluble kaempferol and combretastatin and characterized using various analyses such as dynamic light scattering (DLS) and Fourier-transform infrared (FT-IR) spectroscopy.The anti-angiogenic activity of native, nano-formulated water-soluble kaempferol and combretastatin was investigated by cell viability on HUVEC and A498 cell lines, while chick chorioallantoic membrane (CAM) assay was utilized to assess morphometric and histopathological changes, and mRNA expressions of VEGF-A and FGF2 using qRT-PCR. MTT assay results revealed that the combination of nano-formulated water-soluble kaempferol and combretastatin significantly reduced the cell viability compared to control, individual treatments of native, nano-formulated water-soluble kaempferol, and combretastatin. Morphometric analysis of CAM showed that treatment with nano-formulated water-soluble kaempferol and combretastatin caused a substantial decrease in density, vessel network, branch points, and nets of CAM blood vessels. The histopathological results of CAM showed the irregular shape of blood vessels at the thin stratum of chronic endoderm, and blood capillaries were diminished compared to the control. In addition, the mRNA expression levels of VEGF-A and FGF2 were significantly decreased compared with native forms. Therefore, the findings of this study indicate that nano-formulated water-soluble combretastatin and kaempferol suppress angiogenesis by preventing the activation of endothelial cells and suppressing factors of angiogenesis. Moreover, a combination of nano-formulated water-soluble kaempferol and combretastatin worked much better than individual treatments.
Collapse
Affiliation(s)
- Gowtham Kumar Subbaraj
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education (Deemed to be University), Kelambakkam 603103, India.
| | - Tariq Masoodi
- Laboratory of Cancer Immunology and Genetics, Sidra Medicine, Doha, Qatar
| | - Santhosh Kumar Yasam
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education (Deemed to be University), Kelambakkam 603103, India
| | | | - Langeswaran Kulanthaivel
- Cancer Genetics & Molecular Biology Laboratory, Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu 630003, India
| | - Noor Ahmad Shaik
- Department of Genetics Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sheema Hashem
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
| | - Ammira S Alshabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
19
|
Javed S, Soukhtehzari S, Salmond N, Fernandes N, Williams KC. Development of an in vivo system to model breast cancer metastatic organotropism and evaluate treatment response using the chick embryo. iScience 2023; 26:106305. [PMID: 36950119 PMCID: PMC10025954 DOI: 10.1016/j.isci.2023.106305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/21/2022] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Metastatic lesions produced through the process of systemic tumor cell dissemination and growth at distant sites are challenging to treat and the primary cause of patient mortality. Developing in vivo models of metastasis with utility in evaluating molecular targets and therapeutics in a timely manner would expedite the path to therapeutic discovery. Here, we evaluated breast cancer metastasis and metastatic organotropism using the chick embryo. We developed a method to evaluate metastasis using the MDA231 cell line. Then, using cell lines with demonstrated tropism for the bone, brain, and lung, we evaluated organotropism. Rapid and robust organ-specific metastasis was modeled in the chick embryo and, importantly, recapitulated metastatic organotropism congruent to what has been demonstrated in mice. Treatment response in the metastatic setting was also evaluated and quantified. This work establishes the chick embryo as a model for studies aimed at understanding organotropism and therapeutic response in the metastatic setting.
Collapse
Affiliation(s)
- Sumreen Javed
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, Canada
| | - Sepideh Soukhtehzari
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, Canada
| | - Nikki Salmond
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, Canada
| | - Nazarine Fernandes
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, Canada
| | - Karla C. Williams
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, Canada
- Corresponding author
| |
Collapse
|
20
|
Halgrain M, Bernardet N, Hennequet-Antier C, Hincke M, Réhault-Godbert S. RNA-seq analysis of the active chick embryo chorioallantoic membrane reveals genes that encode proteins assigned to ion transport and innate immunity. Genomics 2023; 115:110564. [PMID: 36642281 DOI: 10.1016/j.ygeno.2023.110564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/06/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
The chicken chorioallantoic membrane (CAM) is an extraembryonic membrane that is vital for the embryo. It undergoes profound cell differentiation between 11 and 15 days of embryonic incubation (EID), which corresponds to the acquisition of its physiological functions. To gain insight into the functional genes that accompany these biological changes, RNA sequencing of the CAM at EID11 and EID15 was performed. Results showed that CAM maturation coincides with the overexpression of 4225 genes, including many genes encoding proteins involved in mineral metabolism, innate immunity, homeostasis, angiogenesis, reproduction, and regulation of hypoxia. Of these genes, some exhibit variability in expression depending on the chicken breed (broiler versus layer breeds). Besides the interest of these results for the poultry sector, the identification of new functional gene candidates opens additional research avenues in the field of developmental biology.
Collapse
Affiliation(s)
| | | | - Christelle Hennequet-Antier
- Université Paris-Saclay, INRAE, MaIAGE, 78350 Jouy-en-Josas, France; Université Paris-Saclay, INRAE, BioinfOmics, MIGALE bioinformatics facility, 78350 Jouy-en-Josas, France
| | - Maxwell Hincke
- Department of Innovation in Medical Education, Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; LE STUDIUM Research Consortium, Loire Valley Institute for Advanced Studies, Orléans-Tours, France
| | | |
Collapse
|
21
|
Oliinyk D, Eigenberger A, Felthaus O, Haerteis S, Prantl L. Chorioallantoic Membrane Assay at the Cross-Roads of Adipose-Tissue-Derived Stem Cell Research. Cells 2023; 12:cells12040592. [PMID: 36831259 PMCID: PMC9953848 DOI: 10.3390/cells12040592] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
With a history of more than 100 years of different applications in various scientific fields, the chicken chorioallantoic membrane (CAM) assay has proven itself to be an exceptional scientific model that meets the requirements of the replacement, reduction, and refinement principle (3R principle). As one of three extraembryonic avian membranes, the CAM is responsible for fetal respiration, metabolism, and protection. The model provides a unique constellation of immunological, vascular, and extracellular properties while being affordable and reliable at the same time. It can be utilized for research purposes in cancer biology, angiogenesis, virology, and toxicology and has recently been used for biochemistry, pharmaceutical research, and stem cell biology. Stem cells and, in particular, mesenchymal stem cells derived from adipose tissue (ADSCs) are emerging subjects for novel therapeutic strategies in the fields of tissue regeneration and personalized medicine. Because of their easy accessibility, differentiation profile, immunomodulatory properties, and cytokine repertoire, ADSCs have already been established for different preclinical applications in the files mentioned above. In this review, we aim to highlight and identify some of the cross-sections for the potential utilization of the CAM model for ADSC studies with a focus on wound healing and tissue engineering, as well as oncological research, e.g., sarcomas. Hereby, the focus lies on the combination of existing evidence and experience of such intersections with a potential utilization of the CAM model for further research on ADSCs.
Collapse
Affiliation(s)
- Dmytro Oliinyk
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
- Correspondence:
| | - Andreas Eigenberger
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Oliver Felthaus
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Silke Haerteis
- Institute for Molecular and Cellular Anatomy, Faculty for Biology and Preclinical Medicine, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
22
|
Chen L, Yuan M, Zhang X, Li Y, Feng Y, Yu J, Coudyzer W, Xie Y, Xu J, Li Y, Li Y, Ni Y. Exploration of Chick Embryo and Chorioallantoic Membrane on Imaging Navigated Platforms for Anticancer Pharmaceutical Evaluations. Technol Cancer Res Treat 2023; 22:15330338231206985. [PMID: 37844882 PMCID: PMC10585999 DOI: 10.1177/15330338231206985] [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: 07/28/2023] [Revised: 08/29/2023] [Accepted: 09/25/2023] [Indexed: 10/18/2023] Open
Abstract
Conforming to the current replace-reduce-refine 3Rs' guidelines in animal experiments, a series of explorative efforts have been made to set up operable biomedical imaging-guided platforms for qualitative and quantitative evaluations on pharmacological effects of tumor vascular-disrupting agents (VDAs), based on the chick embryos (CEs) with its chorioallantoic membrane (CAM), in this overview. The techniques and platforms have been hierarchically elaborated, from macroscopic to microscopic and from overall to specific aspects. A protocol of LED lamplight associated with a new deep-learning algorithm was consolidated to screen out weak CEs by using the CAM vasculature imaging. 3D magnetic resonance imaging (MRI) and laser speckle contrast imaging (LSCI) to monitor the evolution of CE and vascular changes in CAM are introduced. A LSCI-CAM platform for studying the effects of VDAs on normal and cancerous vasculature of CAM and possible molecular mechanisms has been demonstrated. Finally, practical challenges and future perspectives are highlighted. The aim of this article is to help peers in biomedical research to familiarize with the CAM platform and to optimize imaging protocols for the evaluation of vasoactive pharmaceuticals, especially anticancer vascular targeted therapy.
Collapse
Affiliation(s)
- Lei Chen
- KU Leuven, Biomedical Group, Leuven, Belgium
| | - Mingyuan Yuan
- Department of Radiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xinqi Zhang
- Airport Division, Tianjin Cancer Hospital, Tianjin, China
| | - Yongsheng Li
- Airport Division, Tianjin Cancer Hospital, Tianjin, China
| | - Yuanbo Feng
- KU Leuven, Biomedical Group, Leuven, Belgium
| | - Jie Yu
- KU Leuven, Biomedical Group, Leuven, Belgium
| | - Walter Coudyzer
- Department of Radiology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Yiyang Xie
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jiayue Xu
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yuzhen Li
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yue Li
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yicheng Ni
- KU Leuven, Biomedical Group, Leuven, Belgium
- Department of Radiology, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
23
|
Sharma DS, Wadhwa S, Gulati M, Kumar B, Chitranshi N, Gupta VK, Alrouji M, Alhajlah S, AlOmeir O, Vishwas S, Khursheed R, Saini S, Kumar A, Parveen SR, Gupta G, Zacconi F, Chellappan DK, Morris A, Loebenberg R, Dua K, Singh SK. Chitosan modified 5-fluorouracil nanostructured lipid carriers for treatment of diabetic retinopathy in rats: A new dimension to an anticancer drug. Int J Biol Macromol 2023; 224:810-830. [PMID: 36302483 DOI: 10.1016/j.ijbiomac.2022.10.168] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022]
Abstract
Diabetic retinopathy (DR) is one of the chronic complications of diabetes. It includes retinal blood vessels' damage. If untreated, it leads to loss of vision. The existing treatment strategies for DR are expensive, invasive, and need expertise during administration. Hence, there is a need to develop a non-invasive topical formulation that can penetrate deep to the posterior segment of retina and treat the damaged retinal vessels. In addition, it should also provide sustained release. In recent years, novel drug delivery systems (NDDS) have been explored for treating DR and found successful. In this study, chitosan (CS) modified 5-Fluorouracil Nanostructured Lipid Carriers (CS-5-FU-NLCs) were prepared by modified melt emulsification-ultrasonication method and optimized by Box-Behnken Design. The size, polydispersity index, zeta potential and entrapment efficiency of CS-5-FU-NLCs were 163.2 ± 2.3 nm, 0.28 ± 1.52, 21.4 ± 0.5 mV and 85.0 ± 0.2 %, respectively. The in vitro drug release and ex vivo permeation study confirmed higher and sustained drug release in CS-5-FU-NLCs as compared to 5-FU solution. HET-CAM Model ensured the non-irritant nature of CS-5-FU-NLCs. In vivo ocular studies of CS-5-FU-NLCs confirmed antiangiogenic effect of 5-FU by CAM model and diabetic retinopathy induced rat model, indicating successful delivery of 5-FU to the retina.
Collapse
Affiliation(s)
- Deep Shikha Sharma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Sheetu Wadhwa
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India.
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Bimlesh Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Nitin Chitranshi
- Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Australia
| | - Vivek Kumar Gupta
- Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Australia
| | - Mohammed Alrouji
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Sharif Alhajlah
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Othman AlOmeir
- Department of Pharmacy Practice, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Rubiya Khursheed
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Sumant Saini
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Ankit Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Shaik Rahana Parveen
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Flavia Zacconi
- Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile; Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Andrew Morris
- Swansea University Medical School, Swansea University, Singleton Park, Room 262, 1st Floor, Grove Building, Swansea, Wales SA2 8PP, UK
| | - Raimar Loebenberg
- University of Alberta, Faculty of Pharmacy and Pharmaceutical Sciences, Edmonton AB T6G2N8, Alberta, Canada
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia.
| |
Collapse
|
24
|
Butler K, Brinker CJ, Leong HS. Bridging the In Vitro to In Vivo gap: Using the Chick Embryo Model to Accelerate Nanoparticle Validation and Qualification for In Vivo studies. ACS NANO 2022; 16:19626-19650. [PMID: 36453753 PMCID: PMC9799072 DOI: 10.1021/acsnano.2c03990] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 10/17/2022] [Indexed: 06/17/2023]
Abstract
We postulate that nanoparticles (NPs) for use in therapeutic applications have largely not realized their clinical potential due to an overall inability to use in vitro results to predict NP performance in vivo. The avian embryo and associated chorioallantoic membrane (CAM) has emerged as an in vivo preclinical model that bridges the gap between in vitro and in vivo, enabling rapid screening of NP behavior under physiologically relevant conditions and providing a rapid, accessible, economical, and more ethical means of qualifying nanoparticles for in vivo use. The CAM is highly vascularized and mimics the diverging/converging vasculature of the liver, spleen, and lungs that serve as nanoparticle traps. Intravital imaging of fluorescently labeled NPs injected into the CAM vasculature enables immediate assessment and quantification of nano-bio interactions at the individual NP scale in any tissue of interest that is perfused with a microvasculature. In this review, we highlight how utilization of the avian embryo and its CAM as a preclinical model can be used to understand NP stability in blood and tissues, extravasation, biocompatibility, and NP distribution over time, thereby serving to identify a subset of NPs with the requisite stability and performance to introduce into rodent models and enabling the development of structure-property relationships and NP optimization without the sacrifice of large populations of mice or other rodents. We then review how the chicken embryo and CAM model systems have been used to accelerate the development of NP delivery and imaging agents by allowing direct visualization of targeted (active) and nontargeted (passive) NP binding, internalization, and cargo delivery to individual cells (of relevance for the treatment of leukemia and metastatic cancer) and cellular ensembles (e.g., cancer xenografts of interest for treatment or imaging of cancer tumors). We conclude by showcasing emerging techniques for the utilization of the CAM in future nano-bio studies.
Collapse
Affiliation(s)
- Kimberly
S. Butler
- Molecular
and Microbiology, Sandia National Laboratories, Albuquerque, New Mexico 87123, United States
| | - C. Jeffrey Brinker
- Department
of Chemical and Biological Engineering and the Comprehensive Cancer
Center, The University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Hon Sing Leong
- Department
of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto M5G 1L7, Canada
- Biological
Sciences Platform, Sunnybrook Hospital, Toronto M4N 3M5, Canada
| |
Collapse
|
25
|
de Oliveira NK, Ferraz EP, Rosin FCP, Correa L, Deboni MCZ. Poly-ε-caprolactone/poly(rotaxane) seeded with human dental pulp stem cells or osteoblasts promotes angiogenesis: a chorioallantoic membrane assay. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:2051-2066. [PMID: 35719115 DOI: 10.1080/09205063.2022.2091372] [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: 11/29/2021] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 06/15/2023]
Abstract
Biomaterials used for tissue regeneration should ideally provide a favorable environment for cell proliferation and differentiation. Angiogenesis is crucial for supplying oxygen and nutrients necessary for cellular survival at implantation sites. The aim of this study was to evaluate the overall angiogenesis response of a poly ε-caprolactone/poly (rotaxane) blend (poly-blend) carried by human dental pulp stem cells (hDPSCs) or osteoblasts (OB) seeded in the chorioallantoic membranes (CAM) of fertilized chicken eggs on embryonic day 7. They were classified into the following intervention groups: (a) poly(polymeric blend disks free of cells); (b) hDPSC seeded onto CAM; (c) poly/hDPSC (where hDPSCs were seeded onto poly-blend); (d) poly/OB (where osteoblasts were seeded onto poly); (e) OB (where hDPSCs differentiated into osteoblasts were seeded onto CAM); and (f) a negative control when a sterilized silicone ring free of cells or polymer was inserted into CAM. On embryonic day 14, the quantitative and qualitative characteristics of the blood vessels in the CAMs were analyzed macroscopically and microscopically. Macroscopic examination showed that the Poly/hDPSC samples exhibited an increased medium vessel density. Additionally, microscopic observations showed that the Poly/hDPSC group and poly alone resulted in a large lumen area of vascularization. Thus, poly ε-caprolactone/poly (rotaxane) did not impair angiogenesis. Furthermore, poly-blend carried by stem cells of dental pulp origin shows a better vasculogenic potential, which is essential for regenerative therapies.
Collapse
Affiliation(s)
| | - Emanuela Prado Ferraz
- Oral Surgery Department, School of Dentistry, University of São Paulo - FOUSP, São Paulo, SP, Brazil
| | | | - Luciana Correa
- Pathology Department, School of Dentistry, University of São Paulo - FOUSP, São Paulo, SP, Brazil
| | | |
Collapse
|
26
|
Advances in Biomaterials for Promoting Vascularization. CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-022-00217-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
27
|
Sreenivasamurthy SA, Akhter FF, Akhter A, Su Y, Zhu D. Cellular mechanisms of biodegradable zinc and magnesium materials on promoting angiogenesis. BIOMATERIALS ADVANCES 2022; 139:213023. [PMID: 35882117 DOI: 10.1016/j.bioadv.2022.213023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/15/2022] [Accepted: 07/05/2022] [Indexed: 06/15/2023]
Abstract
Biodegradable metals, zinc and magnesium, have been regarded as next-generation, biomedical implant materials to promote tissue repair and regeneration. These implants might also promote the vascularization of surrounding neotissue. Released metallic ions, Zn2+ and Mg2+, show promise in vitro to implement vessel growth by stimulating the expression of pro-angiogenic cytokines, yet there is little known regarding how cellular responses transcend to influence the tissue environment. This study serves to optimize angiogenic behavior using EA.hy926 endothelial cultures exposed to Zn2+ and Mg2+ gradients and observe the translation of these effects on blood vessel development via the in ovo chorioallantoic membrane (CAM) assay. Findings indicate that Zn2+ 10 μM and Mg2+ 10 mM instigate the most prominent effects using endothelial cultures via scratch wound and tube formation assays, yet higher concentrations at Zn2+ 50 μM and Mg2+ 50 mM encourage significant angiogenesis along the CAM. Immunoblotting results also conclude the presence and upregulation of cytokines involved in vessel growth. Optimizing the angiogenic potential of Zn2+ and Mg2+ separately sheds light to design future engineering constructs for promoting blood vessel development and successful assimilation between host and implant tissue.
Collapse
Affiliation(s)
- Sai A Sreenivasamurthy
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11790, United States
| | - Fnu Firoz Akhter
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11790, United States
| | - Asma Akhter
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11790, United States
| | - Yingchao Su
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11790, United States
| | - Donghui Zhu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11790, United States.
| |
Collapse
|
28
|
Cerium oxide decorated 5-fluorouracil loaded chitosan nanoparticles for treatment of hepatocellular carcinoma. Int J Biol Macromol 2022; 216:52-64. [PMID: 35750101 DOI: 10.1016/j.ijbiomac.2022.06.112] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 11/24/2022]
Abstract
Reactive oxygen species (ROS) play a crucial role in the mammalian system in both normal and pathological conditions. Hence, this work prepared and characterized the ROS responsive cerium oxide nanoparticles (CeO2 NPs) decorated 5-fluorouracil (5FU) loaded chitosan (CS) nanoparticles (CS-5FU NPs) for enhanced anticancer activity in hepatocellular carcinoma (HepG2 cells). CeO2 NPs decorated CS-5FU NPs were found to be spherical in shape and black dense aggregated particles sized 200 nm. The functional properties and cubic crystalline structure of CeO2 NPs were studied by Fourier-transform infrared spectroscopy (FTIR) and X-ray diffraction (XRD) analysis, respectively. Further, CS-5FU-CeO2 NPs attenuated the 2,2'-Azobis (2-methylpropionamidine) dihydrochloride (AAPH) induced ROS formation in mouse embryonic fibroblasts (NIH3T3 cells) while enhancing apoptotic cell death in HepG2 cells by controlled delivery of 5FU. Furthermore, CS-5FU-CeO2 NPs have not exhibited toxicity to red blood cells (RBCs) and chick chorioallantoic membrane (CAM). Hence, this work concluded that CeO2 NPs decorated CS-5FU NPs synergistically enhanced anticancer activity in HepG2 cells through the regulation of ROS.
Collapse
|
29
|
Chen L, Wang S, Feng Y, Yu J, Coudyzer W, Van Ongeval C, Geng L, Li Y, Ni Y. Development and characterization of a chick embryo chorioallantoic membrane (CAM) based platform for evaluation of vasoactive medications. Microvasc Res 2022; 142:104372. [PMID: 35483521 DOI: 10.1016/j.mvr.2022.104372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 12/13/2022]
Abstract
Among various anti-cancer therapies, tumor vascular disrupting agents (VDAs) play a crucial role, for which their off-targeting effects on normal vessels need also to be investigated. The purpose of this study was to set up an in-ovo platform that combines a laser speckle contrast imaging (LSCI) modality with chick embryo chorioallantoic membrane (CAM) to real-time monitor vascular diameters and perfusion without and with intravascular injection. Two eggshell windows for both observation or measurement and injection were opened. Dynamic blood perfusion images and corresponding statistic graphs were acquired by using a LSCI unit on CAMs from embryo date (ED) 9 to ED15. A dedicated fine needle catheter was made for slow intravascular administration over 30 min with simultaneous LSCI acquisition. To verify the connectivity between CAM vessels and the embryonic circulations in the egg, contrast-enhanced 3D micro computed tomography (μCT), 2D angiography and histology were executed. This platform was successfully established to acquire, quantify and demonstrate vascular and hemodynamic information from the CAM. Chick embryos even with air cell opened remained alive from ED9 to ED15. Through collecting LSCI derived CAM vascular diameter and perfusion parameters, ED12 was determined as the best time window for vasoactive drug studies. A reverse correlation between CAM vessel diameter and blood perfusion rate was found (p < 0.002). Intravascular infusion and simultaneous LSCI acquisition for 30 min in ovo proved feasible. Contrast-enhanced angiography and histomorphology could characterize the connectivity between CAM vasculature and embryonic circulation. This LSCI-CAM platform was proved effective for investigating the in-ovo hemodynamics, which paves the road for further preclinical research on vasoactive medications including VDAs.
Collapse
Affiliation(s)
- Lei Chen
- KU Leuven, Biomedical Group, Campus Gasthuisberg, Leuven 3000, Belgium.
| | - Shuncong Wang
- KU Leuven, Biomedical Group, Campus Gasthuisberg, Leuven 3000, Belgium.
| | - Yuanbo Feng
- KU Leuven, Biomedical Group, Campus Gasthuisberg, Leuven 3000, Belgium.
| | - Jie Yu
- KU Leuven, Biomedical Group, Campus Gasthuisberg, Leuven 3000, Belgium.
| | - Walter Coudyzer
- Department of Radiology, University Hospitals Leuven, KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | - Chantal Van Ongeval
- Department of Radiology, University Hospitals Leuven, KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | - Lei Geng
- School of Life Science, TianGong University, Tianjin, China.
| | - Yue Li
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| | - Yicheng Ni
- KU Leuven, Biomedical Group, Campus Gasthuisberg, Leuven 3000, Belgium.
| |
Collapse
|
30
|
Shokrani H, Shokrani A, Sajadi SM, Seidi F, Mashhadzadeh AH, Rabiee N, Saeb MR, Aminabhavi T, Webster TJ. Cell-Seeded Biomaterial Scaffolds: The Urgent Need for Unanswered Accelerated Angiogenesis. Int J Nanomedicine 2022; 17:1035-1068. [PMID: 35309965 PMCID: PMC8927652 DOI: 10.2147/ijn.s353062] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
One of the most arduous challenges in tissue engineering is neovascularization, without which there is a lack of nutrients delivered to a target tissue. Angiogenesis should be completed at an optimal density and within an appropriate period of time to prevent cell necrosis. Failure to meet this challenge brings about poor functionality for the tissue in comparison with the native tissue, extensively reducing cell viability. Prior studies devoted to angiogenesis have provided researchers with some biomaterial scaffolds and cell choices for angiogenesis. For example, while most current angiogenesis approaches require a variety of stimulatory factors ranging from biomechanical to biomolecular to cellular, some other promising stimulatory factors have been underdeveloped (such as electrical, topographical, and magnetic). When it comes to choosing biomaterial scaffolds in tissue engineering for angiogenesis, key traits rush to mind including biocompatibility, appropriate physical and mechanical properties (adhesion strength, shear stress, and malleability), as well as identifying the appropriate biomaterial in terms of stability and degradation profile, all of which may leave essential trace materials behind adversely influencing angiogenesis. Nevertheless, the selection of the best biomaterial and cells still remains an area of hot dispute as such previous studies have not sufficiently classified, integrated, or compared approaches. To address the aforementioned need, this review article summarizes a variety of natural and synthetic scaffolds including hydrogels that support angiogenesis. Furthermore, we review a variety of cell sources utilized for cell seeding and influential factors used for angiogenesis with a concentrated focus on biomechanical factors, with unique stimulatory factors. Lastly, we provide a bottom-to-up overview of angiogenic biomaterials and cell selection, highlighting parameters that need to be addressed in future studies.
Collapse
Affiliation(s)
- Hanieh Shokrani
- Department of Chemical Engineering, Sharif University of Technology, Tehran, Iran
| | - Amirhossein Shokrani
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - S Mohammad Sajadi
- Department of Nutrition, Cihan University-Erbil, Erbil, 625, Iraq
- Department of Phytochemistry, SRC, Soran University, Soran, KRG, 624, Iraq
- Correspondence: S Mohammad Sajadi; Navid Rabiee, Email ; ;
| | - Farzad Seidi
- Jiangsu Co–Innovation Center for Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing, 210037, People’s Republic of China
| | - Amin Hamed Mashhadzadeh
- Mechanical and Aerospace Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan, 010000, Kazakhstan
| | - Navid Rabiee
- Department of Physics, Sharif University of Technology, Tehran, Iran
- School of Engineering, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Tejraj Aminabhavi
- School of Advanced Sciences, KLE Technological University, Hubballi, Karnataka, 580 031, India
- Department of Chemistry, Karnatak University, Dharwad, 580 003, India
| | - Thomas J Webster
- School of Health Sciences and Biomedical Engineering, Hebei University, Tianjin, People’s Republic of China
- Center for Biomaterials, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
31
|
Diaz-Gomez L, Gonzalez-Prada I, Millan R, Da Silva-Candal A, Bugallo-Casal A, Campos F, Concheiro A, Alvarez-Lorenzo C. 3D printed carboxymethyl cellulose scaffolds for autologous growth factors delivery in wound healing. Carbohydr Polym 2022; 278:118924. [PMID: 34973742 DOI: 10.1016/j.carbpol.2021.118924] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/02/2021] [Accepted: 11/17/2021] [Indexed: 11/02/2022]
Abstract
This work aims to use carboxymethyl cellulose (CMC) as main structural and functional component of 3D printed scaffolds for healing of diabetic wounds. Differently from previous inks involving small contents in CMC, herein sterile (steam-heated) concentrated CMC solely dispersions (10-20%w/v) were screened regarding printability and fidelity properties. CMC (15%w/v)-citric acid inks showed excellent self-healing rheological properties and stability during storage. CMC scaffolds loaded with platelet rich plasma (PRP) sustained the release of relevant growth factors. CMC scaffolds both with and without PRP promoted angiogenesis in ovo, stem cell migration in vitro, and wound healing in a diabetic model in vivo. Transparent CMC scaffolds allowed direct monitoring of bilateral full-thickness wounds created in rat dorsum. CMC scaffolds facilitated re-epithelialization, granulation, and angiogenesis in full-thickness skin defects, and the performance was improved when combined with PRP. Overall, CMC is pointed out as outstanding component of active dressings for diabetic wounds.
Collapse
Affiliation(s)
- Luis Diaz-Gomez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Iago Gonzalez-Prada
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Rosendo Millan
- Centro de Biomedicina Experimental da USC (CEBEGA), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Andres Da Silva-Candal
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Neurovascular Diseases Laboratory, Neurology Service, University Hospital Complex of A Coruña, Biomedical Research Institute (INIBIC), 15706 A Coruña, Spain
| | - Ana Bugallo-Casal
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
32
|
Kis AM, Macasoi I, Paul C, Radulescu M, Buzatu R, Watz CG, Cheveresan A, Berceanu D, Pinzaru I, Dinu S, Manea A, Poenaru M, Borza C, Dehelean CA. Methotrexate and Cetuximab—Biological Impact on Non-Tumorigenic Models: In Vitro and In Ovo Assessments. Medicina (B Aires) 2022; 58:medicina58020167. [PMID: 35208492 PMCID: PMC8877801 DOI: 10.3390/medicina58020167] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/20/2022] Open
Abstract
Background Objectives: The neoplastic process remains a major health problem facing humanity. Although there are currently different therapeutic options, they raise a multitude of shortcomings related to the toxic effects associated with their administration. Methotrexate (Met) and Cetuximab (Cet) are two basic chemotherapeutics used in cancer practice, but notwithstanding despite many years of use, the mechanisms by which the multitude of side-effects occur are not yet fully understood. Thus, the present study focused on the in vitro and in ovo evaluation of the associated toxic mechanisms on keratinocytes, keys cells in the wound healing process. Materials and Methods: The two chemotherapeutics were tested in eight different concentrations to evaluate keratinocytes viability, the anti-migratory effect, and the influence on the expression of markers involved in the production of cell apoptosis. In addition, the potential irritating effect on the vascular plexus were highlighted by applying the in ovo method, chick chorioallantoic membrane (HET-CAM). Results: The results revealed that Met induced decreased cell viability as well as increased expression of pro-apoptotic genes. In the vascular plexus of the chorioallantoic membrane, Met caused vascular irritation accompanied by capillary hemorrhage and vascular stasis. Conclusions: Summarizing, Cet presents a safer toxicological profile, compared to Met, based on the results obtained from both in vitro (cell viability, wound healing, RT-PCR assays), and in ovo (HET-CAM assay) techniques.
Collapse
Affiliation(s)
- Andreea M. Kis
- Department of ENT, “Victor Babeş” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania; (A.M.K.); (M.P.)
| | - Ioana Macasoi
- Departament of Toxicology and Drug Industry, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania; (I.M.); (I.P.); (C.A.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Corina Paul
- Department of Pediatrics, “Victor Babeş” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania;
| | - Matilda Radulescu
- Department of Microbiology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
- Correspondence: (M.R.); (R.B.)
| | - Roxana Buzatu
- Department of Dental Aesthetics, Faculty of Dental Medicine, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 9 No., Revolutiei Bv., 300041 Timişoara, Romania
- Correspondence: (M.R.); (R.B.)
| | - Claudia G. Watz
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
- Departament of Pharmaceutical Physics, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
| | - Adelina Cheveresan
- Department of Pharmacology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Delia Berceanu
- Department of Microbiology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Iulia Pinzaru
- Departament of Toxicology and Drug Industry, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania; (I.M.); (I.P.); (C.A.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Stefania Dinu
- Department of Pedodontics, Faculty of Dental Medicine, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 9 No., Revolutiei Bv., 300041 Timişoara, Romania;
- Pediatric Dentistry Research Center, Faculty of Dental Medicine, “Victor Babeş” University of Medicine and Pharmacy Timisoara, 9 No., Revolutiei Bv., 300041 Timişoara, Romania
| | - Aniko Manea
- Department of Neonatology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Marioara Poenaru
- Department of ENT, “Victor Babeş” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania; (A.M.K.); (M.P.)
| | - Claudia Borza
- Department of Pathophysiology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Cristina A. Dehelean
- Departament of Toxicology and Drug Industry, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania; (I.M.); (I.P.); (C.A.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| |
Collapse
|
33
|
Vascular Effects of Low-Dose ACE2 Inhibitor MLN-4760—Benefit or Detriment in Essential Hypertension? Biomedicines 2021; 10:biomedicines10010038. [PMID: 35052717 PMCID: PMC8773407 DOI: 10.3390/biomedicines10010038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/17/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infects host cells through angiotensin-converting enzyme 2 (ACE2). Concurrently, the product of ACE2 action, angiotensin 1–7 (Ang 1–7), binds to Mas receptors within the cardiovascular system and provides protective effects. Therefore, it is crucial to reveal the role of ACE2 inhibition, especially within pre-existing cardiovascular pathologies. In our study, we imitated the action of SARS-CoV-2 in organisms using the low dose of the ACE2 inhibitor MLN-4760 with the aim of investigating to what degree ACE2 inhibition is detrimental to the cardiovascular system of spontaneously hypertensive rats (SHRs), which represent a model of human essential hypertension. Our study revealed the complex action of MLN-4760 in SHRs. On the one hand, we found that MLN-4760 had (1) (pro)obesogenic effects that negatively correlated with alternative renin-angiotensin system activity and Ang 1–7 in plasma, (2) negative effects on ACE1 inhibitor (captopril) action, (3) detrimental effects on the small arteries function and (4) anti-angiogenic effect in the model of chick chorioallantoic membrane. On the other hand, MLN-4760 induced compensatory mechanisms involving strengthened Mas receptor-, nitric oxide- and hydrogen sulfide-mediated signal transduction in the aorta, which was associated with unchanged blood pressure, suggesting beneficial action of MLN-4760 when administered at a low dose.
Collapse
|
34
|
Cilostazol induces angiogenesis and regulates oxidative stress in a dose-dependent manner: A chorioallantoic membrane study. TURKISH JOURNAL OF THORACIC AND CARDIOVASCULAR SURGERY 2021; 29:449-456. [PMID: 35096441 PMCID: PMC8762914 DOI: 10.5606/tgkdc.dergisi.2021.22212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/25/2021] [Indexed: 11/21/2022]
Abstract
Background
In this study, we aimed to investigate the effects of cilostazol on angiogenesis and oxidative stress using the chorioallantoic membrane model.
Methods
In this experimental study, the Ross 308 chick embryos were used. The negative control group (n=10) received no intervention. The positive control group (n=10) consisted of eggs treated with epidermal growth factor for inducing angiogenesis. Three cilostazol groups were designed with 10-7 (n=10), 10-6 (n=10), and 10-5 (n=10) M concentrations. Each egg was punctured on the sixth day of incubation, and drug pellets were introduced to the positive control and drug groups at the prespecified doses. Vascular development was evaluated on the eighth day of application. The total oxidant status, total antioxidant capacity, and oxidative stress index levels were determined from albumen liquids obtained with a syringe before and after drug application.
Results
Lower oxidative stress index levels were obtained from the positive control and cilostazol groups compared to the negative control albumens (p=0.001). The increments in vascular junctions and newly developed vascular nodules were evaluated in drug-free and drug-applied chorioallantoic membranes. The highest activity was obtained in the 10-7 M concentration cilostazol group. An increased angiogenic activity was detected in all drug groups in each concentration compared to the negative control group (p=0.001). Angiogenic activity was similar in all the cilostazol-treated groups (p=0.43).
Conclusion
Cilostazol has a positive stimulant effect on angiogenesis and it seems to suppress oxidative stress during embryonic growth. Cilostazol exerts these effects significantly and similarly at different doses.
Collapse
|
35
|
Wiesmann N, Mendler S, Buhr CR, Ritz U, Kämmerer PW, Brieger J. Zinc Oxide Nanoparticles Exhibit Favorable Properties to Promote Tissue Integration of Biomaterials. Biomedicines 2021; 9:biomedicines9101462. [PMID: 34680579 PMCID: PMC8533365 DOI: 10.3390/biomedicines9101462] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022] Open
Abstract
Due to the demographic change, medicine faces a growing demand for tissue engineering solutions and implants. Often, satisfying tissue regeneration is difficult to achieve especially when co-morbidities hamper the healing process. As a novel strategy, we propose the incorporation of zinc oxide nanoparticles (ZnO NPs) into biomaterials to improve tissue regeneration. Due to their wide range of biocompatibility and their antibacterial properties, ZnO NPs are already discussed for different medical applications. As there are versatile possibilities of modifying their form, size, and function, they are becoming increasingly attractive for tissue engineering. In our study, in addition to antibacterial effects of ZnO NPs, we show for the first time that ZnO NPs can foster the metabolic activity of fibroblasts as well as endothelial cells, both cell types being crucial for successful implant integration. With the gelatin sponge method performed on the chicken embryo’s chorioallantoic membrane (CAM), we furthermore confirmed the high biocompatibility of ZnO NPs. In summary, we found ZnO NPs to have very favorable properties for the modification of biomaterials. Here, incorporation of ZnO NPs could help to guide the tissue reaction and promote complication-free healing.
Collapse
Affiliation(s)
- Nadine Wiesmann
- Department of Otorhinolaryngology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (S.M.); (C.R.B.); (J.B.)
- Department of Oral- and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany;
- Correspondence: ; Tel.: +49-6131-17-4034
| | - Simone Mendler
- Department of Otorhinolaryngology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (S.M.); (C.R.B.); (J.B.)
| | - Christoph R. Buhr
- Department of Otorhinolaryngology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (S.M.); (C.R.B.); (J.B.)
| | - Ulrike Ritz
- Department of Orthopedics and Traumatology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany;
| | - Peer W. Kämmerer
- Department of Oral- and Maxillofacial Surgery, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany;
| | - Juergen Brieger
- Department of Otorhinolaryngology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (S.M.); (C.R.B.); (J.B.)
| |
Collapse
|
36
|
Kazakova O, Șoica C, Babaev M, Petrova A, Khusnutdinova E, Poptsov A, Macașoi I, Drăghici G, Avram Ș, Vlaia L, Mioc A, Mioc M, Dehelean C, Voicu A. 3-Pyridinylidene Derivatives of Chemically Modified Lupane and Ursane Triterpenes as Promising Anticancer Agents by Targeting Apoptosis. Int J Mol Sci 2021; 22:ijms221910695. [PMID: 34639035 PMCID: PMC8509773 DOI: 10.3390/ijms221910695] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022] Open
Abstract
Cancer persists as a global challenge due to the extent to which conventional anticancer therapies pose high risks counterbalanced with their therapeutic benefit. Naturally occurring substances stand as an important safer alternative source for anticancer drug development. In the current study, a series of modified lupane and ursane derivatives was subjected to in vitro screening on the NCI-60 cancer cell line panel. Compounds 6 and 7 have been identified as highly active with GI50 values ranging from 0.03 µM to 5.9 µM (compound 6) and 0.18–1.53 µM (compound 7). Thus, these two compounds were further assessed in detail in order to identify a possible antiproliferative mechanism of action. DAPI (4′,6-diamidino-2-phenylindole) staining revealed that both compounds induced nuclei condensation and overall cell morphological changes consistent with apoptotic cell death. rtPCR analysis showed that both compounds induced upregulation of proapoptotic Bak and Bad genes while downregulating Bcl-XL and Bcl-2 antiapoptotic genes. Molecular docking analysis revealed that both compounds exhibited high scores for Bcl-XL inhibition, while compound 7 showed higher in silico Bcl-XL inhibition potential as compared to the native inhibitor ATB-737, suggesting that compounds may induce apoptotic cell death through targeted antiapoptotic protein inhibition, as well.
Collapse
Affiliation(s)
- Oxana Kazakova
- Ufa Institute of Chemistry UFRC, Russian Academy of Science RAS, pr. Oktyabrya 71, 450054 Ufa, Russia; (M.B.); (A.P.); (E.K.); (A.P.)
- Correspondence: (O.K.); (M.M.)
| | - Codruța Șoica
- Formulation and Technology of Drugs Research Center, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.Ș.); (I.M.); (G.D.); (Ș.A.); (L.V.); (A.M.); (C.D.)
- Research Centre Pharmacotoxicol Evaluat, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Marat Babaev
- Ufa Institute of Chemistry UFRC, Russian Academy of Science RAS, pr. Oktyabrya 71, 450054 Ufa, Russia; (M.B.); (A.P.); (E.K.); (A.P.)
| | - Anastasiya Petrova
- Ufa Institute of Chemistry UFRC, Russian Academy of Science RAS, pr. Oktyabrya 71, 450054 Ufa, Russia; (M.B.); (A.P.); (E.K.); (A.P.)
| | - Elmira Khusnutdinova
- Ufa Institute of Chemistry UFRC, Russian Academy of Science RAS, pr. Oktyabrya 71, 450054 Ufa, Russia; (M.B.); (A.P.); (E.K.); (A.P.)
| | - Alexander Poptsov
- Ufa Institute of Chemistry UFRC, Russian Academy of Science RAS, pr. Oktyabrya 71, 450054 Ufa, Russia; (M.B.); (A.P.); (E.K.); (A.P.)
| | - Ioana Macașoi
- Formulation and Technology of Drugs Research Center, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.Ș.); (I.M.); (G.D.); (Ș.A.); (L.V.); (A.M.); (C.D.)
- Research Centre Pharmacotoxicol Evaluat, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - George Drăghici
- Formulation and Technology of Drugs Research Center, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.Ș.); (I.M.); (G.D.); (Ș.A.); (L.V.); (A.M.); (C.D.)
- Research Centre Pharmacotoxicol Evaluat, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Ștefana Avram
- Formulation and Technology of Drugs Research Center, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.Ș.); (I.M.); (G.D.); (Ș.A.); (L.V.); (A.M.); (C.D.)
- Research Centre Pharmacotoxicol Evaluat, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Lavinia Vlaia
- Formulation and Technology of Drugs Research Center, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.Ș.); (I.M.); (G.D.); (Ș.A.); (L.V.); (A.M.); (C.D.)
- Formulation and Technology of Drugs Research Center, “Victor Babeș” University of Medicine and Pharmacy, Faculty of Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Alexandra Mioc
- Formulation and Technology of Drugs Research Center, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.Ș.); (I.M.); (G.D.); (Ș.A.); (L.V.); (A.M.); (C.D.)
- Research Centre Pharmacotoxicol Evaluat, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Marius Mioc
- Formulation and Technology of Drugs Research Center, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.Ș.); (I.M.); (G.D.); (Ș.A.); (L.V.); (A.M.); (C.D.)
- Research Centre Pharmacotoxicol Evaluat, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania
- Correspondence: (O.K.); (M.M.)
| | - Cristina Dehelean
- Formulation and Technology of Drugs Research Center, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.Ș.); (I.M.); (G.D.); (Ș.A.); (L.V.); (A.M.); (C.D.)
- Research Centre Pharmacotoxicol Evaluat, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Adrian Voicu
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania;
| |
Collapse
|
37
|
Grambow E, Sorg H, Sorg CGG, Strüder D. Experimental Models to Study Skin Wound Healing with a Focus on Angiogenesis. Med Sci (Basel) 2021; 9:medsci9030055. [PMID: 34449673 PMCID: PMC8395822 DOI: 10.3390/medsci9030055] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022] Open
Abstract
A large number of models are now available for the investigation of skin wound healing. These can be used to study the processes that take place in a phase-specific manner under both physiological and pathological conditions. Most models focus on wound closure, which is a crucial parameter for wound healing. However, vascular supply plays an equally important role and corresponding models for selective or parallel investigation of microcirculation regeneration and angiogenesis are also described. In this review article, we therefore focus on the different levels of investigation of skin wound healing (in vivo to in virtuo) and the investigation of angiogenesis and its parameters.
Collapse
Affiliation(s)
- Eberhard Grambow
- Department of General, Visceral, Thoracic, Vascular and Transplantation Surgery, Rostock University Medical Center, 18057 Rostock, Germany
- Correspondence:
| | - Heiko Sorg
- Department of Health, University of Witten/Herdecke, Alfred-Herrhausen-Str. 50, 58455 Witten, Germany;
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Klinikum Westfalen, Am Knappschaftskrankenhaus 1, 44309 Dortmund, Germany
| | - Christian G. G. Sorg
- Chair of Management and Innovation in Health Care, Department of Management and Entrepreneurship, Faculty of Management, Economics and Society, Witten/Herdecke University, Alfred-Herrhausen-Straße 50, 58455 Witten, Germany;
| | - Daniel Strüder
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery “Otto Körner”, Rostock University Medical Center, 18057 Rostock, Germany;
| |
Collapse
|
38
|
Comparison of Quantification of Target-Specific Accumulation of [ 18F]F-siPSMA-14 in the HET-CAM Model and in Mice Using PET/MRI. Cancers (Basel) 2021; 13:cancers13164007. [PMID: 34439163 PMCID: PMC8393674 DOI: 10.3390/cancers13164007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 01/07/2023] Open
Abstract
Simple Summary Animal studies are essential for the development of new radiopharmaceuticals to determine specific accumulation and biodistribution. Alternative models, such as the HET-CAM model, offer the possibility of reducing animal experiments in accordance with the 3Rs principles. Accurate quantification of tumor accumulation of a PSMA-specific ligand in the HET-CAM model and comparison with corresponding animal experiments was performed using the imaging modalities PET and MRI. It was demonstrated that the HET-CAM model leads to comparable results and is suitable as an alternative to animal experiments for the initial assessment of target-specific binding of novel radiopharmaceuticals. However, as evaluation of biodistribution in ovo is still limited, further animal experiments with promising compounds are mandatory. Abstract Assessment of biodistribution and specific tumor accumulation is essential for the development of new radiopharmaceuticals and requires animal experiments. The HET-CAM (hens-egg test—chorioallantoic membrane) model can be used in combination with the non-invasive imaging modalities PET and MRI for pre-selection during radiopharmaceutical development to reduce the number of animal experiments required. Critical to the acceptance of this model is the demonstration of the quantifiability and reproducibility of these data compared to the standard animal model. Tumor accumulation and biodistribution of the PSMA-specific radiotracer [18F]F-siPSMA-14 was analyzed in the chick embryo and in an immunodeficient mouse model. Evaluation was based on MRI and PET data in both models. γ-counter measurements and histopathological analyses complemented these data. PSMA-specific accumulation of [18F]F-siPSMA-14 was successfully demonstrated in the HET-CAM model, similar to the results obtained by mouse model studies. The combination of MR and PET imaging allowed precise quantification of peptide accumulation, initial assessment of biodistribution, and accurate determination of tumor volume. Thus, the use of the HET-CAM model is suitable for the pre-selection of new radiopharmaceuticals and potentially reduces animal testing in line with the 3Rs principles of animal welfare.
Collapse
|
39
|
Luptakova L, Dvorcakova S, Demcisakova Z, Belbahri L, Holovska K, Petrovova E. Dimethyl Sulfoxide: Morphological, Histological, and Molecular View on Developing Chicken Liver. TOXICS 2021; 9:toxics9030055. [PMID: 33809222 PMCID: PMC8001493 DOI: 10.3390/toxics9030055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/28/2021] [Accepted: 03/09/2021] [Indexed: 01/04/2023]
Abstract
Dimethyl sulfoxide (DMSO) is widely used as a solvent for small hydrophobic drug molecules. However, the safe volume allowing to avoid its embryotoxic effect has been poorly studied. In this study, we documented the effects of dimethyl sulfoxide (DMSO) in the developing chicken embryo at morphological, histological, and molecular levels. We focused on the developing chicken liver as the main organ involved in the process of detoxification. In our study, 100% DMSO was administered subgerminally onto the eggshell membrane (membrana papyracea) at various volumes (5, 10, 15, 20, 25, 30, 35, and 50 µL) on 4th embryonic day (ED). We focused on histopathological alterations of the liver structure, and noticed the overall impact of DMSO on developing chicken embryos (embryotoxicity, malformation). At the molecular level, we studied cytochrome P450 complex (CYP) isoform's activities in relation to changes of CYP1A5, CYP3A37, and CYP3A80 gene expression. Total embryotoxicity after application of different doses of DMSO on ED 4, and the embryo lethality increased with increasing DMSO amounts. Overall mortality after DMSO administration ranged below 33%. Mortality was increased with higher amounts of DMSO, mainly from 20 µL. The highest mortality was observed for the highest dose of DMSO over 35 µL. The results also showed a decrease in body weight with increased application volumes of DMSO. At the histological level, we observed mainly the presence of lipid droplets and dilated bile canaliculi and sinusoids in samples over the administration of 25 µL of DMSO. While these findings were not statistically significant, DMSO treatment caused a significant different up-regulation of mRNA expression in all studied genes. For CYP1A5, CYP3A37, and CYP3A80 DMSO volumes needed were 15 µL, 10 µL, and 20 µL, respectively. A significant down-regulation of all studied CYP isoform was detected after application of a DMSO dose of 5 µL. Regarding the morphological results, we can assume that the highest safe dose of DMSO without affecting chicken embryo development and its liver is up to 10 µL. This conclusion is corroborated with the presence of number of malformations and body weight reduction, which correlates with histological findings. Moreover, the gene expression results showed that even the lowest administered DMSO volume could affect hepatocytes at the molecular level causing down-regulation of cytochrome P450 complex (CYP1A5, CYP3A37, CYP3A80).
Collapse
Affiliation(s)
- Lenka Luptakova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia;
- Correspondence: ; Tel.: +421-918-919-686
| | - Simona Dvorcakova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia;
| | - Zuzana Demcisakova
- Department of Morphological Disciplines, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (Z.D.); (K.H.); (E.P.)
| | - Lassaad Belbahri
- Laboratory of Soil Biodiversity, Department of Biology, University of Neuchatel, 2000 Neuchatel, Switzerland;
| | - Katarina Holovska
- Department of Morphological Disciplines, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (Z.D.); (K.H.); (E.P.)
| | - Eva Petrovova
- Department of Morphological Disciplines, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (Z.D.); (K.H.); (E.P.)
| |
Collapse
|
40
|
3D-microtissue derived secretome as a cell-free approach for enhanced mineralization of scaffolds in the chorioallantoic membrane model. Sci Rep 2021; 11:5418. [PMID: 33686145 PMCID: PMC7940489 DOI: 10.1038/s41598-021-84123-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
Bone regeneration is a complex process and the clinical translation of tissue engineered constructs (TECs) remains a challenge. The combination of biomaterials and mesenchymal stem cells (MSCs) may enhance the healing process through paracrine effects. Here, we investigated the influence of cell format in combination with a collagen scaffold on key factors in bone healing process, such as mineralization, cell infiltration, vascularization, and ECM production. MSCs as single cells (2D-SCs), assembled into microtissues (3D-MTs) or their corresponding secretomes were combined with a collagen scaffold and incubated on the chicken embryo chorioallantoic membrane (CAM) for 7 days. A comprehensive quantitative analysis was performed on a cellular level by histology and by microcomputed tomography (microCT). In all experimental groups, accumulation of collagen and glycosaminoglycan within the scaffold was observed over time. A pronounced cell infiltration and vascularization from the interface to the surface region of the CAM was detected. The 3D-MT secretome showed a significant mineralization of the biomaterial using microCT compared to all other conditions. Furthermore, it revealed a homogeneous distribution pattern of mineralization deposits in contrast to the cell-based scaffolds, where mineralization was only at the surface. Therefore, the secretome of MSCs assembled into 3D-MTs may represent an interesting therapeutic strategy for a next-generation bone healing concept.
Collapse
|
41
|
Utilisation of Chick Embryo Chorioallantoic Membrane as a Model Platform for Imaging-Navigated Biomedical Research. Cells 2021; 10:cells10020463. [PMID: 33671534 PMCID: PMC7926796 DOI: 10.3390/cells10020463] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/15/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
The fertilised chick egg and particularly its chorioallantoic membrane (CAM) have drawn continuing interest in biomedicine and bioengineering fields, especially for research on vascular study, cancer, drug screening and development, cell factors, stem cells, etc. This literature review systemically introduces the CAM's structural evolution, functions, vascular features and the circulation system, and cell regulatory factors. It also presents the major and updated applications of the CAM in assays for pharmacokinetics and biodistribution, drug efficacy and toxicology testing/screening in preclinical pharmacological research. The time course of CAM applications for different assays and their advantages and limitations are summarised. Among these applications, two aspects are emphasised: (1) potential utility of the CAM for preclinical studies on vascular-disrupting agents (VDAs), promising for anti-cancer vascular-targeted therapy, and (2) modern imaging technologies, including modalities and their applications for real-time visualisation, monitoring and evaluation of the changes in CAM vasculature as well as the interactions occurring after introducing the tested medical, pharmaceutical and biological agents into the system. The aim of this article is to help those working in the biomedical field to familiarise themselves with the chick embryo CAM as an alternative platform and to utilise it to design and optimise experimental settings for their specific research topics.
Collapse
|
42
|
Characterization of Properties, In Vitro and In Vivo Evaluation of Calcium Phosphate/Amino Acid Cements for Treatment of Osteochondral Defects. MATERIALS 2021; 14:ma14020436. [PMID: 33477289 PMCID: PMC7830446 DOI: 10.3390/ma14020436] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 12/13/2022]
Abstract
Novel calcium phosphate cements containing a mixture of four amino acids, glycine, proline, hydroxyproline and either lysine or arginine (CAL, CAK) were characterized and used for treatment of artificial osteochondral defects in knee. It was hypothesized that an enhanced concentration of extracellular collagen amino acids (in complex mixture), in connection with bone cement in defect sites, would support the healing of osteochondral defects with successful formation of hyaline cartilage and subchondral bone. Calcium phosphate cement mixtures were prepared by in situ reaction in a planetary ball mill at aseptic conditions and characterized. It was verified that about 30–60% of amino acids remained adsorbed on hydroxyapatite particles in cements and the addition of amino acids caused around 60% reduction in compressive strength and refinement of hydroxyapatite particles in their microstructure. The significant over-expression of osteogenic genes after the culture of osteoblasts was demonstrated in the cement extracts containing lysine and compared with other cements. The cement pastes were inserted into artificial osteochondral defects in the medial femoral condyle of pigs and, after 3 months post-surgery, tissues were analyzed macroscopically, histologically, immunohistochemically using MRI and X-ray methods. Analysis clearly showed the excellent healing process of artificial osteochondral defects in pigs after treatment with CAL and CAK cements without any inflammation, as well as formation of subchondral bone and hyaline cartilage morphologically and structurally identical to the original tissues. Good integration of the hyaline neocartilage with the surrounding tissue, as well as perfect interconnection between the neocartilage and new subchondral bone tissue, was demonstrated. Tissues were stable after 12 months’ healing.
Collapse
|
43
|
Heimes D, Wiesmann N, Eckrich J, Brieger J, Mattyasovszky S, Proff P, Weber M, Deschner J, Al-Nawas B, Kämmerer PW. In Vivo Modulation of Angiogenesis and Immune Response on a Collagen Matrix via Extracorporeal Shockwaves. Int J Mol Sci 2020; 21:ijms21207574. [PMID: 33066403 PMCID: PMC7589066 DOI: 10.3390/ijms21207574] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/02/2020] [Accepted: 10/11/2020] [Indexed: 12/12/2022] Open
Abstract
The effective management of tissue integration and immunological responses to transplants decisively co-determines the success of soft and hard tissue reconstruction. The aim of this in vivo study was to evaluate the eligibility of extracorporeal shock wave therapy (ESWT) with respect to its ability to modulate angiogenesis and immune response to a collagen matrix (CM) for tissue engineering in the chorioallantoic membrane (CAM) assay, which is performed with fertilized chicken eggs. CM were placed on the CAM on embryonic development day (EDD) 7; at EDD-10, ESWT was conducted at 0.12 mJ/mm2 with 500 impulses each. One and four days later, angiogenesis represented by vascularized area, vessel density, and vessel junctions as well as HIF-1α and VEGF gene expression were evaluated. Furthermore, immune response (iNOS2, MMP-9, and MMP-13 via qPCR) was assessed and compared between ESWT- and non-ESWT-groups. At EDD-14, the vascularized area (+115% vs. +26%) and the increase in vessel junctions (+751% vs. +363%) were significantly higher in the ESWT-group. ESWT significantly increased MMP-9 gene expression at EDD-11 and significantly decreased MMP-13 gene expression at EDD-14 as compared to the controls. Using the CAM assay, an enhanced angiogenesis and neovascularization in CM after ESWT were observed. Furthermore, ESWT could reduce the inflammatory activity after a latency of four days.
Collapse
Affiliation(s)
- Diana Heimes
- Department of Oral- and Maxillofacial and Plastic Surgery, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany; (N.W.); (B.A.-N.); (P.W.K.)
- Correspondence: ; Tel.: +49-6131-17-5086
| | - Nadine Wiesmann
- Department of Oral- and Maxillofacial and Plastic Surgery, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany; (N.W.); (B.A.-N.); (P.W.K.)
- Molecular Tumor Biology, Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center of the Johannes Gutenberg University, Langenbeckstraße 1, 55131 Mainz, Germany; (J.E.); (J.B.)
| | - Jonas Eckrich
- Molecular Tumor Biology, Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center of the Johannes Gutenberg University, Langenbeckstraße 1, 55131 Mainz, Germany; (J.E.); (J.B.)
| | - Juergen Brieger
- Molecular Tumor Biology, Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center of the Johannes Gutenberg University, Langenbeckstraße 1, 55131 Mainz, Germany; (J.E.); (J.B.)
| | - Stefan Mattyasovszky
- Department of Orthopedics and Traumatology, University Medical Center of the Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
| | - Peter Proff
- Department of Orthodontics, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany;
| | - Manuel Weber
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - James Deschner
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Augustusplatz 2, 55131 Mainz, Germany;
| | - Bilal Al-Nawas
- Department of Oral- and Maxillofacial and Plastic Surgery, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany; (N.W.); (B.A.-N.); (P.W.K.)
| | - Peer W. Kämmerer
- Department of Oral- and Maxillofacial and Plastic Surgery, University Medical Center Mainz, Augustusplatz 2, 55131 Mainz, Germany; (N.W.); (B.A.-N.); (P.W.K.)
| |
Collapse
|