1
|
Simon L, Constanzo J, Terraza-Aguirre C, Ibn Elfekih Z, Berthelot J, Benkhaled BT, Haute T, Pednekar K, Clark K, Emerson SJ, Atis S, Benedetti C, Langlois S, Marquant A, Prakash J, Wang A, Devoisselle JM, Montier T, Djouad F, Pouget JP, Lapinte V, Morille M. Surface modification of extracellular vesicles with polyoxazolines to enhance their plasma stability and tumor accumulation. Biomaterials 2025; 313:122748. [PMID: 39180918 DOI: 10.1016/j.biomaterials.2024.122748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
Extracellular vesicles (EVs) are future promising therapeutics, but their instability in vivo after administration remains an important barrier to their further development. Many groups evaluated EV surface modification strategies to add a targeting group with the aim of controlling EV biodistribution. Conversely, fewer groups focused on their stabilization to obtain "stealth" allogenic EVs. Modulating their stabilization and biodistribution is an essential prerequisite for their development as nano-therapeutics. Here, we explored polyoxazolines with lipid anchors association to the EV membrane (POxylation as an alternative to PEGylation) to stabilize EVs in plasma and control their biodistribution, while preserving their native properties. We found that this modification maintained and seemed to potentiate the immunomodulatory properties of EVs derived from mesenchymal stem/stromal cells (MSC). Using a radiolabeling protocol to track EVs at a therapeutically relevant concentration in vivo, we demonstrated that POxylation is a promising option to stabilize EVs in plasma because it increased EV half-life by 6 fold at 6 h post-injection. Moreover, EV accumulation in tumors was higher after POxylation than after PEGylation.
Collapse
Affiliation(s)
- L Simon
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Constanzo
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | | | - Z Ibn Elfekih
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Berthelot
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - B T Benkhaled
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - T Haute
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - K Pednekar
- Department of Advanced Organ Bioengineering and Therapeutics, Engineered Therapeutics Section, Technical Medical Centre, University of Twente, 7500 AE, Enschede, the Netherlands
| | - K Clark
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - S J Emerson
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - S Atis
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | - C Benedetti
- Montpellier Ressources Imagerie, BioCampus, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - S Langlois
- Montpellier Ressources Imagerie, BioCampus, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - A Marquant
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Prakash
- Department of Advanced Organ Bioengineering and Therapeutics, Engineered Therapeutics Section, Technical Medical Centre, University of Twente, 7500 AE, Enschede, the Netherlands
| | - A Wang
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - J M Devoisselle
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - T Montier
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France; CHU de Brest, Service de Génétique Médicale et de Biologie de La Reproduction, Centre de Référence des Maladies Rares Maladies Neuromusculaires, 29200, Brest, France
| | - F Djouad
- IRMB, University of Montpellier, INSERM, 34295, Montpellier, France; Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, 34095, Montpellier, France
| | - J P Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | - V Lapinte
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - Marie Morille
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France; Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
2
|
Wang C, Yao H, Shi J, Zhang Z, Cong B, Wu Z, Shang X, Hu X, Yang J, Sun H, Gu Z, Cheng G, Chong H, Wang DA, Zhao Y. Injectable tissue-engineered human cartilage matrix composite fibrin glue for regeneration of articular cartilage defects. BIOMATERIALS ADVANCES 2025; 167:214095. [PMID: 39504587 DOI: 10.1016/j.bioadv.2024.214095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/20/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024]
Abstract
Due to the lack of blood vessels and nerves, the ability of cartilage to repair itself is limited, and the injury of articular cartilage urgently needs effective treatment. Currently, the limitation of clinical repair for cartilage defects is that it is difficult to form pure hyaline cartilage repair, and the source of cartilage tissue and cells is limited. To obtain high-purity regenerated hyaline cartilage, we proposed to construct an injectable hydrogel precursor by using human living hyaline cartilage graft (hLhCG) secreted by human chondrocytes as the dispersed phase and fibrinogen solution as the continuous phase, by double injection with thrombin, three-dimensional network hydrogel structure was formed under the action of thrombin to repair joint defects. The component phenotypes of hLhCG and biomechanical properties of composite gel scaffolds were verified. After 12 weeks of injection of the mixed phase at the defect site, the regenerated tissues are similar in composition to adjacent natural tissues and exhibit similar biomechanical properties. The phenotype of regenerated cartilage was verified, confirming the successful regeneration of hyaline cartilage.
Collapse
Affiliation(s)
- Chirun Wang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China.
| | - Junli Shi
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, 999077, Hong Kong Special Administrative Region
| | - Bo Cong
- Department of Osteoarthropathy, Yantaishan Hospital, Yantai 264001, Shandong, PR China
| | - Zhonglian Wu
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Xianfeng Shang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Xu Hu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, 999077, Hong Kong Special Administrative Region
| | - Jian Yang
- Clinical Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, PR China
| | - Haidi Sun
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Zehao Gu
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Gong Cheng
- Department of Osteoarthropathy, Yantaishan Hospital, Yantai 264001, Shandong, PR China
| | - Hui Chong
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, 999077, Hong Kong Special Administrative Region.
| | - Yuchi Zhao
- Department of Osteoarthropathy, Yantaishan Hospital, Yantai 264001, Shandong, PR China.
| |
Collapse
|
3
|
Li C, Xu G, Wang Y, Huang L, Cai F, Meng L, Jin B, Jiang Z, Sun H, Zhao H, Lu X, Sang X, Huang P, Li F, Yang H, Mao Y, Zheng H. Acoustic-holography-patterned primary hepatocytes possess liver functions. Biomaterials 2024; 311:122691. [PMID: 38996673 DOI: 10.1016/j.biomaterials.2024.122691] [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: 02/04/2024] [Revised: 06/03/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024]
Abstract
Acoustic holography (AH), a promising approach for cell patterning, emerges as a powerful tool for constructing novel invitro 3D models that mimic organs and cancers features. However, understanding changes in cell function post-AH remains limited. Furthermore, replicating complex physiological and pathological processes solely with cell lines proves challenging. Here, we employed acoustical holographic lattice to assemble primary hepatocytes directly isolated from mice into a cell cluster matrix to construct a liver-shaped tissue sample. For the first time, we evaluated the liver functions of AH-patterned primary hepatocytes. The patterned model exhibited large numbers of self-assembled spheroids and superior multifarious core hepatocyte functions compared to cells in 2D and traditional 3D culture models. AH offers a robust protocol for long-term in vitro culture of primary cells, underscoring its potential for future applications in disease pathogenesis research, drug testing, and organ replacement therapy.
Collapse
Affiliation(s)
- Changcan Li
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China; Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Gang Xu
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yinhan Wang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Laixin Huang
- Shenzhen Institute of Advanced Technology, And Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| | - Feiyan Cai
- Shenzhen Institute of Advanced Technology, And Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| | - Long Meng
- Shenzhen Institute of Advanced Technology, And Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| | - Bao Jin
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Zhuoran Jiang
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Hang Sun
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Xingting Sang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Pengyu Huang
- Institute of Biomedical Engineering, PUMC & Chinese Academy of Medical Sciences (CAMS), Tianjin, China
| | - Fei Li
- Shenzhen Institute of Advanced Technology, And Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China.
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China.
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, China.
| | - Hairong Zheng
- Shenzhen Institute of Advanced Technology, And Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
4
|
Mehdikhani M, Yilgör P, Poursamar SA, Etemadi N, Gokyer S, Navid S, Farzan M, Farzan M, Babaei M, Rafienia M. A hybrid 3D-printed and electrospun bilayer pharmaceutical membrane based on polycaprolactone/chitosan/polyvinyl alcohol for wound healing applications. Int J Biol Macromol 2024; 282:136692. [PMID: 39437946 DOI: 10.1016/j.ijbiomac.2024.136692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Skin injuries resulting from physical trauma pose significant health risks, necessitating advanced wound care solutions. This investigation introduces an innovative bilayer wound dressing composed of 3D-printed propolis-coated polycaprolactone (PCL/PP) and an electrospun composite of polyvinyl alcohol, chitosan, polycaprolactone, and diltiazem (PVA/CTS/PCL/DTZ). SEM analysis revealed a bilayer structure with 89.23 ± 51.47 % porosity and uniformly distributed nanofibers. The scaffold tensile strength, with pore sizes of 100, 300, and 500 μm, was comparable to native skin. However, smaller pore sizes reduced water vapor transmission from 4211.59 ± 168.53 to 2358.49 ± 203.63 g/m2. The incorporation of DTZ lowered the contact angle to 35.23 ± 3.65°, while the addition of PCL reduced the degradation rate and modulated the release of DTZ by approximately 50 %. Moreover, lower pH increased the degradation rate and decreased swelling. The inclusion of propolis enhanced antibacterial activity, and 10 % DTZ promoted the viability, proliferation, and migration of fibroblasts and adipose-derived stem cells. However, increasing DTZ concentration to 12 % reduced cell viability. In vivo tests on rats demonstrated effective wound healing and anti-inflammatory properties of the bilayer samples. Regarding the aforementioned results, the PCL/PP-PVA/CTS/PCL/DTZ (10 % w/w) bilayer wound dressing is a promising candidate for wound healing applications.
Collapse
Affiliation(s)
- Mehdi Mehdikhani
- Department of Biomedical Engineering, Faculty of Engineering, University of Isfahan, Isfahan, Iran; Research and Technology Center for International Scientific Studies and Collaboration (CISSC), Ministry of Science, Tehran, Iran.
| | - Pinar Yilgör
- Department of Biomedical Engineering, Faculty of Engineering, Ankara University, Ankara, Türkiye
| | - Seyed Ali Poursamar
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Niloofar Etemadi
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seyda Gokyer
- Department of Biomedical Engineering, Faculty of Engineering, Ankara University, Ankara, Türkiye
| | - Sepehr Navid
- Core Research Facilities, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahan Farzan
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mahour Farzan
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Melika Babaei
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Rafienia
- Biosensor Research Center (BRC), Isfahan University of Medical Sciences (IUMS), Isfahan, Iran.
| |
Collapse
|
5
|
Chavoshnejad P, Li G, Solhtalab A, Liu D, Razavi MJ. A theoretical framework for predicting the heterogeneous stiffness map of brain white matter tissue. Phys Biol 2024; 21:066004. [PMID: 39427682 DOI: 10.1088/1478-3975/ad88e4] [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: 07/22/2024] [Accepted: 10/20/2024] [Indexed: 10/22/2024]
Abstract
Finding the stiffness map of biological tissues is of great importance in evaluating their healthy or pathological conditions. However, due to the heterogeneity and anisotropy of biological fibrous tissues, this task presents challenges and significant uncertainty when characterized only by single-mode loading experiments. In this study, we propose a new theoretical framework to map the stiffness landscape of fibrous tissues, specifically focusing on brain white matter tissue. Initially, a finite element (FE) model of the fibrous tissue was subjected to six loading cases, and their corresponding stress-strain curves were characterized. By employing multiobjective optimization, the material constants of an equivalent anisotropic material model were inversely extracted to best fit all six loading modes simultaneously. Subsequently, large-scale FE simulations were conducted, incorporating various fiber volume fractions and orientations, to train a convolutional neural network capable of predicting the equivalent anisotropic material properties solely based on the fibrous architecture of any given tissue. The proposed method, leveraging brain fiber tractography, was applied to a localized volume of white matter, demonstrating its effectiveness in precisely mapping the anisotropic behavior of fibrous tissue. In the long-term, the proposed method may find applications in traumatic brain injury, brain folding studies, and neurodegenerative diseases, where accurately capturing the material behavior of the tissue is crucial for simulations and experiments.
Collapse
Affiliation(s)
- Poorya Chavoshnejad
- Department of Mechanical Engineering, Binghamton University, State University of New York, Binghamton, NY 13902, United States of America
| | - Guangfa Li
- Department of Mechanical Engineering, Binghamton University, State University of New York, Binghamton, NY 13902, United States of America
| | - Akbar Solhtalab
- Department of Mechanical Engineering, Binghamton University, State University of New York, Binghamton, NY 13902, United States of America
| | - Dehao Liu
- Department of Mechanical Engineering, Binghamton University, State University of New York, Binghamton, NY 13902, United States of America
| | - Mir Jalil Razavi
- Department of Mechanical Engineering, Binghamton University, State University of New York, Binghamton, NY 13902, United States of America
| |
Collapse
|
6
|
McLeod KA, Di Gregorio M, Tinney D, Carmichael J, Zuanazzi D, Siqueira WL, Rizkalla A, Hamilton DW. Galectin-3/Gelatin Electrospun Scaffolds Modulate Collagen Synthesis in Skin Healing but Do Not Improve Wound Closure Kinetics. Bioengineering (Basel) 2024; 11:960. [PMID: 39451336 PMCID: PMC11504234 DOI: 10.3390/bioengineering11100960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Chronic wounds remain trapped in a pro-inflammatory state, with strategies targeted at inducing re-epithelialization and the proliferative phase of healing desirable. As a member of the lectin family, galectin-3 is implicated in the regulation of macrophage phenotype and epithelial migration. We investigated if local delivery of galectin-3 enhanced skin healing in a full-thickness excisional C57BL/6 mouse model. An electrospun gelatin scaffold loaded with galectin-3 was developed and compared to topical delivery of galectin-3. Electrospun gelatin/galectin-3 scaffolds had an average fiber diameter of 200 nm, with 83% scaffold porosity approximately and an average pore diameter of 1.15 μm. The developed scaffolds supported dermal fibroblast adhesion, matrix deposition, and proliferation in vitro. In vivo treatment of 6 mm full-thickness excisional wounds with gelatin/galectin-3 scaffolds did not influence wound closure, re-epithelialization, or macrophage phenotypes, but increased collagen synthesis. In comparison, topical delivery of galectin-3 [6.7 µg/mL] significantly increased arginase-I cell density at day 7 versus untreated and gelatin/galectin-3 scaffolds (p < 0.05). A preliminary assessment of increasing the concentration of topical galectin-3 demonstrated that at day 7, galectin-3 [12.5 µg/mL] significantly increased both epithelial migration and collagen content in a concentration-dependent manner. In conclusion, local delivery of galectin 3 shows potential efficacy in modulating skin healing in a concentration-dependent manner.
Collapse
Affiliation(s)
- Karrington A. McLeod
- Faculty of Engineering, School of Biomedical Engineering, University of Western Ontario, London, ON N6A 3K7, Canada (A.R.)
| | - Madeleine Di Gregorio
- Faculty of Engineering, School of Biomedical Engineering, University of Western Ontario, London, ON N6A 3K7, Canada (A.R.)
| | - Dylan Tinney
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada; (D.T.); (J.C.)
| | - Justin Carmichael
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada; (D.T.); (J.C.)
| | - David Zuanazzi
- Biochemistry Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (D.Z.); (W.L.S.)
| | - Walter L. Siqueira
- Biochemistry Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (D.Z.); (W.L.S.)
- College of Dentistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Amin Rizkalla
- Faculty of Engineering, School of Biomedical Engineering, University of Western Ontario, London, ON N6A 3K7, Canada (A.R.)
- School of Dentistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Douglas W. Hamilton
- Faculty of Engineering, School of Biomedical Engineering, University of Western Ontario, London, ON N6A 3K7, Canada (A.R.)
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada; (D.T.); (J.C.)
- School of Dentistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
7
|
Chen Q, Steinmetz K, Oh JK, Travaš-Sejdić J, Domigan LJ. Engineering an Extracellular Matrix Mimic Using Hemoglobin Protein Nanofibrils. ACS APPLIED BIO MATERIALS 2024; 7:6089-6100. [PMID: 39183644 DOI: 10.1021/acsabm.4c00713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Extracellular matrix (ECM) is essential for tissue development, providing structural support and a microenvironment that is necessary for cells. As tissue engineering advances, there is a growing demand for ECM mimics. Polycaprolactone (PCL) is a commonly used synthetic polymer for ECM mimic materials. However, its biologically inactive surface limits its direct application in tissue engineering. Our study aimed to improve the biocompatibility of PCL by incorporating hemoglobin nanofibrils (HbFs) into PCL using an electrospinning technique. HbFs were formed from bovine hemoglobin (Hb) extracted from industrial byproducts and designed to offer PCL an improved cell adhesion property. The fabricated HbFs@PCL electrospun scaffold exhibits improved fibroblast adherence, proliferation, and deeper fibroblast infiltration into the scaffold compared with the pure PCL scaffold, indicating its potential to be an ECM mimic. This study represents the pioneering utilization of Hb-sourced nanofibrils in the electrospun PCL scaffolds for tissue engineering applications.
Collapse
Affiliation(s)
- Qun Chen
- Chemical and Materials Engineering, The University of Auckland, 5 Grafton Road, Auckland 1010, New Zealand
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - Kai Steinmetz
- School of Biological Sciences, The University of Auckland, 3A Symonds Street, Auckland 1010, New Zealand
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
- The Riddet Institute-Advancing Frontiers in Food Science, Palmerston North 4410, New Zealand
- New Harvest, California 95811, United States
| | - Jin Kyo Oh
- Chemical and Materials Engineering, The University of Auckland, 5 Grafton Road, Auckland 1010, New Zealand
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - Jadranka Travaš-Sejdić
- Centre for Innovative Materials for Health, School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland 1023, New Zealand
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - Laura J Domigan
- Chemical and Materials Engineering, The University of Auckland, 5 Grafton Road, Auckland 1010, New Zealand
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
- The Riddet Institute-Advancing Frontiers in Food Science, Palmerston North 4410, New Zealand
| |
Collapse
|
8
|
Rijns L, Baker MB, Dankers PYW. Using Chemistry To Recreate the Complexity of the Extracellular Matrix: Guidelines for Supramolecular Hydrogel-Cell Interactions. J Am Chem Soc 2024; 146:17539-17558. [PMID: 38888174 PMCID: PMC11229007 DOI: 10.1021/jacs.4c02980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/17/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
Hydrogels have emerged as a promising class of extracellular matrix (ECM)-mimicking materials in regenerative medicine. Here, we briefly describe current state-of-the-art of ECM-mimicking hydrogels, ranging from natural to hybrid to completely synthetic versions, giving the prelude to the importance of supramolecular interactions to make true ECM mimics. The potential of supramolecular interactions to create ECM mimics for cell culture is illustrated through a focus on two different supramolecular hydrogel systems, both developed in our laboratories. We use some recent, significant findings to present important design principles underlying the cell-material interaction. To achieve cell spreading, we propose that slow molecular dynamics (monomer exchange within fibers) is crucial to ensure the robust incorporation of cell adhesion ligands within supramolecular fibers. Slow bulk dynamics (stress-relaxation─fiber rearrangements, τ1/2 ≈ 1000 s) is required to achieve cell spreading in soft gels (<1 kPa), while gel stiffness overrules dynamics in stiffer gels. Importantly, this resonates with the findings of others which specialize in different material types: cell spreading is impaired in case substrate relaxation occurs faster than clutch binding and focal adhesion lifetime. We conclude with discussing considerations and limitations of the supramolecular approach as well as provide a forward thinking perspective to further understand supramolecular hydrogel-cell interactions. Future work may utilize the presented guidelines underlying cell-material interactions to not only arrive at the next generation of ECM-mimicking hydrogels but also advance other fields, such as bioelectronics, opening up new opportunities for innovative applications.
Collapse
Affiliation(s)
- Laura Rijns
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
- Department
of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Matthew B. Baker
- Department
of Complex Tissue Regeneration, MERLN Institute for Technology Inspired
Regenerative Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology
Inspired Regenerative Medicine, Maastricht
University, 6200 MD Maastricht, The Netherlands
| | - Patricia Y. W. Dankers
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
- Department
of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Department
of Chemical Engineering and Chemistry, Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
9
|
Rasouli M, Shahghasempour L, Shirbaghaee Z, Hosseinzadeh S, Abbaszadeh HA, Fattahi R, Ranjbari J, Soleimani M. Mesenchymal stem cell therapy using Pal-KTTKS-enriched carboxylated cellulose improves burn wound in rat model. Arch Dermatol Res 2024; 316:353. [PMID: 38850353 DOI: 10.1007/s00403-024-03082-1] [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: 09/06/2023] [Revised: 09/06/2023] [Accepted: 04/26/2024] [Indexed: 06/10/2024]
Abstract
Despite the great progress in developing wound dressings, delayed wound closure still remains a global challenge. Thus, developing novel wound dressings and employing advanced strategies, including tissue engineering, are urgently desired. The carboxylated cellulose was developed through the in situ synthesis method and further reinforced by incorporating pal-KTTKS to stimulate collagen synthesis and improve wound healing. The developed composites supported cell adhesion and proliferation and showed good biocompatibility. To boost wound-healing performance, adipose-derived mesenchymal stem cells (MSC) were seeded on the pal-KTTKS-enriched composites to be implanted in a rat model of burn wound healing. Healthy male rats were randomly divided into four groups and wound-healing performance of Vaseline gauze (control), carboxylated cellulose (CBC), pal-KTTKS-enriched CBC (KTTKS-CBC), and MSCs seeded on the KTTKS-CBC composites (MSC-KTTKS-CBC) were evaluated on days 3, 7, and 14 post-implantation. In each group, the designed therapeutic dressings were renewed every 5 days to increase wound-healing performance. We found that KTTKS-CBC and MSC-KTTKS-CBC composites exhibited significantly better wound healing capability, as evidenced by significantly alleviated inflammation, increased collagen deposition, improved angiogenesis, and considerably accelerated wound closure. Nevertheless, the best wound-healing performance was observed in the MSC-KTTKS-CBC groups among all four groups. This research suggests that the MSC-KTTKS-CBC composite offers a great deal of promise as a wound dressing to enhance wound regeneration and expedite wound closure in the clinic.
Collapse
Affiliation(s)
- Mehdi Rasouli
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Lida Shahghasempour
- Department of Microbiology, Islamic Azad University, Karaj BranchKaraj, Iran
| | - Zeinab Shirbaghaee
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Simzar Hosseinzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat-Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Fattahi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Ranjbari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Masoud Soleimani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Ferrai C, Schulte C. Mechanotransduction in stem cells. Eur J Cell Biol 2024; 103:151417. [PMID: 38729084 DOI: 10.1016/j.ejcb.2024.151417] [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: 12/27/2023] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Nowadays, it is an established concept that the capability to reach a specialised cell identity via differentiation, as in the case of multi- and pluripotent stem cells, is not only determined by biochemical factors, but that also physical aspects of the microenvironment play a key role; interpreted by the cell through a force-based signalling pathway called mechanotransduction. However, the intricate ties between the elements involved in mechanotransduction, such as the extracellular matrix, the glycocalyx, the cell membrane, Integrin adhesion complexes, Cadherin-mediated cell/cell adhesion, the cytoskeleton, and the nucleus, are still far from being understood in detail. Here we report what is currently known about these elements in general and their specific interplay in the context of multi- and pluripotent stem cells. We furthermore merge this overview to a more comprehensive picture, that aims to cover the whole mechanotransductive pathway from the cell/microenvironment interface to the regulation of the chromatin structure in the nucleus. Ultimately, with this review we outline the current picture of the interplay between mechanotransductive cues and epigenetic regulation and how these processes might contribute to stem cell dynamics and fate.
Collapse
Affiliation(s)
- Carmelo Ferrai
- Institute of Pathology, University Medical Centre Göttingen, Germany.
| | - Carsten Schulte
- Department of Biomedical and Clinical Sciences and Department of Physics "Aldo Pontremoli", University of Milan, Italy.
| |
Collapse
|
11
|
Barajaa MA, Otsuka T, Ghosh D, Kan HM, Laurencin CT. Development of porcine skeletal muscle extracellular matrix-derived hydrogels with improved properties and low immunogenicity. Proc Natl Acad Sci U S A 2024; 121:e2322822121. [PMID: 38687784 PMCID: PMC11087813 DOI: 10.1073/pnas.2322822121] [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: 12/26/2023] [Accepted: 03/18/2024] [Indexed: 05/02/2024] Open
Abstract
Hydrogels derived from decellularized extracellular matrices (ECM) of animal origin show immense potential for regenerative applications due to their excellent cytocompatibility and biomimetic properties. Despite these benefits, the impact of decellularization protocols on the properties and immunogenicity of these hydrogels remains relatively unexplored. In this study, porcine skeletal muscle ECM (smECM) underwent decellularization using mechanical disruption (MD) and two commonly employed decellularization detergents, sodium deoxycholate (SDC) or Triton X-100. To mitigate immunogenicity associated with animal-derived ECM, all decellularized tissues were enzymatically treated with α-galactosidase to cleave the primary xenoantigen-the α-Gal antigen. Subsequently, the impact of the different decellularization protocols on the resultant hydrogels was thoroughly investigated. All methods significantly reduced total DNA content in hydrogels. Moreover, α-galactosidase treatment was crucial for cleaving α-Gal antigens, suggesting that conventional decellularization methods alone are insufficient. MD preserved total protein, collagen, sulfated glycosaminoglycan, laminin, fibronectin, and growth factors more efficiently than other protocols. The decellularization method impacted hydrogel gelation kinetics and ultrastructure, as confirmed by turbidimetric and scanning electron microscopy analyses. MD hydrogels demonstrated high cytocompatibility, supporting satellite stem cell recruitment, growth, and differentiation into multinucleated myofibers. In contrast, the SDC and Triton X-100 protocols exhibited cytotoxicity. Comprehensive in vivo immunogenicity assessments in a subcutaneous xenotransplantation model revealed MD hydrogels' biocompatibility and low immunogenicity. These findings highlight the significant influence of the decellularization protocol on hydrogel properties. Our results suggest that combining MD with α-galactosidase treatment is an efficient method for preparing low-immunogenic smECM-derived hydrogels with enhanced properties for skeletal muscle regenerative engineering and clinical applications.
Collapse
Affiliation(s)
- Mohammed A. Barajaa
- Department of Biomedical Engineering, College of Engineering, Imam Abdulrahman Bin Faisal University, Dammam34212, Saudi Arabia
| | - Takayoshi Otsuka
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT06030
| | - Debolina Ghosh
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT06030
| | - Ho-Man Kan
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT06030
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT06030
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT06269
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT06030
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT06269
- Department of Chemical & Bimolecular Engineering, University of Connecticut, Storrs, CT06269
| |
Collapse
|
12
|
Ortega JA, Soares de Aguiar GP, Chandravanshi P, Levy N, Engel E, Álvarez Z. Exploring the properties and potential of the neural extracellular matrix for next-generation regenerative therapies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1962. [PMID: 38723788 DOI: 10.1002/wnan.1962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 05/24/2024]
Abstract
The extracellular matrix (ECM) is a dynamic and complex network of proteins and molecules that surrounds cells and tissues in the nervous system and orchestrates a myriad of biological functions. This review carefully examines the diverse interactions between cells and the ECM, as well as the transformative chemical and physical changes that the ECM undergoes during neural development, aging, and disease. These transformations play a pivotal role in shaping tissue morphogenesis and neural activity, thereby influencing the functionality of the central nervous system (CNS). In our comprehensive review, we describe the diverse behaviors of the CNS ECM in different physiological and pathological scenarios and explore the unique properties that make ECM-based strategies attractive for CNS repair and regeneration. Addressing the challenges of scalability, variability, and integration with host tissues, we review how advanced natural, synthetic, and combinatorial matrix approaches enhance biocompatibility, mechanical properties, and functional recovery. Overall, this review highlights the potential of decellularized ECM as a powerful tool for CNS modeling and regenerative purposes and sets the stage for future research in this exciting field. This article is categorized under: Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Implantable Materials and Surgical Technologies > Nanomaterials and Implants.
Collapse
Affiliation(s)
- J Alberto Ortega
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Gisele P Soares de Aguiar
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Palash Chandravanshi
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Natacha Levy
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Elisabeth Engel
- IMEM-BRT Group, Department of Materials Science and Engineering, EEBE, Technical University of Catalonia (UPC), Barcelona, Spain
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Zaida Álvarez
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
13
|
Crossley RM, Johnson S, Tsingos E, Bell Z, Berardi M, Botticelli M, Braat QJS, Metzcar J, Ruscone M, Yin Y, Shuttleworth R. Modeling the extracellular matrix in cell migration and morphogenesis: a guide for the curious biologist. Front Cell Dev Biol 2024; 12:1354132. [PMID: 38495620 PMCID: PMC10940354 DOI: 10.3389/fcell.2024.1354132] [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: 12/11/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
The extracellular matrix (ECM) is a highly complex structure through which biochemical and mechanical signals are transmitted. In processes of cell migration, the ECM also acts as a scaffold, providing structural support to cells as well as points of potential attachment. Although the ECM is a well-studied structure, its role in many biological processes remains difficult to investigate comprehensively due to its complexity and structural variation within an organism. In tandem with experiments, mathematical models are helpful in refining and testing hypotheses, generating predictions, and exploring conditions outside the scope of experiments. Such models can be combined and calibrated with in vivo and in vitro data to identify critical cell-ECM interactions that drive developmental and homeostatic processes, or the progression of diseases. In this review, we focus on mathematical and computational models of the ECM in processes such as cell migration including cancer metastasis, and in tissue structure and morphogenesis. By highlighting the predictive power of these models, we aim to help bridge the gap between experimental and computational approaches to studying the ECM and to provide guidance on selecting an appropriate model framework to complement corresponding experimental studies.
Collapse
Affiliation(s)
- Rebecca M. Crossley
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Samuel Johnson
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Erika Tsingos
- Computational Developmental Biology Group, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - Zoe Bell
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Massimiliano Berardi
- LaserLab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Optics11 life, Amsterdam, Netherlands
| | | | - Quirine J. S. Braat
- Department of Applied Physics and Science Education, Eindhoven University of Technology, Eindhoven, Netherlands
| | - John Metzcar
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States
- Department of Informatics, Indiana University, Bloomington, IN, United States
| | | | - Yuan Yin
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
14
|
Golebiowska AA, Intravaia JT, Sathe VM, Kumbar SG, Nukavarapu SP. Decellularized extracellular matrix biomaterials for regenerative therapies: Advances, challenges and clinical prospects. Bioact Mater 2024; 32:98-123. [PMID: 37927899 PMCID: PMC10622743 DOI: 10.1016/j.bioactmat.2023.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
Tissue engineering and regenerative medicine have shown potential in the repair and regeneration of tissues and organs via the use of engineered biomaterials and scaffolds. However, current constructs face limitations in replicating the intricate native microenvironment and achieving optimal regenerative capacity and functional recovery. To address these challenges, the utilization of decellularized tissues and cell-derived extracellular matrix (ECM) has emerged as a promising approach. These biocompatible and bioactive biomaterials can be engineered into porous scaffolds and grafts that mimic the structural and compositional aspects of the native tissue or organ microenvironment, both in vitro and in vivo. Bioactive dECM materials provide a unique tissue-specific microenvironment that can regulate and guide cellular processes, thereby enhancing regenerative therapies. In this review, we explore the emerging frontiers of decellularized tissue-derived and cell-derived biomaterials and bio-inks in the field of tissue engineering and regenerative medicine. We discuss the need for further improvements in decellularization methods and techniques to retain structural, biological, and physicochemical characteristics of the dECM products in a way to mimic native tissues and organs. This article underscores the potential of dECM biomaterials to stimulate in situ tissue repair through chemotactic effects for the development of growth factor and cell-free tissue engineering strategies. The article also identifies the challenges and opportunities in developing sterilization and preservation methods applicable for decellularized biomaterials and grafts and their translation into clinical products.
Collapse
Affiliation(s)
| | - Jonathon T. Intravaia
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Vinayak M. Sathe
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Sangamesh G. Kumbar
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Syam P. Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| |
Collapse
|
15
|
Jain K, Lim KYE, Sheetz MP, Kanchanawong P, Changede R. Intrinsic self-organization of integrin nanoclusters within focal adhesions is required for cellular mechanotransduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567975. [PMID: 38045378 PMCID: PMC10690202 DOI: 10.1101/2023.11.20.567975] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Upon interaction with the extracellular matrix, the integrin receptors form nanoclusters as a first biochemical response to ligand binding. Here, we uncover a critical biodesign principle where these nanoclusters are spatially self-organized, facilitating effective mechanotransduction. Mouse Embryonic Fibroblasts (MEFs) with integrin β3 nanoclusters organized themselves with an intercluster distance of ∼550 nm on uniformly coated fibronectin substrates, leading to larger focal adhesions. We determined that this spatial organization was driven by cell-intrinsic factors since there was no pre-existing pattern on the substrates. Altering this spatial organization using cyclo-RGD functionalized Titanium nanodiscs (of 100 nm, corroborating to the integrin nanocluster size) spaced at intervals of 300 nm (almost half), 600 nm (normal) or 1000 nm (almost double) resulted in abrogation in mechanotransduction, indicating that a new parameter i.e., an optimal intercluster distance is necessary for downstream function. Overexpression of α-actinin, which induces a kink in the integrin tail, disrupted the establishment of the optimal intercluster distance, while simultaneous co-overexpression of talin head with α-actinin rescued it, indicating a concentration-dependent competition, and that cytoplasmic activation of integrin by talin head is required for the optimal intercluster organization. Additionally, talin head-mediated recruitment of FHOD1 that facilitates local actin polymerization at nanoclusters, and actomyosin contractility were also crucial for establishing the optimal intercluster distance and a robust mechanotransduction response. These findings demonstrate that cell-intrinsic machinery plays a vital role in organizing integrin receptor nanoclusters within focal adhesions, encoding essential information for downstream mechanotransduction signalling.
Collapse
|
16
|
Morrison TX, Gramlich WM. Tunable, thiol-ene, interpenetrating network hydrogels of norbornene-modified carboxymethyl cellulose and cellulose nanofibrils. Carbohydr Polym 2023; 319:121173. [PMID: 37567714 DOI: 10.1016/j.carbpol.2023.121173] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 06/25/2023] [Accepted: 06/30/2023] [Indexed: 08/13/2023]
Abstract
Carboxymethyl cellulose modified with norbornene groups (NorCMC) and cellulose nanofibrils (CNFs) produced through mechanical refining without chemical pretreatment formed interpenetrating network hydrogels through a UV-light initiated thiol-ene reaction. The molar ratio of thiols in crosslinkers to norbornene groups off the NorCMC (T:N), total polymer weight percent in the hydrogel, and weight percent of CNFs of the total polymer content of the hydrogels were varied to control hydrogel properties. This method enabled orders of magnitude changes to behavior. Swelling in aqueous environments could be significant (>150 %) without CNFs to minimal (<15 %) with the use of 50 % CNFs. NorCMC and CNF networks interacted synergistically to create hydrogels with compression modulus values spanning 1 to 150 kPa - the values of most biological tissues. T:N and total polymer weight percent could be varied to create hydrogels with different CNF content, but the same compression modulus, targeting 10 and 100 kPa hydrogels and providing a system that can independently vary fibrillar content and bulk modulus. Analysis of the effective crosslinks, thiol-ene network mesh size, and burst release of the polymer indicated synergistic interactions of the NorCMC thiol-ene and CNFs networks. These interactions enhanced modulus and degradation control of the network under physiological conditions.
Collapse
Affiliation(s)
| | - William M Gramlich
- Department of Chemistry, University of Maine, Orono, ME 04469, USA; Advanced Structures and Composites Center, University of Maine, Orono, ME 04469, USA; Institute of Medicine, University of Maine, Orono, ME 04469, USA.
| |
Collapse
|
17
|
Lemma ED, Motta A, Bucciarelli A, Mozetic P. Editorial: Advanced three-dimensional platforms for tissue regeneration: when the microenvironment matters. Front Bioeng Biotechnol 2023; 11:1265642. [PMID: 37622002 PMCID: PMC10446473 DOI: 10.3389/fbioe.2023.1265642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Affiliation(s)
| | - Antonella Motta
- Department of Industrial Engineering, University of Trento, Trento, Italy
| | | | - Pamela Mozetic
- Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
- Institute of Nanotechnology (NANOTEC), National Research Council, Lecce, Italy
| |
Collapse
|
18
|
Ghanem MA, Maxwell AD, Dalecki D, Sapozhnikov OA, Bailey MR. Phase holograms for the three-dimensional patterning of unconstrained microparticles. Sci Rep 2023; 13:9160. [PMID: 37280230 PMCID: PMC10244404 DOI: 10.1038/s41598-023-35337-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/16/2023] [Indexed: 06/08/2023] Open
Abstract
Acoustic radiation forces can remotely manipulate particles. Forces from a standing wave field align microscale particles along the nodal or anti-nodal locations of the field to form three-dimensional (3D) patterns. These patterns can be used to form 3D microstructures for tissue engineering applications. However, standing wave generation requires more than one transducer or a reflector, which is challenging to implement in vivo. Here, a method is developed and validated to manipulate microspheres using a travelling wave from a single transducer. Diffraction theory and an iterative angular spectrum approach are employed to design phase holograms to shape the acoustic field. The field replicates a standing wave and aligns polyethylene microspheres in water, which are analogous to cells in vivo, at pressure nodes. Using Gor'kov potential to calculate the radiation forces on the microspheres, axial forces are minimized, and transverse forces are maximized to create stable particle patterns. Pressure fields from the phase holograms and resulting particle aggregation patterns match predictions with a feature similarity index > 0.92, where 1 is a perfect match. The resulting radiation forces are comparable to those produced from a standing wave, which suggests opportunities for in vivo implementation of cell patterning toward tissue engineering applications.
Collapse
Affiliation(s)
- Mohamed A Ghanem
- Center for Industrial and Medical Ultrasound, Applied Physics Laboratory, University of Washington, 1013 NE 40th St., Seattle, WA, 98105, USA.
| | - Adam D Maxwell
- Department of Urology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Diane Dalecki
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Oleg A Sapozhnikov
- Center for Industrial and Medical Ultrasound, Applied Physics Laboratory, University of Washington, 1013 NE 40th St., Seattle, WA, 98105, USA
- Physics Faculty, Moscow State University, Moscow, 119991, Russia
| | - Michael R Bailey
- Center for Industrial and Medical Ultrasound, Applied Physics Laboratory, University of Washington, 1013 NE 40th St., Seattle, WA, 98105, USA
- Department of Urology, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
19
|
Ketebo AA, Din SU, Lee G, Park S. Mechanobiological Analysis of Nanoparticle Toxicity. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13101682. [PMID: 37242097 DOI: 10.3390/nano13101682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/06/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023]
Abstract
Nanoparticles (NPs) are commonly used in healthcare and nanotherapy, but their toxicity at high concentrations is well-known. Recent research has shown that NPs can also cause toxicity at low concentrations, disrupting various cellular functions and leading to altered mechanobiological behavior. While researchers have used different methods to investigate the effects of NPs on cells, including gene expression and cell adhesion assays, the use of mechanobiological tools in this context has been underutilized. This review emphasizes the importance of further exploring the mechanobiological effects of NPs, which could reveal valuable insights into the mechanisms behind NP toxicity. To investigate these effects, different methods, including the use of polydimethylsiloxane (PDMS) pillars to study cell motility, traction force production, and rigidity sensing contractions, have been employed. Understanding how NPs affect cell cytoskeletal functions through mechanobiology could have significant implications, such as developing innovative drug delivery systems and tissue engineering techniques, and could improve the safety of NPs for biomedical applications. In summary, this review highlights the significance of incorporating mechanobiology into the study of NP toxicity and demonstrates the potential of this interdisciplinary field to advance our knowledge and practical use of NPs.
Collapse
Affiliation(s)
- Abdurazak Aman Ketebo
- School of Mechanical Engineering, Sungkyunkwan University, Suwon 16499, Republic of Korea
| | - Shahab Ud Din
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University, Suwon 16499, Republic of Korea
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
20
|
Gupta D, Wiklander OP, Wood MJ, El-Andaloussi S. Biodistribution of therapeutic extracellular vesicles. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2023; 4:170-190. [PMID: 39697988 PMCID: PMC11648525 DOI: 10.20517/evcna.2023.12] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 12/20/2024]
Abstract
The field of extracellular vesicles (EVs) has seen a tremendous paradigm shift in the past two decades, from being regarded as cellular waste bags to being considered essential mediators in intercellular communication. Their unique ability to transfer macromolecules across cells and biological barriers has made them a rising star in drug delivery. Mounting evidence suggests that EVs can be explored as efficient drug delivery vehicles for a range of therapeutic macromolecules. In contrast to many synthetic delivery systems, these vesicles appear exceptionally well tolerated in vivo. This tremendous development in the therapeutic application of EVs has been made through technological advancement in labelling and understanding the in vivo biodistribution of EVs. Here in this review, we have summarised the recent findings in EV in vivo pharmacokinetics and discussed various biological barriers that need to be surpassed to achieve tissue-specific delivery.
Collapse
Affiliation(s)
- Dhanu Gupta
- Department of Paediatrics. University of Oxford, Oxford OX3 7TY, UK
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge 14151, Sweden
| | - Oscar P.B Wiklander
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge 14151, Sweden
| | - Matthew J.A Wood
- Department of Paediatrics. University of Oxford, Oxford OX3 7TY, UK
| | - Samir El-Andaloussi
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Huddinge 14151, Sweden
| |
Collapse
|
21
|
Jones CL, Penney BT, Theodossiou SK. Engineering Cell-ECM-Material Interactions for Musculoskeletal Regeneration. Bioengineering (Basel) 2023; 10:bioengineering10040453. [PMID: 37106640 PMCID: PMC10135874 DOI: 10.3390/bioengineering10040453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
The extracellular microenvironment regulates many of the mechanical and biochemical cues that direct musculoskeletal development and are involved in musculoskeletal disease. The extracellular matrix (ECM) is a main component of this microenvironment. Tissue engineered approaches towards regenerating muscle, cartilage, tendon, and bone target the ECM because it supplies critical signals for regenerating musculoskeletal tissues. Engineered ECM-material scaffolds that mimic key mechanical and biochemical components of the ECM are of particular interest in musculoskeletal tissue engineering. Such materials are biocompatible, can be fabricated to have desirable mechanical and biochemical properties, and can be further chemically or genetically modified to support cell differentiation or halt degenerative disease progression. In this review, we survey how engineered approaches using natural and ECM-derived materials and scaffold systems can harness the unique characteristics of the ECM to support musculoskeletal tissue regeneration, with a focus on skeletal muscle, cartilage, tendon, and bone. We summarize the strengths of current approaches and look towards a future of materials and culture systems with engineered and highly tailored cell-ECM-material interactions to drive musculoskeletal tissue restoration. The works highlighted in this review strongly support the continued exploration of ECM and other engineered materials as tools to control cell fate and make large-scale musculoskeletal regeneration a reality.
Collapse
Affiliation(s)
- Calvin L Jones
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Dr MS2085, Boise, ID 83725, USA
| | - Brian T Penney
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Dr MS2085, Boise, ID 83725, USA
| | - Sophia K Theodossiou
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Dr MS2085, Boise, ID 83725, USA
| |
Collapse
|
22
|
ANTXR1 as a potential sensor of extracellular mechanical cues. Acta Biomater 2023; 158:80-86. [PMID: 36638946 DOI: 10.1016/j.actbio.2023.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/18/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
Cell adhesion molecules mediate cell-cell or cell-matrix interactions, some of which are mechanical sensors, such as integrins. Emerging evidence indicates that anthrax toxin receptor 1 (ANTXR1), a newly identified cell adhesion molecule, can also sense extracellular mechanical signals such as hydrostatic pressure and extracellular matrix (ECM) rigidity. ANTXR1 can interact with ECM through connecting intracellular cytoskeleton and ECM molecules (just like integrins) to regulate numerous biological processes, such as cell adhesion, cell migration or ECM homeostasis. Although with high structural similarity to integrins, its functions and downstream signal transduction are independent from those of integrins. In this perspective, based on existing evidence in literature, we analyzed the structural and functional evidence that ANTXR1 can act as a potential sensor for extracellular mechanical cues. To our knowledge, this is the first in-depth overview of ANTXR1 from the perspective of mechanobiology. STATEMENT OF SIGNIFICANCE: An overview of ANTXR1 from the perspective of mechanobiology; An analysis of mechanical sensitivity of ANTXR1 in structure and function; A summary of existing evidence of ANTXR1 as a potential mechanosensor.
Collapse
|
23
|
Rasouli M, Hosseinzadeh S, Mortazavi SM, Fattahi R, Ranjbari J, Soleimani M. Do Carboxymethyl Cellulose and Pal-KTTKS Make Bacterial Cellulose a Superior Wound Dressing or Skin Scaffold? POLYM-PLAST TECH MAT 2023. [DOI: 10.1080/25740881.2023.2175222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Affiliation(s)
- Mehdi Rasouli
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Simzar Hosseinzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Maryam Mortazavi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Fattahi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Ranjbari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Mana G, Valdembri D, Askari JA, Li Z, Caswell P, Zhu C, Humphries MJ, Ballestrem C, Serini G. The βI domain promotes active β1 integrin clustering into mature adhesion sites. Life Sci Alliance 2023; 6:e202201388. [PMID: 36410791 PMCID: PMC9679427 DOI: 10.26508/lsa.202201388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/22/2022] Open
Abstract
Modulation of integrin function is required in many physiological and pathological settings, such as angiogenesis and cancer. Integrin allosteric changes, clustering, and trafficking cooperate to regulate cell adhesion and motility on extracellular matrix proteins via mechanisms that are partly defined. By exploiting four monoclonal antibodies recognizing distinct conformational epitopes, we show that in endothelial cells (ECs), the extracellular βI domain, but not the hybrid or I-EGF2 domain of active β1 integrins, promotes their FAK-regulated clustering into tensin 1-containing fibrillar adhesions and impairs their endocytosis. In this regard, the βI domain-dependent clustering of active β1 integrins is necessary to favor fibronectin-elicited directional EC motility, which cannot be effectively promoted by β1 integrin conformational activation alone.
Collapse
Affiliation(s)
- Giulia Mana
- Candiolo Cancer Institute - FPO, IRCCS, Candiolo (TO), Italy
- Department of Oncology, University of Torino School of Medicine, Candiolo (TO), Italy
| | - Donatella Valdembri
- Candiolo Cancer Institute - FPO, IRCCS, Candiolo (TO), Italy
- Department of Oncology, University of Torino School of Medicine, Candiolo (TO), Italy
| | - Janet A Askari
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Zhenhai Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Patrick Caswell
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Martin J Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Christoph Ballestrem
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Guido Serini
- Candiolo Cancer Institute - FPO, IRCCS, Candiolo (TO), Italy
- Department of Oncology, University of Torino School of Medicine, Candiolo (TO), Italy
| |
Collapse
|
25
|
Solbu AA, Caballero D, Damigos S, Kundu SC, Reis RL, Halaas Ø, Chahal AS, Strand BL. Assessing cell migration in hydrogels: An overview of relevant materials and methods. Mater Today Bio 2023; 18:100537. [PMID: 36659998 PMCID: PMC9842866 DOI: 10.1016/j.mtbio.2022.100537] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/05/2022] [Accepted: 12/28/2022] [Indexed: 12/29/2022] Open
Abstract
Cell migration is essential in numerous living processes, including embryonic development, wound healing, immune responses, and cancer metastasis. From individual cells to collectively migrating epithelial sheets, the locomotion of cells is tightly regulated by multiple structural, chemical, and biological factors. However, the high complexity of this process limits the understanding of the influence of each factor. Recent advances in materials science, tissue engineering, and microtechnology have expanded the toolbox and allowed the development of biomimetic in vitro assays to investigate the mechanisms of cell migration. Particularly, three-dimensional (3D) hydrogels have demonstrated a superior ability to mimic the extracellular environment. They are therefore well suited to studying cell migration in a physiologically relevant and more straightforward manner than in vivo approaches. A myriad of synthetic and naturally derived hydrogels with heterogeneous characteristics and functional properties have been reported. The extensive portfolio of available hydrogels with different mechanical and biological properties can trigger distinct biological responses in cells affecting their locomotion dynamics in 3D. Herein, we describe the most relevant hydrogels and their associated physico-chemical characteristics typically employed to study cell migration, including established cell migration assays and tracking methods. We aim to give the reader insight into existing literature and practical details necessary for performing cell migration studies in 3D environments.
Collapse
Affiliation(s)
- Anita Akbarzadeh Solbu
- Department of Biotechnology and Food Sciences, NOBIPOL, NTNU- Norwegian University of Science and Technology, Trondheim, Norway
| | - David Caballero
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal
| | - Spyridon Damigos
- Department of Biotechnology and Food Sciences, NOBIPOL, NTNU- Norwegian University of Science and Technology, Trondheim, Norway
| | - Subhas C. Kundu
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal
| | - Øyvind Halaas
- Department of Clinical and Molecular Medicine, NTNU- Norwegian University of Science and Technology, Trondheim, Norway
| | - Aman S. Chahal
- Department of Biotechnology and Food Sciences, NOBIPOL, NTNU- Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU- Norwegian University of Science and Technology, Trondheim, Norway
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Berit L. Strand
- Department of Biotechnology and Food Sciences, NOBIPOL, NTNU- Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
26
|
Mierke CT. Physical and biological advances in endothelial cell-based engineered co-culture model systems. Semin Cell Dev Biol 2023; 147:58-69. [PMID: 36732105 DOI: 10.1016/j.semcdb.2023.01.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023]
Abstract
Scientific knowledge in the field of cell biology and mechanobiology heavily leans on cell-based in vitro experiments and models that favor the examination and comprehension of certain biological processes and occurrences across a variety of environments. Cell culture assays are an invaluable instrument for a vast spectrum of biomedical and biophysical investigations. The quality of experimental models in terms of simplicity, reproducibility, and combinability with other methods, and in particular the scale at which they depict cell fate in native tissues, is critical to advancing the knowledge of the comprehension of cell-cell and cell-matrix interactions in tissues and organs. Typically, in vitro models are centered on the experimental tinkering of mammalian cells, most often cultured as monolayers on planar, two-dimensional (2D) materials. Notwithstanding the significant advances and numerous findings that have been accomplished with flat biology models, their usefulness for generating further new biological understanding is constrained because the simple 2D setting does not reproduce the physiological response of cells in natural living tissues. In addition, the co-culture systems in a 2D stetting weakly mirror their natural environment of tissues and organs. Significant advances in 3D cell biology and matrix engineering have resulted in the creation and establishment of a new type of cell culture shapes that more accurately represents the in vivo microenvironment and allows cells and their interactions to be analyzed in a biomimetic approach. Contemporary biomedical and biophysical science has novel advances in technology that permit the design of more challenging and resilient in vitro models for tissue engineering, with a particular focus on scaffold- or hydrogel-based formats, organotypic cultures, and organs-on-chips, which cover the purposes of co-cultures. Even these complex systems must be kept as simplified as possible in order to grasp a particular section of physiology too very precisely. In particular, it is highly appreciated that they bridge the space between conventional animal research and human (patho)physiology. In this review, the recent progress in 3D biomimetic culturation is presented with a special focus on co-cultures, with an emphasis on the technological building blocks and endothelium-based co-culture models in cancer research that are available for the development of more physiologically relevant in vitro models of human tissues under normal and diseased conditions. Through applications and samples of various physiological and disease models, it is possible to identify the frontiers and future engagement issues that will have to be tackled to integrate synthetic biomimetic culture systems far more successfully into biomedical and biophysical investigations.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, Leipzig, Germany.
| |
Collapse
|
27
|
Wang H, Sun WQ, Wang J. Complete proteomic profiling of regenerative bio-scaffolds with a two-step trypsinization method. J Biomed Mater Res B Appl Biomater 2023; 111:62-72. [PMID: 35822935 DOI: 10.1002/jbm.b.35132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/20/2022] [Accepted: 06/28/2022] [Indexed: 01/01/2023]
Abstract
Regenerative bio-scaffolds, widely used for clinical tissue reconstruction and tissue repairs, are functionally diversified and structurally complex decellularized tissue materials (e.g., extracellular matrix, ECM). ECM is naturally cross-linked and can be further selectively cross-linked upon processing. Identification, quantification and bioinformatics functional comparison of all ECM proteins are challenging for regenerative bio-scaffolds. In this study, we have applied proteomic profiling with a two-step sequential trypsinization method, and identified and quantified 300-400 constituent proteins in three commercially available regenerative bio-scaffolds (BioDesign Surgisis, ReGen tissue matrix, and ThormalGEN mesh). These proteins were classified into four categories and 14 subcategories based on their mainly biological function. The main components of regenerative bio-scaffolds were highly abundant ECM structural proteins, and the minor parts of bio-scaffolds were lowly abundant, less cross-linked, functionally more diversified proteins, especially extracellular fluid proteins that were easily solubilized by trypsin. The comparative analysis has revealed large differences in the number, type, abundance and function of identified proteins, as well as the extent of decellularization and cross-linking among regenerative bio-scaffolds. So, the proteomic profiling with a two-step sequential trypsinization method could not only provide the molecular basis to better understand the degradation process of regenerative bio-scaffolds in vivo and different clinical outcomes among various regenerative bio-scaffolds, facilitate the exploration of the response mechanisms in the host's early clinical stages of ECM-induced tissue regeneration that is still poorly understood, but also can be used for optimization of the decellularization and cross-linking process, product characterization and rational design of new ECM products.
Collapse
Affiliation(s)
- Huidan Wang
- Institute of Biothermal Science and Technology, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Wendell Q Sun
- Institute of Biothermal Science and Technology, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jian Wang
- National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
28
|
Xiao J, Wei Q, Xue J, Yang Z, Deng Z, Zhao F. Preparation and In Vitro Bioactivity Study of a Novel Hollow Mesoporous Bioactive Glass Nanofiber Scaffold. Molecules 2022; 27:molecules27227973. [PMID: 36432072 PMCID: PMC9697868 DOI: 10.3390/molecules27227973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
In this study, a novel three-dimensional hollow mesoporous bioactive glass nanofiber scaffold has been synthesized with a template-assisted sol-gel method using bacterial cellulose (BC) as a template and nonionic triblock copolymer (P123) as a pore-directing agent, ethyl orthosilicate (TEOS), calcium nitrate tetrahydrate (CN), and triethyl phosphate (TEP) as glass precursors. Scanning and transmission electron microscopies, X-ray diffraction, nitrogen adsorption-desorption, and nuclear magnetic resonance method were applied to characterize the morphology, crystal structure, and chemical structure of the mesoporous bioactive glass nanofiber scaffold. Furthermore, the in vitro bioactivity and biocompatibility were also explored. The obtained scaffold depicted nanofiber-like morphology and interconnected three-dimensional network structure that replicated the BC template. The scaffold showed a large specific surface area (230.0 cm2 g-1) and pore volume (0.2 m3 g-1). More importantly, the scaffold exhibited excellent apatite-forming ability and cellular biocompatibility. We believe that the hollow mesoporous bioactive glass nanofiber scaffold has great potential application in bone tissue regeneration.
Collapse
Affiliation(s)
- Jian Xiao
- School of Physics and Electronic Information, Shangrao Normal University, Shangrao 334001, China
- School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
- Correspondence: (J.X.); (F.Z.)
| | - Qianghua Wei
- Primary Health Department, Shangrao Health Committee, Shangrao 334001, China
| | - Jinhong Xue
- Basic Department, Jinci College of Shanxi Medical University, Taiyuan 030025, China
| | - Zide Yang
- School of Physics and Electronic Information, Shangrao Normal University, Shangrao 334001, China
| | - Zhicheng Deng
- Institute of Science and Technology, China Railway Nanchang Group Co., Ltd., Nanchang 330002, China
| | - Fulai Zhao
- School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
- Correspondence: (J.X.); (F.Z.)
| |
Collapse
|
29
|
Terrassoux L, Claux H, Bacari S, Meignan S, Furlan A. A Bloody Conspiracy. Blood Vessels and Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2022; 14:cancers14194581. [PMID: 36230504 PMCID: PMC9558972 DOI: 10.3390/cancers14194581] [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: 08/18/2022] [Revised: 09/10/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary The tumor microenvironment has risen over the last years as a significant contributor to the failure of antitumoral strategies due to its numerous pro-tumorigenic activities. In this review, we focused on two features of this microenvironment, namely angiogenesis and immunity, which have been the targets of therapies to tackle tumors via its microenvironmental part over the last decade. Increasing our knowledge of the complex interactions within this ecosystem is mandatory to optimize these therapeutic approaches. The development of innovative experimental models is of great help in reaching this goal. Abstract Cancer progression occurs in concomitance with a profound remodeling of the cellular microenvironment. Far from being a mere passive event, the re-orchestration of interactions between the various cell types surrounding tumors highly contributes to the progression of the latter. Tumors notably recruit and stimulate the sprouting of new blood vessels through a process called neo-angiogenesis. Beyond helping the tumor cope with an increased metabolic demand associated with rapid growth, this also controls the metastatic dissemination of cancer cells and the infiltration of immune cells in the tumor microenvironment. To decipher this critical interplay for the clinical progression of tumors, the research community has developed several valuable models in the last decades. This review offers an overview of the various instrumental solutions currently available, including microfluidic chips, co-culture models, and the recent rise of organoids. We highlight the advantages of each technique and the specific questions they can address to better understand the tumor immuno-angiogenic ecosystem. Finally, we discuss this development field’s fundamental and applied perspectives.
Collapse
Affiliation(s)
- Lisa Terrassoux
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Hugo Claux
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Salimata Bacari
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Samuel Meignan
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Alessandro Furlan
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
- Correspondence:
| |
Collapse
|
30
|
Cenhrang K, Robart L, Castiaux AD, Martin RS. 3D printed devices with integrated collagen scaffolds for cell culture studies including transepithelial/transendothelial electrical resistance (TEER) measurements. Anal Chim Acta 2022; 1221:340166. [PMID: 35934386 PMCID: PMC9511703 DOI: 10.1016/j.aca.2022.340166] [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: 04/27/2022] [Revised: 07/07/2022] [Accepted: 07/10/2022] [Indexed: 11/01/2022]
Abstract
In this paper, we describe the use of 3D printed devices for both static and flow studies that contain electrospun collagen scaffolds and can accommodate transepithelial/transendothelial electrical resistance (TEER) measurements. Electrospinning was used to create the collagen scaffold, followed by an optimized 1-Ethyl-3-(3-dimethylaminopropyl) carbodiimide/N-Hydroxysuccinimide (EDC/NHS) cross-linking procedure to produce stable collagen fibers that are similar in size to fibers in vivo. LC/MS was used to study the leaching of solvent and NHS from the scaffold, with several rinsing steps being shown to eliminate the leaching and promote the culture of Madin-Darby Canine Kidney (MDCK) epithelial cells on the scaffold. Both static and flow 2-part devices were successfully fabricated by 3D printing using either VeroClear or MED610 material (PolyJet printing) and assembling the scaffold between laser cut Teflon gaskets. The devices were designed to easily accommodate commonly used STX2 chopstick electrodes for TEER measurements. A detailed comparison was made between the use of collagen scaffolds vs other electrospun materials for cell culture. The collagen extracellular matrix model displayed a high barrier functionality for up to 7 days. In addition, a different 3D printed device with a collagen scaffold is described to incorporate continuous flow and replenishment of media under the cell layer in a manner that also enables periodic recording of TEER measurements. Overall, this work shows that the combination of biological ECM materials such as collagen into microfluidic devices that incorporate flow have great potential to form more realistic cell culture models in areas such as blood brain barrier research.
Collapse
Affiliation(s)
| | - Logan Robart
- Department of Chemistry, Saint Louis University, USA
| | - Andre D Castiaux
- Department of Chemistry, Saint Louis University, USA; Center for Additive Manufacturing, Saint Louis University, USA
| | - R Scott Martin
- Department of Chemistry, Saint Louis University, USA; Center for Additive Manufacturing, Saint Louis University, USA.
| |
Collapse
|
31
|
Eichholz K, Freeman F, Pitacco P, Nulty J, Ahern D, Burdis R, Browe D, Garcia O, Hoey D, Kelly DJ. Scaffold microarchitecture regulates angiogenesis and the regeneration of large bone defects. Biofabrication 2022; 14. [PMID: 35947963 DOI: 10.1088/1758-5090/ac88a1] [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: 02/16/2022] [Accepted: 08/10/2022] [Indexed: 11/11/2022]
Abstract
Emerging 3D printing technologies can provide exquisite control over the external shape and internal architecture of scaffolds and tissue engineered constructs, enabling systematic studies to explore how geometric design features influence the regenerative process. Here we used fused deposition modelling (FDM) and melt electrowriting (MEW) to investigate how scaffold microarchitecture influences the healing of large bone defects. FDM was used to fabricate scaffolds with relatively large fibre diameters and low porosities, while MEW was used to fabricate scaffolds with smaller fibre diameters and higher porosities, with both scaffolds being designed to have comparable surface areas. Scaffold microarchitecture significantly influenced the healing response following implantation into critically sized femoral defects in rats, with the FDM scaffolds supporting the formation of larger bone spicules through its pores, while the MEW scaffolds supported the formation of a more round bone front during healing. After 12 weeks in vivo, both MEW and FDM scaffolds supported significantly higher levels of defect vascularisation compared to empty controls, while the MEW scaffolds supported higher levels of new bone formation. Somewhat surprisingly, this superior healing in the MEW group did not correlate with higher levels of angiogenesis, with the FDM scaffold supporting greater total vessel formation and the formation of larger vessels, while the MEW scaffold promoted the formation of a dense microvasculature with minimal evidence of larger vessels infiltrating the defect region. To conclude, the small fibre diameter, high porosity and high specific surface area of the MEW scaffold proved beneficial for osteogenesis and bone regeneration, demonstrating that changes in scaffold architecture enabled by this additive manufacturing technique can dramatically modulate angiogenesis and tissue regeneration without the need for complex exogenous growth factors. These results provide a valuable insight into the importance of 3D printed scaffold architecture when developing new bone tissue engineering strategies.
Collapse
Affiliation(s)
- Kian Eichholz
- Department of Mechanical and Manufacturing Engineering, University of Dublin Trinity College, Parsons Building, Dublin, IRELAND
| | - Fiona Freeman
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Parsons building, Dublin, 2, IRELAND
| | - Pierluca Pitacco
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Parsons Building, Dublin 2, Dublin, 2, IRELAND
| | - Jessica Nulty
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Parsons Building, Dublin 2, Dublin, 2, IRELAND
| | - Daniel Ahern
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Parsons Building, Dublin 2, Dublin, 2, IRELAND
| | - Ross Burdis
- Trinity Biomedical Institute, Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Dublin, D02 PN40, IRELAND
| | - David Browe
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Parsons building, Dublin, 2, IRELAND
| | - Orquidea Garcia
- Johnson & Johnson 3D Printing Innovation & Customer Solutions, Johnson & Johnson Services Inc, Irvine, California, 0000, UNITED STATES
| | - David Hoey
- Department of Mechanical and Manufacturing Engineering, University of Dublin Trinity College, Parsons building, Dublin, 2, IRELAND
| | - Daniel John Kelly
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Parsons Building, Dublin 2, Dublin, 2, IRELAND
| |
Collapse
|
32
|
Vahala D, Choi YS. Modelling the Tumor Microenvironment: Recapitulating Nano- and Micro-Scale Properties that Regulate Tumor Progression. Front Cell Dev Biol 2022; 10:908799. [PMID: 35800896 PMCID: PMC9254080 DOI: 10.3389/fcell.2022.908799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/20/2022] [Indexed: 12/02/2022] Open
Abstract
Breast cancer remains a significant burden with 1 in 8 women affected and metastasis posing a significant challenge for patient survival. Disease progression involves remodeling of the extracellular matrix (ECM). In breast cancer, tissue stiffness increases owing to an increase in collagen production by recruited cancer-associated fibroblasts (CAFs). These stromal modifications are notable during primary tumor growth and have a dualistic action by creating a hard capsule to prevent penetration of anti-cancer therapies and forming a favorable environment for tumor progression. Remodeling of the tumor microenvironment immediately presented to cells can include changes in protein composition, concentration and structural arrangement and provides the first mechanical stimuli in the metastatic cascade. Not surprisingly, metastatic cancer cells possess the ability to mechanically adapt, and their adaptability ensures not only survival but successful invasion within altered environments. In the past decade, the importance of the microenvironment and its regulatory role in diseases have gained traction and this is evident in the shift from plastic culture to the development of novel biomaterials that mimic in vivo tissue. With these advances, elucidations can be made into how ECM remodeling and more specifically, altered cell-ECM adhesions, regulate tumor growth and cancer cell plasticity. Such enabling tools in mechanobiology will identify fundamental mechanisms in cancer progression that eventually help develop preventative and therapeutic treatment from a clinical perspective. This review will focus on current platforms engineered to mimic the micro and nano-properties of the tumor microenvironment and subsequent understanding of mechanically regulated pathways in cancer.
Collapse
|
33
|
Poornima K, Francis AP, Hoda M, Eladl MA, Subramanian S, Veeraraghavan VP, El-Sherbiny M, Asseri SM, Hussamuldin ABA, Surapaneni KM, Mony U, Rajagopalan R. Implications of Three-Dimensional Cell Culture in Cancer Therapeutic Research. Front Oncol 2022; 12:891673. [PMID: 35646714 PMCID: PMC9133474 DOI: 10.3389/fonc.2022.891673] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022] Open
Abstract
Replicating the naturalistic biomechanical milieu of cells is a primary requisite to uncover the fundamental life processes. The native milieu is significantly not replicated in the two-dimensional (2D) cell cultures. Alternatively, the current three-dimensional (3D) culture techniques can replicate the properties of extracellular matrix (ECM), though the recreation of the original microenvironment is challenging. The organization of cells in a 3D manner contributes to better insight about the tumorigenesis mechanism of the in vitro cancer models. Gene expression studies are susceptible to alterations in their microenvironment. Physiological interactions among neighboring cells also contribute to gene expression, which is highly replicable with minor modifications in 3D cultures. 3D cell culture provides a useful platform for identifying the biological characteristics of tumor cells, particularly in the drug sensitivity area of translational medicine. It promises to be a bridge between traditional 2D culture and animal experiments and is of great importance for further research in tumor biology. The new imaging technology and the implementation of standard protocols can address the barriers interfering with the live cell observation in a natural 3D physiological environment.
Collapse
Affiliation(s)
- Kolluri Poornima
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Pondicherry, India
| | - Arul Prakash Francis
- Centre of Molecular Medicine and Diagnostics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Muddasarul Hoda
- Department of Biological Sciences, Aliah University, Kolkata, India
| | - Mohamed Ahmed Eladl
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Srividya Subramanian
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Pondicherry, India
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | - Saad Mohamed Asseri
- Department of Clinical Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | | | - Krishna Mohan Surapaneni
- Departments of Biochemistry, Molecular Virology, Research, Clinical Skills, and Simulation, Panimalar Medical College Hospital and Research Institute, Chennai, India
| | - Ullas Mony
- Centre of Molecular Medicine and Diagnostics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Rukkumani Rajagopalan
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Pondicherry, India
| |
Collapse
|
34
|
Riquelme-Guzmán C, Beck T, Edwards-Jorquera S, Schlüßler R, Müller P, Guck J, Möllmert S, Sandoval-Guzmán T. In vivo assessment of mechanical properties during axolotl development and regeneration using confocal Brillouin microscopy. Open Biol 2022; 12:220078. [PMID: 35728623 PMCID: PMC9213112 DOI: 10.1098/rsob.220078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In processes such as development and regeneration, where large cellular and tissue rearrangements occur, cell fate and behaviour are strongly influenced by tissue mechanics. While most well-established tools probing mechanical properties require an invasive sample preparation, confocal Brillouin microscopy captures mechanical parameters optically with high resolution in a contact-free and label-free fashion. In this work, we took advantage of this tool and the transparency of the highly regenerative axolotl to probe its mechanical properties in vivo for the first time. We mapped the Brillouin frequency shift with high resolution in developing limbs and regenerating digits, the most studied structures in the axolotl. We detected a gradual increase in the cartilage Brillouin frequency shift, suggesting decreasing tissue compressibility during both development and regeneration. Moreover, we were able to correlate such an increase with the regeneration stage, which was undetected with fluorescence microscopy imaging. The present work evidences the potential of Brillouin microscopy to unravel the mechanical changes occurring in vivo in axolotls, setting the basis to apply this technique in the growing field of epimorphic regeneration.
Collapse
Affiliation(s)
- Camilo Riquelme-Guzmán
- CRTD/Center for Regenerative Therapies TU Dresden, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Department of Internal Medicine 3, Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Timon Beck
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Sandra Edwards-Jorquera
- Department of Internal Medicine 3, Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Raimund Schlüßler
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Paul Müller
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Jochen Guck
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Stephanie Möllmert
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Tatiana Sandoval-Guzmán
- Department of Internal Medicine 3, Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,Paul Langerhans Institute Dresden, Helmholtz Centre Munich, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
35
|
Kao SH, Liang SY, Cheng PL, Tu HL. Surface Viscosity-Dependent Neurite Initiation in Cortical Neurons. Adv Biol (Weinh) 2022; 6:e2101325. [PMID: 35362269 DOI: 10.1002/adbi.202101325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/14/2022] [Indexed: 01/27/2023]
Abstract
Dynamic extracellular environments profoundly affect the behavior and function of cells both biochemically and mechanically. Neurite initiation is the first step for neurons to establish intricate neuronal networks. How such a process is modulated by mechanical factors is not fully understood. Particularly, it is unknown whether the molecular clutch model, which has been used to explain cell responses to matrix rigidity, also holds for neurite initiation. To study how mechanical properties modulate neurite initiation, substrates with various well-defined surface viscosities using supported lipid bilayers (SLBs) are synthesized. The results show that ligands with intermediate viscosity greatly maximize neurite initiation in primary neurons, while neurite initiation is drastically limited on substrates with higher or lower viscosity. Importantly, biochemical characterizations reveal altered focal adhesion and calpain activity are associated with distinct neurite initiation patterns. Collectively, these results indicate that neurite initiation is surface viscosity-dependent; there is an optimal range of surface viscosities to drive neurite initiation. Upon binding to ligands of varying viscosities, calpain activity is differentially triggered and leads to distinct levels of neurite outgrowth. These findings not only enhance the understanding of how extracellular environments regulate neurons, but also demonstrate the potential utility of SLBs for neural tissue engineering applications.
Collapse
Affiliation(s)
- Shih-Han Kao
- Institute of Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Shu-Yang Liang
- Institute of Molecular Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Pei-Lin Cheng
- Institute of Molecular Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Hsiung-Lin Tu
- Institute of Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| |
Collapse
|
36
|
Jin B, Kong W, Zhao X, Chen S, Sun Q, Feng J, Song D, Han D. Substrate stiffness affects the morphology, proliferation, and radiosensitivity of cervical squamous carcinoma cells. Tissue Cell 2021; 74:101681. [PMID: 34837739 DOI: 10.1016/j.tice.2021.101681] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/24/2021] [Accepted: 11/09/2021] [Indexed: 12/22/2022]
Abstract
Cervical cancer is associated with the highest morbidity rate among gynecological cancers. Radiotherapy plays an important role in the treatment of cervical cancer. However, a considerable number of patients are radiation resistant, leading to a poor prognosis. Matrix stiffness is related to the occurrence, development, and chemoresistance of solid tumors. The association between matrix stiffness and radiosensitivity in cervical cancer cells remains unknown. Here, we sought to determine the effect of matrix stiffness on the phenotype and radiosensitivity of cervical cancer cells. Cervical squamous carcinoma SiHa cells were grown on substrates of different stiffnesses (0.5, 5, and 25 kPa). Cell morphology, proliferation, and radiosensitivity were examined. Cells grown on hard substrates displayed stronger proliferative activity, larger size, and higher differentiation degree, which was reflected in a more mature skeleton assembly, more abundant pseudopodia formation, and smaller nuclear/cytoplasmic ratio. In addition, SiHa cells exhibited stiffness-dependent resistance to radiation, possibly via altered apoptosis-related protein expression. Our findings demonstrate that matrix stiffness affects the morphology, proliferation, and radiosensitivity of SiHa cells. Tissue stiffness may be an indicator of the sensitivity of a patient to radiotherapy. Thus, the data provide insights into the diagnosis of cervical cancer and the design of future radiotherapies.
Collapse
Affiliation(s)
- Bixia Jin
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China
| | - Weimin Kong
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China.
| | - Xuanyu Zhao
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China
| | - Shuning Chen
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China
| | - Quanmei Sun
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Jiantao Feng
- Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Dan Song
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China
| | - Dong Han
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| |
Collapse
|
37
|
Yeo M, Kim G. Electrohydrodynamic-direct-printed cell-laden microfibrous structure using alginate-based bioink for effective myotube formation. Carbohydr Polym 2021; 272:118444. [PMID: 34420709 DOI: 10.1016/j.carbpol.2021.118444] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 11/26/2022]
Abstract
In this study, a fully aligned microfibrous structure fabricated using fibrin-assisted alginate bioink and electrohydrodynamic direct-printing was proposed for skeletal muscle tissue engineering. To safely construct the aligned alginate/fibrin microfibrous structure laden with myoblasts or endothelial cells, various printing conditions, such as an applied electric field, distance between the nozzle and target, and nozzle moving speed, were selected appropriately. Furthermore, to accelerate the formation of myotubes more efficiently, the alginate/fibrin bioink with vascular endothelial cells was co-printed into a spatially patterned structure within a myoblast-laden structure. The myoblast-laden structure co-cultured with endothelial cells presented fully aligned myotube formation and significantly greater myogenic differentiation compared to the myoblast-laden structure without the endothelial cells owing to the more abundant secretion of angiogenic cytokines. Also, when adipose stem cell- and endothelial cell-laden fibrous structure was implanted in a mouse volumetric muscle loss model, accelerated volumetric muscle repair was observed compared to the defect model. Based on the results, this study demonstrates an alginate-based bioink and new bio-fabricating method to obtain microfibrous cell-laden alginate/fibrin structures with mechanically stable and topographical cues. The proposed method can provide a myoblast/endothelial cell-laden fibrous alginate structure to efficiently induce engineering of skeletal muscle tissue, which could be used in muscle-on-a-chip or recovering structures of volumetric muscle defects.
Collapse
Affiliation(s)
- Miji Yeo
- Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University (SKKU), Suwon 16419, South Korea
| | - GeunHyung Kim
- Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University (SKKU), Suwon 16419, South Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
38
|
Jamal A, Bernardini A, Dini D. Microscale characterisation of the time-dependent mechanical behaviour of brain white matter. J Mech Behav Biomed Mater 2021; 125:104917. [PMID: 34710852 DOI: 10.1016/j.jmbbm.2021.104917] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/06/2021] [Accepted: 10/16/2021] [Indexed: 01/08/2023]
Abstract
Brain mechanics is a topic of deep interest because of the significant role of mechanical cues in both brain function and form. Specifically, capturing the heterogeneous and anisotropic behaviour of cerebral white matter (WM) is extremely challenging and yet the data on WM at a spatial resolution relevant to tissue components are sparse. To investigate the time-dependent mechanical behaviour of WM, and its dependence on local microstructural features when subjected to small deformations, we conducted atomic force microscopy (AFM) stress relaxation experiments on corpus callosum (CC), corona radiata (CR) and fornix (FO) of fresh ovine brain. Our experimental results show a dependency of the tissue mechanical response on axons orientation, with e.g. the stiffness of perpendicular and parallel samples is different in all three regions of WM whereas the relaxation behaviour is different for the CC and FO regions. An inverse modelling approach was adopted to extract Prony series parameters of the tissue components, i.e. axons and extra cellular matrix with its accessory cells, from experimental data. Using a bottom-up approach, we developed analytical and FEA estimates that are in good agreement with our experimental results. Our systematic characterisation of sheep brain WM using a combination of AFM experiments and micromechanical models provide a significant contribution for predicting localised time-dependent mechanics of brain tissue. This information can lead to more accurate computational simulations, therefore aiding the development of surgical robotic solutions for drug delivery and accurate tissue mimics, as well as the determination of criteria for tissue injury and predict brain development and disease progression.
Collapse
Affiliation(s)
- Asad Jamal
- Department of Mechanical Engineering, Imperial College London, London, SW7 2AZ, UK.
| | - Andrea Bernardini
- Department of Mechanical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Daniele Dini
- Department of Mechanical Engineering, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
39
|
Micro-scaffolds as synthetic cell niches: recent advances and challenges. Curr Opin Biotechnol 2021; 73:290-299. [PMID: 34619481 DOI: 10.1016/j.copbio.2021.08.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/19/2021] [Accepted: 08/21/2021] [Indexed: 01/01/2023]
Abstract
Micro-fabrication and nano-fabrication provide useful approaches to address fundamental biological questions by mimicking the physiological microenvironment in which cells carry out their functions. In particular, 2D patterns and 3D scaffolds obtained via lithography, direct laser writing, and other techniques allow for shaping hydrogels, synthetic polymers and biologically derived materials to create structures for (single) cell culture. Applications of micro-scaffolds mimicking cell niches include stem cell self-renewal, differentiation, and lineage specification. This review moves from technological aspects of scaffold microfabrication for cell biological applications to a broad overview of advances in (stem) cell research: achievements for embryonic, induced pluripotent, mesenchymal, and neural stem cells are treated in detail, while a particular section is dedicated to micro-scaffolds used to study single cells in basic cell biology.
Collapse
|
40
|
Park W, Gao G, Cho DW. Tissue-Specific Decellularized Extracellular Matrix Bioinks for Musculoskeletal Tissue Regeneration and Modeling Using 3D Bioprinting Technology. Int J Mol Sci 2021; 22:7837. [PMID: 34360604 PMCID: PMC8346156 DOI: 10.3390/ijms22157837] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/20/2021] [Accepted: 07/20/2021] [Indexed: 12/11/2022] Open
Abstract
The musculoskeletal system is a vital body system that protects internal organs, supports locomotion, and maintains homeostatic function. Unfortunately, musculoskeletal disorders are the leading cause of disability worldwide. Although implant surgeries using autografts, allografts, and xenografts have been conducted, several adverse effects, including donor site morbidity and immunoreaction, exist. To overcome these limitations, various biomedical engineering approaches have been proposed based on an understanding of the complexity of human musculoskeletal tissue. In this review, the leading edge of musculoskeletal tissue engineering using 3D bioprinting technology and musculoskeletal tissue-derived decellularized extracellular matrix bioink is described. In particular, studies on in vivo regeneration and in vitro modeling of musculoskeletal tissue have been focused on. Lastly, the current breakthroughs, limitations, and future perspectives are described.
Collapse
Affiliation(s)
- Wonbin Park
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang 37673, Korea;
| | - Ge Gao
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China;
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang 37673, Korea;
- POSTECH-Catholic Biomedical Engineering Institute, Pohang University of Science and Technology, Pohang 37673, Korea
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| |
Collapse
|
41
|
Tai CS, Lan KC, Wang E, Chan FE, Hsieh MT, Huang CW, Weng SL, Chen PC, Chen WL. Nanotopography as Artificial Microenvironment for Accurate Visualization of Metastasis Development via Simulation of ECM Dynamics. NANO LETTERS 2021; 21:1400-1411. [PMID: 33522822 DOI: 10.1021/acs.nanolett.0c04209] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Metastatic progression is mediated by complex interactions between deregulated extracellular matrix (ECM) and cancer cells and remains a major challenge in cancer management. To investigate the role of ECM dynamics in promoting metastasis development, we developed an artificial microenvironment (AME) platform comprised of nanodot arrays of increasing diameter. Cells cultured on the platform showed increasing signs of mesenchymal-like cell transition as AME diameter increased, suggesting accurate simulation of ECM-mediated gene regulation. Gene expression was analyzed to determine genes significant to transition, which were then used to select appropriate small molecule drugs for time course treatments. Our results suggest that the platform can identify critical target genes as well as possible drug candidates. Overall, the AME platform allows for the study of intricate ECM-induced gene expression trends across metastasis development that would otherwise be difficult to visualize in vivo and may open new avenues toward successful personalized cancer management.
Collapse
Affiliation(s)
- Chun-San Tai
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Kuan-Chun Lan
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Erick Wang
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Fu-Erh Chan
- Department of Materials and Mineral Resources Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Ming-Ting Hsieh
- Department of Materials and Mineral Resources Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Ching-Wen Huang
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- Division of Thoracic Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Shun-Long Weng
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu City, Taiwan
| | - Po-Chun Chen
- Department of Materials and Mineral Resources Engineering, National Taipei University of Technology, Taipei, Taiwan
- Institute of Material Science and Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Wen Liang Chen
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
42
|
Previdi A, Piazzoni C, Borghi F, Schulte C, Lorenzelli L, Giacomozzi F, Bucciarelli A, Malgaroli A, Lamanna J, Moro A, Racchetti G, Podestà A, Lenardi C, Milani P. Micropatterning of Substrates for the Culture of Cell Networks by Stencil-Assisted Additive Nanofabrication. MICROMACHINES 2021; 12:mi12010094. [PMID: 33477416 PMCID: PMC7829752 DOI: 10.3390/mi12010094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 11/16/2022]
Abstract
The fabrication of in vitro neuronal cell networks where cells are chemically or electrically connected to form functional circuits with useful properties is of great interest. Standard cell culture substrates provide ensembles of cells that scarcely reproduce physiological structures since their spatial organization and connectivity cannot be controlled. Supersonic Cluster Beam Deposition (SCBD) has been used as an effective additive method for the large-scale fabrication of interfaces with extracellular matrix-mimicking surface nanotopography and reproducible morphological properties for cell culture. Due to the high collimation of SCBD, it is possible to exploit stencil masks for the fabrication of patterned films and reproduce features as small as tens of micrometers. Here, we present a protocol to fabricate micropatterned cell culture substrates based on the deposition of nanostructured cluster-assembled zirconia films by stencil-assisted SCBD. The effectiveness of this approach is demonstrated by the fabrication of micrometric patterns able to confine primary astrocytes. Calcium waves propagating in the astrocyte networks are shown.
Collapse
Affiliation(s)
- Anita Previdi
- CIMaINa and Dipartimento di Fisica, Università degli Studi di Milano, Via Celoria 16, 20133 Milano, Italy; (A.P.); (C.P.); (F.B.); (C.S.); (A.P.); (C.L.)
| | - Claudio Piazzoni
- CIMaINa and Dipartimento di Fisica, Università degli Studi di Milano, Via Celoria 16, 20133 Milano, Italy; (A.P.); (C.P.); (F.B.); (C.S.); (A.P.); (C.L.)
| | - Francesca Borghi
- CIMaINa and Dipartimento di Fisica, Università degli Studi di Milano, Via Celoria 16, 20133 Milano, Italy; (A.P.); (C.P.); (F.B.); (C.S.); (A.P.); (C.L.)
| | - Carsten Schulte
- CIMaINa and Dipartimento di Fisica, Università degli Studi di Milano, Via Celoria 16, 20133 Milano, Italy; (A.P.); (C.P.); (F.B.); (C.S.); (A.P.); (C.L.)
| | - Leandro Lorenzelli
- Center for Materials and Microsystems (CMM), Bruno Kessler Foundation (FBK), Via Sommarive 18, 38123 Trento, Italy; (L.L.); (F.G.); (A.B.)
| | - Flavio Giacomozzi
- Center for Materials and Microsystems (CMM), Bruno Kessler Foundation (FBK), Via Sommarive 18, 38123 Trento, Italy; (L.L.); (F.G.); (A.B.)
| | - Alessio Bucciarelli
- Center for Materials and Microsystems (CMM), Bruno Kessler Foundation (FBK), Via Sommarive 18, 38123 Trento, Italy; (L.L.); (F.G.); (A.B.)
| | - Antonio Malgaroli
- Center for Behavioral Neuroscience and Communication (BNC), Università Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milano, Italy; (A.M.); (J.L.); (A.M.); (G.R.)
| | - Jacopo Lamanna
- Center for Behavioral Neuroscience and Communication (BNC), Università Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milano, Italy; (A.M.); (J.L.); (A.M.); (G.R.)
| | - Andrea Moro
- Center for Behavioral Neuroscience and Communication (BNC), Università Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milano, Italy; (A.M.); (J.L.); (A.M.); (G.R.)
| | - Gabriella Racchetti
- Center for Behavioral Neuroscience and Communication (BNC), Università Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milano, Italy; (A.M.); (J.L.); (A.M.); (G.R.)
| | - Alessandro Podestà
- CIMaINa and Dipartimento di Fisica, Università degli Studi di Milano, Via Celoria 16, 20133 Milano, Italy; (A.P.); (C.P.); (F.B.); (C.S.); (A.P.); (C.L.)
| | - Cristina Lenardi
- CIMaINa and Dipartimento di Fisica, Università degli Studi di Milano, Via Celoria 16, 20133 Milano, Italy; (A.P.); (C.P.); (F.B.); (C.S.); (A.P.); (C.L.)
| | - Paolo Milani
- CIMaINa and Dipartimento di Fisica, Università degli Studi di Milano, Via Celoria 16, 20133 Milano, Italy; (A.P.); (C.P.); (F.B.); (C.S.); (A.P.); (C.L.)
- Correspondence:
| |
Collapse
|
43
|
Zhang X, Liu Y, Clark KL, Padget AM, Alexander PG, Dai J, Zhu W, Lin H. Mesenchymal stem cell-derived extracellular matrix (mECM): a bioactive and versatile scaffold for musculoskeletal tissue engineering. ACTA ACUST UNITED AC 2020; 16:012002. [PMID: 32906098 DOI: 10.1088/1748-605x/abb6b3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cell-derived extracellular matrix (mECM) has received increased attention in the fields of tissue engineering and scaffold-assisted regeneration. mECM exhibits many unique characteristics, such as robust bioactivity, biocompatibility, ease of use, and the potential for autologous tissue engineering. As the use of mECM has increased in musculoskeletal tissue engineering, it should be noted that mECM generated from current methods has inherited insufficiencies, such as low mechanical properties and lack of internal architecture. In this review, we first summarize the development and use of mECM as a scaffold for musculoskeletal tissue regeneration and highlight our current progress on moving this technology toward clinical application. Then we review recent methods to improve the properties of mECM that will overcome current weaknesses. Lastly, we propose future studies that will pave the road for mECM application in regenerating tissues in humans.
Collapse
Affiliation(s)
- Xiurui Zhang
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America. Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, People's Republic of China. These authors contributed equally to this work
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Ghosh S, Haldar S, Gupta S, Bisht A, Chauhan S, Kumar V, Roy P, Lahiri D. Anisotropically Conductive Biodegradable Scaffold with Coaxially Aligned Carbon Nanotubes for Directional Regeneration of Peripheral Nerves. ACS APPLIED BIO MATERIALS 2020; 3:5796-5812. [PMID: 35021810 DOI: 10.1021/acsabm.0c00534] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fascicular rearrangement of an injured peripheral nerve requires reconnection of nerve sprouts from anterior and Büngner bands from distal sides of the lesion, failing to which leads to inefficient regeneration of the injured nerve. However, existing neural scaffolds have limited neuroregeneration efficiency because of either the lack of alignment of fibers and a conductive second phase, leading to compromised electrical conductivity, or the lack of extracellular matrix components and in vivo validation. The present study reports a biocompatible, multiwall carbon nanotube (MWCNT)-reinforced, anisotropically conductive, electrospun, aligned nanofibrous scaffold, ensuring maximal peripheral nerve regeneration. Electrospinning parameters were modulated to deposit random and parallel fibers in separate scaffolds for comparative analysis on the effect of fiber alignment on regeneration. Both types of scaffolds were reinforced with MWCNTs to impart electrical conductivity. Nonreinforced scaffolds were nonconductive. In this comparative study, MWCNT-reinforced, aligned scaffolds showed better tensile property with increased conductivity along the direction of alignment, thereby ensuring an escalated neural-regeneration rate. Collectively, in vitro studies established the scaffolds to be highly biocompatible, promoting cell growth and proliferation. With 85% more anisotropic conductivity in the direction of the alignment and the degradation kinetics tuned to the regeneration regime, the MWCNT-reinforced, aligned scaffold efficiently healed injured sciatic nerves in rats within 30 days. Rigorous revivification of the tissue was due to coordinated Wallerian degeneration and expedited guided axonal regeneration. Structural and functional analysis of nerves in vivo showed the aligned, MWCNT-reinforced scaffold to be very efficient in peripheral sciatic nerve regeneration. This study notes the efficacy of the coaxially aligned, MWCNT-reinforced neural scaffold, with a capability of establishing remarkable advancement in the field of peripheral neural regeneration.
Collapse
Affiliation(s)
- Souvik Ghosh
- Biomaterials and Multiscale Mechanics Lab, Department of Metallurgical and Materials Engineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.,Centre of Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.,Molecular Endocrinology Lab, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Swati Haldar
- Centre of Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Sumeet Gupta
- Department of Pharmacy, Maharshi Markandeshwar University (Deemed to Be University), Mullana, Haryana 133207, India
| | - Ankita Bisht
- Biomaterials and Multiscale Mechanics Lab, Department of Metallurgical and Materials Engineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Samrat Chauhan
- Department of Pharmacy, Maharshi Markandeshwar University (Deemed to Be University), Mullana, Haryana 133207, India
| | - Viney Kumar
- Molecular Endocrinology Lab, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Partha Roy
- Molecular Endocrinology Lab, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Debrupa Lahiri
- Biomaterials and Multiscale Mechanics Lab, Department of Metallurgical and Materials Engineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.,Centre of Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| |
Collapse
|
45
|
Ambekar YS, Singh M, Scarcelli G, Rueda EM, Hall BM, Poché RA, Larin KV. Characterization of retinal biomechanical properties using Brillouin microscopy. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:JBO-200208LR. [PMID: 32981240 PMCID: PMC7519206 DOI: 10.1117/1.jbo.25.9.090502] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/04/2020] [Indexed: 05/03/2023]
Abstract
SIGNIFICANCE The retina is critical for vision, and several diseases may alter its biomechanical properties. However, assessing the biomechanical properties of the retina nondestructively is a challenge due to its fragile nature and location within the eye globe. Advancements in Brillouin spectroscopy have provided the means for nondestructive investigations of retina biomechanical properties. AIM We assessed the biomechanical properties of mouse retinas using Brillouin microscopy noninvasively and showed the potential of Brillouin microscopy to differentiate the type and layers of retinas based on stiffness. APPROACH We used Brillouin microscopy to quantify stiffness of fresh and paraformaldehyde (PFA)-fixed retinas. As further proof-of-concept, we demonstrated a change in the stiffness of a retina with N-methyl-D-aspartate (NMDA)-induced damage, compared to an undamaged sample. RESULTS We found that the retina layers with higher cell body density had higher Brillouin modulus compared to less cell-dense layers. We have also demonstrated that PFA-fixed retina samples were stiffer compared with fresh samples. Further, NMDA-induced neurotoxicity leads to retinal ganglion cell (RGC) death and reactive gliosis, increasing the stiffness of the RGC layer. CONCLUSION Brillouin microscopy can be used to characterize the stiffness distribution of the layers of the retina and can be used to differentiate tissue at different conditions based on biomechanical properties.
Collapse
Affiliation(s)
- Yogeshwari S. Ambekar
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
| | - Manmohan Singh
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
| | - Giuliano Scarcelli
- University of Maryland, Fischell Department of Bioengineering, College Park, Maryland, United States
| | - Elda M. Rueda
- Baylor College of Medicine, Department of Molecular Physiology and Biophysics, Houston, Texas, United States
| | - Benjamin M. Hall
- Baylor College of Medicine, Department of Molecular Physiology and Biophysics, Houston, Texas, United States
| | - Ross A. Poché
- Baylor College of Medicine, Department of Molecular Physiology and Biophysics, Houston, Texas, United States
- Address all correspondence to Ross A. Poché, E-mail: ; Kirill V. Larin, E-mail:
| | - Kirill V. Larin
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
- Baylor College of Medicine, Department of Molecular Physiology and Biophysics, Houston, Texas, United States
- Address all correspondence to Ross A. Poché, E-mail: ; Kirill V. Larin, E-mail:
| |
Collapse
|
46
|
Chighizola M, Previdi A, Dini T, Piazzoni C, Lenardi C, Milani P, Schulte C, Podestà A. Adhesion force spectroscopy with nanostructured colloidal probes reveals nanotopography-dependent early mechanotransductive interactions at the cell membrane level. NANOSCALE 2020; 12:14708-14723. [PMID: 32618323 DOI: 10.1039/d0nr01991g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Mechanosensing, the ability of cells to perceive and interpret the microenvironmental biophysical cues (such as the nanotopography), impacts strongly cellular behaviour through mechanotransductive processes and signalling. These events are predominantly mediated by integrins, the principal cellular adhesion receptors located at the cell/extracellular matrix (ECM) interface. Because of the typical piconewton force range and nanometre length scale of mechanotransductive interactions, achieving a detailed understanding of the spatiotemporal dynamics occurring at the cell/microenvironment interface is challenging; sophisticated interdisciplinary methodologies are required. Moreover, an accurate control over the nanotopographical features of the microenvironment is essential, in order to systematically investigate and precisely assess the influence of the different nanotopographical motifs on the mechanotransductive process. In this framework, we were able to study and quantify the impact of microenvironmental nanotopography on early cellular adhesion events by means of adhesion force spectroscopy based on innovative colloidal probes mimicking the nanotopography of natural ECMs. These probes provided the opportunity to detect nanotopography-specific modulations of the molecular clutch force loading dynamics and integrin clustering at the level of single binding events, in the critical time window of nascent adhesion formation. Following this approach, we found that the nanotopographical features are responsible for an excessive force loading in single adhesion sites after 20-60 s of interaction, causing a drop in the number of adhesion sites. However, by manganese treatment we demonstrated that the availability of activated integrins is a critical regulatory factor for these nanotopography-dependent dynamics.
Collapse
Affiliation(s)
- M Chighizola
- C.I.Ma.I.Na. and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, via Celoria 16, 20133 Milan, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Barber-Pérez N, Georgiadou M, Guzmán C, Isomursu A, Hamidi H, Ivaska J. Mechano-responsiveness of fibrillar adhesions on stiffness-gradient gels. J Cell Sci 2020; 133:jcs242909. [PMID: 32393601 PMCID: PMC7328166 DOI: 10.1242/jcs.242909] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/22/2020] [Indexed: 01/01/2023] Open
Abstract
Fibrillar adhesions are important structural and adhesive components in fibroblasts, and are required for fibronectin fibrillogenesis. While nascent and focal adhesions are known to respond to mechanical cues, the mechanoresponsive nature of fibrillar adhesions remains unclear. Here, we used ratiometric analysis of paired adhesion components to determine an appropriate fibrillar adhesion marker. We found that active α5β1-integrin exhibits the most definitive fibrillar adhesion localization compared to other proteins, such as tensin-1, reported to be in fibrillar adhesions. To elucidate the mechanoresponsiveness of fibrillar adhesions, we designed a cost-effective and reproducible technique to fabricate physiologically relevant stiffness gradients on thin polyacrylamide (PA) hydrogels, embedded with fluorescently labelled beads. We generated a correlation curve between bead density and hydrogel stiffness, thus enabling a readout of stiffness without the need for specialized knowhow, such as atomic force microscopy (AFM). We find that stiffness promotes growth of fibrillar adhesions in a tensin-1-dependent manner. Thus, the formation of these extracellular matrix-depositing structures is coupled to the mechanical parameters of the cell environment and may enable cells to fine-tune their matrix environment in response to changing physical conditions.
Collapse
Affiliation(s)
- Nuria Barber-Pérez
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FIN-20520 Turku, Finland
| | - Maria Georgiadou
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FIN-20520 Turku, Finland
| | - Camilo Guzmán
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FIN-20520 Turku, Finland
| | - Aleksi Isomursu
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FIN-20520 Turku, Finland
| | - Hellyeh Hamidi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FIN-20520 Turku, Finland
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FIN-20520 Turku, Finland
- Department of Biochemistry, University of Turku, FIN-20520 Turku, Finland
| |
Collapse
|
48
|
Costa G, Gentile F. A nanomechanical model enables comprehensive characterization of biological tissues in ultrasound imaging. Biomed Phys Eng Express 2020; 6:035026. [PMID: 33438671 DOI: 10.1088/2057-1976/ab8740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Sonography, elastography, sonoelastography are ultrasound imaging techniques commonly used in the clinical practice for the diagnosis of many pathological conditions. These highly reliable, non-invasive methods use high frequency, elastic pressure waves (ultrasounds) to interrogate the internal structure of biological tissues and organs, and the continuum mechanics hypothesis to reconstruct, from the output of the system, the biophysical characteristics of the samples. Nevertheless, continuum mechanics disregards the discrete nature of tissues and organs, resulting in an inability for the model to describe some important tissue biophysical characteristics such as the cell size and their spatial layout. Here, we used the theory of doublet mechanics - a discrete nano-mechanical field theory - to model the propagation of ultrasounds in a multilayered biological tissue. We found that the output of the model exhibits a very high sensitivity to the macro and micro characteristics of the tissue, including cell size. We used results from the model to correlate the internal structure of the samples to the reflection coefficient, i.e. the continuum level response of the system. This model, and its more sophisticated evolutions that will be developed over time, can complement traditional ultrasound imaging, and provide ways to analyze non-invasively living tissues with a resolution inaccessible to conventional techniques of analysis, including positron emission tomography, computer tomography, and magnetic resonance.
Collapse
Affiliation(s)
- G Costa
- Institute for Microelectronics and Microsystems, National Research Council (CNR), 80131 Naples, Italy
| | | |
Collapse
|
49
|
Pradhan S, Banda OA, Farino CJ, Sperduto JL, Keller KA, Taitano R, Slater JH. Biofabrication Strategies and Engineered In Vitro Systems for Vascular Mechanobiology. Adv Healthc Mater 2020; 9:e1901255. [PMID: 32100473 PMCID: PMC8579513 DOI: 10.1002/adhm.201901255] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/24/2020] [Indexed: 12/17/2022]
Abstract
The vascular system is integral for maintaining organ-specific functions and homeostasis. Dysregulation in vascular architecture and function can lead to various chronic or acute disorders. Investigation of the role of the vascular system in health and disease has been accelerated through the development of tissue-engineered constructs and microphysiological on-chip platforms. These in vitro systems permit studies of biochemical regulation of vascular networks and parenchymal tissue and provide mechanistic insights into the biophysical and hemodynamic forces acting in organ-specific niches. Detailed understanding of these forces and the mechanotransductory pathways involved is necessary to develop preventative and therapeutic strategies targeting the vascular system. This review describes vascular structure and function, the role of hemodynamic forces in maintaining vascular homeostasis, and measurement approaches for cell and tissue level mechanical properties influencing vascular phenomena. State-of-the-art techniques for fabricating in vitro microvascular systems, with varying degrees of biological and engineering complexity, are summarized. Finally, the role of vascular mechanobiology in organ-specific niches and pathophysiological states, and efforts to recapitulate these events using in vitro microphysiological systems, are explored. It is hoped that this review will help readers appreciate the important, but understudied, role of vascular-parenchymal mechanotransduction in health and disease toward developing mechanotherapeutics for treatment strategies.
Collapse
Affiliation(s)
- Shantanu Pradhan
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Omar A. Banda
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Cindy J. Farino
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John L. Sperduto
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Keely A. Keller
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Ryan Taitano
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John H. Slater
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE 19716, USA
- Delaware Biotechnology Institute, 15 Innovation Way, Newark, DE 19711, USA
| |
Collapse
|
50
|
Young J, Hua X, Somsel H, Reichart F, Kessler H, Spatz JP. Integrin Subtypes and Nanoscale Ligand Presentation Influence Drug Sensitivity in Cancer Cells. NANO LETTERS 2020; 20:1183-1191. [PMID: 31908168 PMCID: PMC7020138 DOI: 10.1021/acs.nanolett.9b04607] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Cancer cell-matrix interactions have been shown to enhance cancer cell survival via the activation of pro-survival signaling pathways. These pathways are initiated at the site of interaction, i.e., integrins, and thus, their inhibition has been the target of therapeutic strategies. Individual roles for fibronectin-binding integrin subtypes αvβ3 and α5β1 have been shown for various cellular processes; however, a systematic comparison of their function in adhesion-dependent chemoresistance is lacking. Here, we utilize integrin subtype-specific peptidomimetics for αvβ3 and α5β1, both as blocking agents on fibronectin-coated surfaces and as surface-immobilized adhesion sites, in order to parse out their role in breast cancer cell survival. Block copolymer micelle nanolithography is utilized to immobilize peptidomimetics onto highly ordered gold nanoparticle arrays with biologically relevant interparticle spacings (35, 50, or 70 nm), thereby providing a platform for ascertaining the dependence of ligand spacing in chemoprotection. We show that several cellular properties-morphology, focal adhesion formation, and migration-are intricately linked to both the integrin subtype and their nanospacing. Importantly, we show that chemotherapeutic drug sensitivity is highly dependent on both parameters, with smaller ligand spacing generally hindering survival. Furthermore, we identify ligand type-specific patterns of drug sensitivity, with enhanced chemosurvival when cells engage αvβ3 vs α5β1 on fibronectin; however, this is heavily reliant on nanoscale spacing, as the opposite is observed when ligands are spaced at 70 nm. These data imply that even nanoscale alterations in extracellular matrix properties have profound effects on cancer cell survival and can thus inform future therapies and drug testing platforms.
Collapse
Affiliation(s)
- Jennifer
L. Young
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, 69120 Heidelberg, Germany
- Department
of Biophysical Chemistry, Heidelberg University, 69120 Heidelberg, Germany
| | - Ximeng Hua
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, 69120 Heidelberg, Germany
- Department
of Biophysical Chemistry, Heidelberg University, 69120 Heidelberg, Germany
| | - Heidi Somsel
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, 69120 Heidelberg, Germany
- Department
of Biophysical Chemistry, Heidelberg University, 69120 Heidelberg, Germany
| | - Florian Reichart
- Department
of Chemistry, Technical University of Munich, 85748 Garching, Germany
| | - Horst Kessler
- Department
of Chemistry, Technical University of Munich, 85748 Garching, Germany
| | - Joachim P. Spatz
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, 69120 Heidelberg, Germany
- Department
of Biophysical Chemistry, Heidelberg University, 69120 Heidelberg, Germany
- E-mail: . Phone: +49 6221 486-420
| |
Collapse
|