1
|
Nwokoye PN, Abilez OJ. Blood vessels in a dish: the evolution, challenges, and potential of vascularized tissues and organoids. Front Cardiovasc Med 2024; 11:1336910. [PMID: 38938652 PMCID: PMC11210405 DOI: 10.3389/fcvm.2024.1336910] [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: 11/12/2023] [Accepted: 04/19/2024] [Indexed: 06/29/2024] Open
Abstract
Vascular pathologies are prevalent in a broad spectrum of diseases, necessitating a deeper understanding of vascular biology, particularly in overcoming the oxygen and nutrient diffusion limit in tissue constructs. The evolution of vascularized tissues signifies a convergence of multiple scientific disciplines, encompassing the differentiation of human pluripotent stem cells (hPSCs) into vascular cells, the development of advanced three-dimensional (3D) bioprinting techniques, and the refinement of bioinks. These technologies are instrumental in creating intricate vascular networks essential for tissue viability, especially in thick, complex constructs. This review provides broad perspectives on the past, current state, and advancements in key areas, including the differentiation of hPSCs into specific vascular lineages, the potential and challenges of 3D bioprinting methods, and the role of innovative bioinks mimicking the native extracellular matrix. We also explore the integration of biophysical cues in vascularized tissues in vitro, highlighting their importance in stimulating vessel maturation and functionality. In this review, we aim to synthesize these diverse yet interconnected domains, offering a broad, multidisciplinary perspective on tissue vascularization. Advancements in this field will help address the global organ shortage and transform patient care.
Collapse
Affiliation(s)
- Peter N. Nwokoye
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Oscar J. Abilez
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
- Division of Pediatric CT Surgery, Stanford University, Stanford, CA, United States
- Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, United States
- Bio-X Program, Stanford University, Stanford, CA, United States
| |
Collapse
|
2
|
Rathna RP, Kulandhaivel M. Advancements in wound healing: integrating biomolecules, drug delivery carriers, and targeted therapeutics for enhanced tissue repair. Arch Microbiol 2024; 206:199. [PMID: 38563993 DOI: 10.1007/s00203-024-03910-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/15/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024]
Abstract
Wound healing, a critical biological process vital for tissue restoration, has spurred a global market exceeding $15 billion for wound care products and $12 billion for scar treatment. Chronic wounds lead to delayed or impaired wound healing. Natural bioactive compounds, prized for minimal side effects, stand out as promising candidates for effective wound healing. In response, researchers are turning to nanotechnology, employing the encapsulation of these agents into drug delivery carriers. Drug delivery system will play a crucial role in enabling targeted delivery of therapeutic agents to promote tissue regeneration and address underlying issues such as inflammation, infection, and impaired angiogenesis in chronic wound healing. Drug delivery carriers offer distinct advantages, exhibiting a substantial ratio of surface area to volume and altered physical and chemical properties. These carriers facilitate sustained and controlled release, proving particularly advantageous for the extended process of wound healing, that typically comprise a diverse range of components, integrating both natural and synthetic polymers. Additionally, they often incorporate bioactive molecules. Despite their properties, including poor solubility, rapid degradation, and limited bioavailability, various natural bioactive agents face challenges in clinical applications. With a global research, emphasis on harnessing nanomaterial for wound healing application, this research overview engages advancing drug delivery technologies to augment the effectiveness of tissue regeneration using bioactive molecules. Recent progress in drug delivery has poised to enhance the therapeutic efficacy of natural compounds in wound healing applications.
Collapse
Affiliation(s)
- R Preethi Rathna
- Department of Microbiology, Karpagam Academy of Higher Education, Coimbatore, Tamilnadu, 641021, India
| | - M Kulandhaivel
- Department of Microbiology, Karpagam Academy of Higher Education, Coimbatore, Tamilnadu, 641021, India.
| |
Collapse
|
3
|
Shafiee A, Sun J, Ahmed IA, Phua F, Rossi GR, Lin CY, Souza-Fonseca-Guimaraes F, Wolvetang EJ, Brown J, Khosrotehrani K. Development of Physiologically Relevant Skin Organoids from Human Induced Pluripotent Stem Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304879. [PMID: 38044307 DOI: 10.1002/smll.202304879] [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: 06/09/2023] [Revised: 09/28/2023] [Indexed: 12/05/2023]
Abstract
The development of skin organs for studying developmental pathways, modeling diseases, or regenerative medicine purposes is a major endeavor in the field. Human induced pluripotent stem cells (hiPSCs) are successfully used to derive skin cells, but the field is still far from meeting the goal of creating skin containing appendages, such as hair follicles and sweat glands. Here, the goal is to generate skin organoids (SKOs) from human skin fibroblast or placental CD34+ cell-derived hiPSCs. With all three hiPSC lines, complex SKOs with stratified skin layers and pigmented hair follicles are generated with different efficacies. In addition, the hiPSC-derived SKOs develop sebaceous glands, touch-receptive Merkel cells, and more importantly eccrine sweat glands. Together, physiologically relevant skin organoids are developed by direct induction of embryoid body formation, along with simultaneous inactivation of transforming growth factor beta signaling, activation of fibroblast growth factor signaling, and inhibition of bone morphogenetic protein signaling pathways. The skin organoids created in this study can be used as valuable platforms for further research into human skin development, disease modeling, or reconstructive surgeries.
Collapse
Affiliation(s)
- Abbas Shafiee
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD, 4029, Australia
- Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Brisbane, QLD, 4029, Australia
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Jane Sun
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Imaan A Ahmed
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Felicia Phua
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Gustavo R Rossi
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Cheng-Yu Lin
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102, Australia
| | | | - Ernst J Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Jason Brown
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD, 4029, Australia
- Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Brisbane, QLD, 4029, Australia
| | - Kiarash Khosrotehrani
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD, 4029, Australia
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102, Australia
| |
Collapse
|
4
|
Wang X, Ma Y, Lu F, Chang Q. The diversified hydrogels for biomedical applications and their imperative roles in tissue regeneration. Biomater Sci 2023; 11:2639-2660. [PMID: 36790251 DOI: 10.1039/d2bm01486f] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Repair and regeneration of tissues after injury are complex pathophysiological processes. Microbial infection, malnutrition, and an ischemic and hypoxic microenvironment in the injured area can impede the typical healing cascade. Distinguished by biomimicry of the extracellular matrix, high aqueous content, and diverse functions, hydrogels have revolutionized clinical practices in tissue regeneration owing to their outstanding hydrophilicity, biocompatibility, and biodegradability. Various hydrogels such as smart hydrogels, nanocomposite hydrogels, and acellular matrix hydrogels are widely used for applications ranging from bench-scale to an industrial scale. In this review, some emerging hydrogels in the biomedical field are briefly discussed. The protective roles of hydrogels in wound dressings and their diverse biological effects on multiple tissues such as bone, cartilage, nerve, muscle, and adipose tissue are also discussed. The vehicle functions of hydrogels for chemicals and cell payloads are detailed. Additionally, this review emphasizes the particular characteristics of hydrogel products that promote tissue repair and reconstruction such as anti-infection, inflammation regulation, and angiogenesis.
Collapse
Affiliation(s)
- Xinhui Wang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 510515, China.
| | - Yuan Ma
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 510515, China.
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 510515, China.
| | - Qiang Chang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 510515, China.
| |
Collapse
|
5
|
Suvarnapathaki S, Nguyen A, Goulopoulos A, Camci-Unal G. Oxygen-Generating Scaffolds for Cardiac Tissue Engineering Applications. ACS Biomater Sci Eng 2023; 9:409-426. [PMID: 36469567 PMCID: PMC11416866 DOI: 10.1021/acsbiomaterials.2c00853] [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] [Indexed: 12/12/2022]
Abstract
Homogeneous vascularization of implanted tissue constructs can extend to 5 weeks, during which cell death can occur due to inadequate availability of oxygen. Researchers are engineering biomaterials that generate and release oxygen in a regulated manner, in an effort to overcome this hurdle. A main limitation of the existing oxygen-generating biomaterials is the uncontrolled release of oxygen, which is ultimately detrimental to the cells. This study demonstrates the incorporation of calcium peroxide (CaO2) within a hydrophobic polymer, polycaprolactone (PCL), to yield composite scaffolds with controlled oxygen release kinetics sustained over 5 weeks. Oxygen-generating microparticles coencapsulated with cardiomyocytes in a gelatin-based hydrogel matrix can serve as model systems for cardiac tissue engineering. Specifically, the results reveal that the oxygen-generating microspheres significantly improve the scaffold mechanical strength ranging from 5 kPa to 35 kPa, have an average scaffold pore size of 50-100 μm, swelling ratios of 33.3-29.8%, and degradation with 10-49% remaining mass at the end of a 48 h accelerated enzymatic degradation. The oxygen-generating scaffolds demonstrate improvement in cell viability, proliferation, and metabolic activity compared to the negative control group when cultured under hypoxia. Additionally, the optimized oxygen-generating constructs display no cytotoxicity or apoptosis. These oxygen-generating scaffolds can possibly assist the in vivo translation of cardiac tissue constructs.
Collapse
Affiliation(s)
- Sanika Suvarnapathaki
- Biomedical Engineering and Biotechnology Program, University of Massachusetts Lowell, One University Avenue, Lowell, Massachusetts 01854, United States
- Department of Chemical Engineering, University of Massachusetts Lowell, One University Avenue, Lowell, Massachusetts 01854, United States
| | - Angelina Nguyen
- Department of Chemical Engineering, University of Massachusetts Lowell, One University Avenue, Lowell, Massachusetts 01854, United States
| | - Anastasia Goulopoulos
- Department of Chemical Engineering, University of Massachusetts Lowell, One University Avenue, Lowell, Massachusetts 01854, United States
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts Lowell, One University Avenue, Lowell, Massachusetts 01854, United States
- Department of Surgery, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01605, United States
| |
Collapse
|
6
|
Neary M, Owen A, Olagunju A. A Holistic Review of the Preclinical Landscape for Long-Acting Anti-infective Drugs Using HIV as a Paradigm. Clin Infect Dis 2022; 75:S490-S497. [PMID: 36410386 PMCID: PMC10200324 DOI: 10.1093/cid/ciac685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Lack of predictive preclinical models is a key contributor to the steep attrition rate in drug development. Successful clinical translation may be higher for new chemical entities or existing approved drugs reformulated for long-acting (LA) administration if preclinical studies designed to identify any new uncertainties are predictive of human exposure and response. In this review, we present an overview of standard preclinical assessments deployed for LA formulations and delivery systems, using human immunodeficiency virus LA therapeutics preclinical development as a paradigm. Key progress in the preclinical development of novel LA antiretrovirals formulations and delivery systems are summarized, including bispecific broadly neutralizing monoclonal antibody and small molecule technologies for codelivery of multiple drugs with disparate solubility properties. There are new opportunities to take advantage of recent developments in tissue engineering and 3-dimensional in vitro modeling to advance preclinical modeling of anti-infective activity, developmental and reproductive toxicity assessment, and to apply quantitative modeling and simulation strategies. These developments are likely to drive the progression of more LA anti-infective drugs and multipurpose technologies into clinical development in the coming years.
Collapse
Affiliation(s)
- Megan Neary
- Department of Pharmacology and Therapeutics, Centre of Excellence for Long-acting Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, Merseyside, United Kingdom
| | - Andrew Owen
- Department of Pharmacology and Therapeutics, Centre of Excellence for Long-acting Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, Merseyside, United Kingdom
| | - Adeniyi Olagunju
- Department of Pharmacology and Therapeutics, Centre of Excellence for Long-acting Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, Merseyside, United Kingdom
| |
Collapse
|
7
|
Zhao L, Wang X. 3D printed microfluidics for cell biological applications. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
8
|
Microfluidic 3D Platform to Evaluate Endothelial Progenitor Cell Recruitment by Bioactive Materials. Acta Biomater 2022; 151:264-277. [PMID: 35981686 DOI: 10.1016/j.actbio.2022.08.019] [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: 02/02/2022] [Revised: 08/04/2022] [Accepted: 08/10/2022] [Indexed: 12/30/2022]
Abstract
Most of the conventional in vitro models to test biomaterial-driven vascularization are too simplistic to recapitulate the complex interactions taking place in the actual cell microenvironment, which results in a poor prediction of the in vivo performance of the material. However, during the last decade, cell culture models based on microfluidic technology have allowed attaining unprecedented levels of tissue biomimicry. In this work, we propose a microfluidic-based 3D model to evaluate the effect of bioactive biomaterials capable of releasing signalling cues (such as ions or proteins) in the recruitment of endogenous endothelial progenitor cells, a key step in the vascularization process. The usability of the platform is demonstrated using experimentally-validated finite element models and migration and proliferation studies with rat endothelial progenitor cells (rEPCs) and bone marrow-derived rat mesenchymal stromal cells (BM-rMSCs). As a proof of concept of biomaterial evaluation, the response of rEPCs to an electrospun composite made of polylactic acid with calcium phosphates nanoparticles (PLA+CaP) was compared in a co-culture microenvironment with BM-rMSC to a regular PLA control. Our results show a significantly higher rEPCs migration and the upregulation of several pro-inflammatory and proangiogenic proteins in the case of the PLA+CaP. The effects of osteopontin (OPN) on the rEPCs migratory response were also studied using this platform, suggesting its important role in mediating their recruitment to a calcium-rich microenvironment. This new tool could be applied to screen the capacity of a variety of bioactive scaffolds to induce vascularization and accelerate the preclinical testing of biomaterials. STATEMENT OF SIGNIFICANCE: : For many years researchers have used neovascularization models to evaluate bioactive biomaterials both in vitro, with low predictive results due to their poor biomimicry and minimal control over cell cues such as spatiotemporal biomolecule signaling, and in vivo models, presenting drawbacks such as being highly costly, time-consuming, poor human extrapolation, and ethically controversial. We describe a compact microphysiological platform designed for the evaluation of proangiogenesis in biomaterials through the quantification of the level of sprouting in a mimicked endothelium able to react to gradients of biomaterial-released signals in a fibrin-based extracellular matrix. This model is a useful tool to perform preclinical trustworthy studies in tissue regeneration and to better understand the different elements involved in the complex process of vascularization.
Collapse
|
9
|
Zhang K, Du Z, Yuan T, Huang J, Zhao X, Mi S. Long-term cultured microvascular networks on chip for tumor vascularization research and drug testing. BIOMICROFLUIDICS 2022; 16:044101. [PMID: 35845724 PMCID: PMC9282889 DOI: 10.1063/5.0090027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/13/2022] [Indexed: 06/15/2023]
Abstract
The vascular structure of the tumor microenvironment (TME) plays an essential role in the process of metastasis. In vitro microvascular structures that can be maintained for a long time will greatly promote metastasis research. In this study, we constructed a mimicking breast cancer invasion model based on a microfluidic chip platform, and the maintenance time of the self-assembled microvascular networks significantly improved by culturing with fibroblasts (up to 13 days). Using this model, we quantified the invasion ability of breast cancer cells and angiogenesis sprouts caused by cancer cells, and the intravasation behavior of cancer cells was also observed in sprouts. We found that cancer cells could significantly cause angiogenesis by promoting sprouting behaviors of the self-assembled human umbilical vein endothelial cells, which, in turn, promoted the invasion behavior of cancer cells. The drug test results showed that the drug resistance of the widely used anti-cancer drugs 5-Fluorouracil (5-FU) and Doxorubicin (DOX) in the 3D model was higher than that in the 2D model. Meanwhile, we also proved that 5-FU and DOX had the effect of destroying tumor blood vessels. The anti-angiogenic drug Apatinib (VEGFR inhibitor) enhanced the drug effect of DOX on MDA-MB-231 cells, further proving the promoting effect of angiogenesis on the invasion ability of cancer cells. These results indicate that our model is of great value in reconstructing TME and drug testing in vitro.
Collapse
Affiliation(s)
- Ke Zhang
- Open FIESTA Center, International Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Zhichang Du
- College of Mechanical and Energy Engineering, Jimei University, Xiamen, China
| | - Tianying Yuan
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, China
| | - Jiajun Huang
- Bio-manufacturing Engineering Laboratory, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Xiaoyu Zhao
- Open FIESTA Center, International Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Shengli Mi
- Author to whom correspondence should be addressed:
| |
Collapse
|
10
|
Introini V, Govendir MA, Rayner JC, Cicuta P, Bernabeu M. Biophysical Tools and Concepts Enable Understanding of Asexual Blood Stage Malaria. Front Cell Infect Microbiol 2022; 12:908241. [PMID: 35711656 PMCID: PMC9192966 DOI: 10.3389/fcimb.2022.908241] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/27/2022] [Indexed: 12/02/2022] Open
Abstract
Forces and mechanical properties of cells and tissues set constraints on biological functions, and are key determinants of human physiology. Changes in cell mechanics may arise from disease, or directly contribute to pathogenesis. Malaria gives many striking examples. Plasmodium parasites, the causative agents of malaria, are single-celled organisms that cannot survive outside their hosts; thus, thost-pathogen interactions are fundamental for parasite’s biological success and to the host response to infection. These interactions are often combinations of biochemical and mechanical factors, but most research focuses on the molecular side. However, Plasmodium infection of human red blood cells leads to changes in their mechanical properties, which has a crucial impact on disease pathogenesis because of the interaction of infected red blood cells with other human tissues through various adhesion mechanisms, which can be probed and modelled with biophysical techniques. Recently, natural polymorphisms affecting red blood cell biomechanics have also been shown to protect human populations, highlighting the potential of understanding biomechanical factors to inform future vaccines and drug development. Here we review biophysical techniques that have revealed new aspects of Plasmodium falciparum invasion of red blood cells and cytoadhesion of infected cells to the host vasculature. These mechanisms occur differently across Plasmodium species and are linked to malaria pathogenesis. We highlight promising techniques from the fields of bioengineering, immunomechanics, and soft matter physics that could be beneficial for studying malaria. Some approaches might also be applied to other phases of the malaria lifecycle and to apicomplexan infections with complex host-pathogen interactions.
Collapse
Affiliation(s)
- Viola Introini
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Viola Introini,
| | - Matt A. Govendir
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
| | - Julian C. Rayner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Pietro Cicuta
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Maria Bernabeu
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
| |
Collapse
|
11
|
Wang Y, Kankala RK, Ou C, Chen A, Yang Z. Advances in hydrogel-based vascularized tissues for tissue repair and drug screening. Bioact Mater 2022; 9:198-220. [PMID: 34820566 PMCID: PMC8586021 DOI: 10.1016/j.bioactmat.2021.07.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
The construction of biomimetic vasculatures within the artificial tissue models or organs is highly required for conveying nutrients, oxygen, and waste products, for improving the survival of engineered tissues in vitro. In recent times, the remarkable progress in utilizing hydrogels and understanding vascular biology have enabled the creation of three-dimensional (3D) tissues and organs composed of highly complex vascular systems. In this review, we give an emphasis on the utilization of hydrogels and their advantages in the vascularization of tissues. Initially, the significance of vascular elements and the regeneration mechanisms of vascularization, including angiogenesis and vasculogenesis, are briefly introduced. Further, we highlight the importance and advantages of hydrogels as artificial microenvironments in fabricating vascularized tissues or organs, in terms of tunable physical properties, high similarity in physiological environments, and alternative shaping mechanisms, among others. Furthermore, we discuss the utilization of such hydrogels-based vascularized tissues in various applications, including tissue regeneration, drug screening, and organ-on-chips. Finally, we put forward the key challenges, including multifunctionalities of hydrogels, selection of suitable cell phenotype, sophisticated engineering techniques, and clinical translation behind the development of the tissues with complex vasculatures towards their future development.
Collapse
Affiliation(s)
- Ying Wang
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, 523059, PR China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, Guangdong, 510080, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian, 361021, PR China
| | - Caiwen Ou
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, 523059, PR China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, Guangdong, 510080, PR China
| | - Aizheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian, 361021, PR China
| | - Zhilu Yang
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, 523059, PR China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, Guangdong, 510080, PR China
| |
Collapse
|
12
|
Wang W, Liu Y, Liu Z, Li S, Deng C, Yang X, Deng Q, Sun Y, Zhang Y, Ma Z, Li W, Liu Y, Zhou X, Li T, Zhu J, Wang J, Dai K. Evaluation of Interleukin-4-Loaded Sodium Alginate-Chitosan Microspheres for Their Support of Microvascularization in Engineered Tissues. ACS Biomater Sci Eng 2021; 7:4946-4958. [PMID: 34525809 DOI: 10.1021/acsbiomaterials.1c00882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Defects in the formation of microvascular networks, which provide oxygen and nutrients to cells, are the main reason for the engraftment failure of clinically applicable engineered tissues. Inflammatory responses and immunomodulation can promote the vascularization of the engineered tissues. We developed a capillary construct composed of a gelatin methacrylate-based cell-laden hydrogel framework complexed with interleukin-4 (IL-4)-loaded alginate-chitosan (AC) microspheres and endothelial progenitor cells (EPCs) and RAW264.7 macrophages as model cells. The AC microspheres maintained and guided the EPCs through electrostatic adhesion, facilitating the formation of microvascular networks. The IL-4-loaded microspheres promoted the polarization of the macrophages into the M2 type, leading to a reduction in pro-inflammatory factors and enhancement of the vascularization. Hematoxylin and eosin staining and immunohistochemical analysis revealed that, without IL-4 or AC microspheres, the scaffold was less effective in angiogenesis. We provide an alternative and promising approach for constructing vascularized tissues.
Collapse
Affiliation(s)
- Wenhao Wang
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.,Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yuehua Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Road, Zhengzhou 450052, People's Republic of China
| | - Zifan Liu
- School of Biological Science and Medical Engineering, BUAA, 37 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Shuai Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Changxu Deng
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiaoxiao Yang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qian Deng
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Yeye Sun
- Hunan Polytechnic of Environment and Biology, Hengyang 422000, China
| | - Yuxin Zhang
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zhenjiang Ma
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wentao Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yang Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiaojun Zhou
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Tao Li
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Junfeng Zhu
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jinwu Wang
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.,Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Kerong Dai
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.,Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
13
|
Shin DS, Anseth KS. Recent advances in 3D models of tumor invasion. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 19:100310. [PMID: 34308009 PMCID: PMC8294077 DOI: 10.1016/j.cobme.2021.100310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This review presents recent advances in the design of in vitro cancer models to study tumor cell migration, metastasis, and invasion in three-dimensions (3D). These cancer models are divided into two categories based on the biophysiological processes and structures simulated, namely (i) spheroid invasion models or (ii) vascularization models. Some recent advances to spheroid invasion models include new methods to make them amenable to high-throughput settings. In vascularization models, cancer cell extravasation, intravasation, and angiogenesis have been emulated. Finally, 3D bioprinting and microfluidic technologies are allowing researchers to recapitulate some of the complex architectural and microenvironmental changes that can drive cancer cells migration from the extracellular matrix and basement membrane to blood vessels.
Collapse
Affiliation(s)
- Della S. Shin
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA 80303
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA 80303
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA 80303
| |
Collapse
|
14
|
Matthiesen I, Voulgaris D, Nikolakopoulou P, Winkler TE, Herland A. Continuous Monitoring Reveals Protective Effects of N-Acetylcysteine Amide on an Isogenic Microphysiological Model of the Neurovascular Unit. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101785. [PMID: 34174140 DOI: 10.1002/smll.202101785] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/21/2021] [Indexed: 05/20/2023]
Abstract
Microphysiological systems mimic the in vivo cellular ensemble and microenvironment with the goal of providing more human-like models for biopharmaceutical research. In this study, the first such model of the blood-brain barrier (BBB-on-chip) featuring both isogenic human induced pluripotent stem cell (hiPSC)-derived cells and continuous barrier integrity monitoring with <2 min temporal resolution is reported. Its capabilities are showcased in the first microphysiological study of nitrosative stress and antioxidant prophylaxis. Relying on off-stoichiometry thiol-ene-epoxy (OSTE+) for fabrication greatly facilitates assembly and sensor integration compared to the prevalent polydimethylsiloxane devices. The integrated cell-substrate endothelial resistance monitoring allows for capturing the formation and breakdown of the BBB model, which consists of cocultured hiPSC-derived endothelial-like and astrocyte-like cells. Clear cellular disruption is observed when exposing the BBB-on-chip to the nitrosative stressor linsidomine, and the barrier permeability and barrier-protective effects of the antioxidant N-acetylcysteine amide are reported. Using metabolomic network analysis reveals further drug-induced changes consistent with prior literature regarding, e.g., cysteine and glutathione involvement. A model like this opens new possibilities for drug screening studies and personalized medicine, relying solely on isogenic human-derived cells and providing high-resolution temporal readouts that can help in pharmacodynamic studies.
Collapse
Affiliation(s)
- Isabelle Matthiesen
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Malvinas Väg 10 pl 5, Stockholm, 100 44, Sweden
| | - Dimitrios Voulgaris
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Malvinas Väg 10 pl 5, Stockholm, 100 44, Sweden
- AIMES, Center for Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Solnavägen 9/B8, Solna, 171 65, Sweden
| | - Polyxeni Nikolakopoulou
- AIMES, Center for Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Solnavägen 9/B8, Solna, 171 65, Sweden
| | - Thomas E Winkler
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Malvinas Väg 10 pl 5, Stockholm, 100 44, Sweden
| | - Anna Herland
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Malvinas Väg 10 pl 5, Stockholm, 100 44, Sweden
- AIMES, Center for Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Solnavägen 9/B8, Solna, 171 65, Sweden
| |
Collapse
|
15
|
Tronolone JJ, Jain A. Engineering new microvascular networks on-chip: ingredients, assembly, and best practices. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007199. [PMID: 33994903 PMCID: PMC8114943 DOI: 10.1002/adfm.202007199] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Indexed: 05/23/2023]
Abstract
Tissue engineered grafts show great potential as regenerative implants for diseased or injured tissues within the human body. However, these grafts suffer from poor nutrient perfusion and waste transport, thus decreasing their viability post-transplantation. Graft vascularization is therefore a major area of focus within tissue engineering because biologically relevant conduits for nutrient and oxygen perfusion can improve viability post-implantation. Many researchers utilize microphysiological systems as testing platforms for potential grafts due to an ability to integrate vascular networks as well as biological characteristics such as fluid perfusion, 3D architecture, compartmentalization of tissue-specific materials, and biophysical and biochemical cues. While many methods of vascularizing these systems exist, microvascular self-assembly has great potential for bench-to-clinic translation as it relies on naturally occurring physiological events. In this review, we highlight the past decade of literature and critically discuss the most important and tunable components yielding a self-assembled vascular network on chip: endothelial cell source, tissue-specific supporting cells, biomaterial scaffolds, biochemical cues, and biophysical forces. This article discusses the bioengineered systems of angiogenesis, vasculogenesis, and lymphangiogenesis, and includes a brief overview of multicellular systems. We conclude with future avenues of research to guide the next generation of vascularized microfluidic models and future tissue engineered grafts.
Collapse
Affiliation(s)
- James J Tronolone
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Abhishek Jain
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77808, USA
| |
Collapse
|
16
|
Agarwal T, Kazemi S, Costantini M, Perfeito F, Correia CR, Gaspar V, Montazeri L, De Maria C, Mano JF, Vosough M, Makvandi P, Maiti TK. Oxygen releasing materials: Towards addressing the hypoxia-related issues in tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111896. [PMID: 33641899 DOI: 10.1016/j.msec.2021.111896] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/09/2021] [Accepted: 01/16/2021] [Indexed: 02/07/2023]
Abstract
Manufacturing macroscale cell-laden architectures is one of the biggest challenges faced nowadays in the domain of tissue engineering. Such living constructs, in fact, pose strict requirements for nutrients and oxygen supply that can hardly be addressed through simple diffusion in vitro or without a functional vasculature in vivo. In this context, in the last two decades, a substantial amount of work has been carried out to develop smart materials that could actively provide oxygen-release to contrast local hypoxia in large-size constructs. This review provides an overview of the currently available oxygen-releasing materials and their synthesis and mechanism of action, highlighting their capacities under in vitro tissue cultures and in vivo contexts. Additionally, we also showcase an emerging concept, herein termed as "living materials as releasing systems", which relies on the combination of biomaterials with photosynthetic microorganisms, namely algae, in an "unconventional" attempt to supply the damaged or re-growing tissue with the necessary supply of oxygen. We envision that future advances focusing on tissue microenvironment regulated oxygen-supplying materials would unlock an untapped potential for generating a repertoire of anatomic scale, living constructs with improved cell survival, guided differentiation, and tissue-specific biofunctionality.
Collapse
Affiliation(s)
- Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Sara Kazemi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marco Costantini
- Institute of Physical Chemistry - Polish Academy of Sciences, Warsaw, Poland
| | - Francisca Perfeito
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Clara R Correia
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
| | - Vítor Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Carmelo De Maria
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Pooyan Makvandi
- Center for MicroBioRobotics (CMBR), Istituto Italiano di Tecnologia, Pisa, Italy
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| |
Collapse
|
17
|
Parlato S, Grisanti G, Sinibaldi G, Peruzzi G, Casciola CM, Gabriele L. Tumor-on-a-chip platforms to study cancer-immune system crosstalk in the era of immunotherapy. LAB ON A CHIP 2021; 21:234-253. [PMID: 33315027 DOI: 10.1039/d0lc00799d] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Immunotherapy is a powerful therapeutic approach able to re-educate the immune system to fight cancer. A key player in this process is the tumor microenvironment (TME), which is a dynamic entity characterized by a complex array of tumor and stromal cells as well as immune cell populations trafficking to the tumor site through the endothelial barrier. Recapitulating these multifaceted dynamics is critical for studying the intimate interactions between cancer and the immune system and to assess the efficacy of emerging immunotherapies, such as immune checkpoint inhibitors (ICIs) and adoptive cell-based products. Microfluidic devices offer a unique technological approach to build tumor-on-a-chip reproducing the multiple layers of complexity of cancer-immune system crosstalk. Here, we seek to review the most important biological and engineering developments of microfluidic platforms for studying cancer-immune system interactions, in both solid and hematological tumors, highlighting the role of the vascular component in immune trafficking. Emphasis is given to image processing and related algorithms for real-time monitoring and quantitative evaluation of the cellular response to microenvironmental dynamic changes. The described approaches represent a valuable tool for preclinical evaluation of immunotherapeutic strategies.
Collapse
Affiliation(s)
- Stefania Parlato
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, Italy.
| | | | | | | | | | | |
Collapse
|
18
|
Jin X, Shang Y, Zou Y, Xiao M, Huang H, Zhu S, Liu N, Li J, Wang W, Zhu P. Injectable Hypoxia-Induced Conductive Hydrogel to Promote Diabetic Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2020; 12:56681-56691. [PMID: 33274927 DOI: 10.1021/acsami.0c13197] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Injectable hydrogels with the capability to cast a hypoxic microenvironment is of great potentialities to develop novel therapies for tissue regeneration. However, the relative research still remains at the conceptual phase. Herein, we chose diabetic wound as a representative injury model to explore the actual therapeutic results of tissue injury by injectable hypoxia-induced hydrogels. To enhance recovery and widen applicability, the hypoxia-induced system was incorporated with a conductive network by an original sequentially interpenetrating technique based on the combination of a fast "click chemistry" and a slow enzymatic mediated cross-linking. Hyperbranched poly(β-amino ester)-tetraaniline (PBAE-TA) was cross-linked with thiolated hyaluronic acid (HA-SH) via a thiol-ene click reaction, contributing to the rapid formation of the first conductive network, where vanillin-grafted gelatin (Geln-Van) and laccase (Lac) with a slow cross-linking rate were employed in casting a hypoxic microenvironment. The as-prepared injectable hydrogels possessed both suitable conductivity and sustainable hypoxia-inducing capability to upregulate the hypoxia-inducible factor-1α and connexin 43 expressions of the encapsulated adipose-derived stem cells, which enhanced vascular regeneration and immunoregulation and further promoted the reconstruction of blood vessels, hair follicles, and dermal collagen matrix, eventually leading to the recovery of diabetic rat skin wounds and restoration of skin functions. This work provides a promising strategy to broaden the applicability of diverse hydrogels with a long time-consuming gelation process and to integrate different networks with various biological functions for the therapies of diabetic wounds and other complex clinical symptoms.
Collapse
Affiliation(s)
- Xin Jin
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300350, China
| | - Yingying Shang
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300350, China
| | - Yang Zou
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300350, China
| | - Meng Xiao
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300350, China
| | - Huanlei Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Shuoji Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Nanbo Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Jiani Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| | - Wei Wang
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300350, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
| |
Collapse
|
19
|
Bai J, Haase K, Roberts JJ, Hoffmann J, Nguyen HT, Wan Z, Zhang S, Sarker B, Friedman N, Ristić-Lehmann Č, Kamm RD. A novel 3D vascular assay for evaluating angiogenesis across porous membranes. Biomaterials 2020; 268:120592. [PMID: 33348261 DOI: 10.1016/j.biomaterials.2020.120592] [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: 07/26/2020] [Revised: 11/24/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023]
Abstract
Microfluidic technology has been extensively applied to model the functional units of human organs and tissues. Since vasculature is a key component of any functional tissue, a variety of techniques to mimic vasculature in vitro have been developed to address complex physiological and pathological processes in 3D tissues. Herein, we developed a novel, in vitro, microfluidic-based model to probe microvasculature growth into and across implanted porous membranes. Using ePTFE and polycarbonate as examples, we characterize the vascularization potential of these thin porous membranes using this device. This tool will allow for the assessment of porous materials early in their development, prior to their use for encapsulating implants or drugs, while minimizing the need for animal studies. Employing quantitative morphometric analysis and measurements of vascular permeability, we demonstrate our model to be an effective platform for evaluation of angiogenic potential of an implanted membrane biomaterial. Results show that endothelial cells can either migrate as single cells or form continuous sprouts across porous membranes, which is a material structure-dependent behavior. Our model is advantageous over conventional Transwell assays as it is amenable to quantitative assessment of vascular sprouting in 3D, and in contrast to animal models it can be employed more efficiently and with real-time assessment capabilities. This new tool could be applied either to test the suitability of a wide range of biomaterials for implantation or to screen different pro-angiogenic factors for therapeutic applications, and will advance the design of new biomaterials.
Collapse
Affiliation(s)
- Jing Bai
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kristina Haase
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Justine J Roberts
- W. L. Gore & Associates, Inc., Flagstaff, AZ, 86004/Cambridge, MA, 02142, USA
| | - Joseph Hoffmann
- W. L. Gore & Associates, Inc., Flagstaff, AZ, 86004/Cambridge, MA, 02142, USA
| | - Huu Tuan Nguyen
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Zhengpeng Wan
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Shun Zhang
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Bapi Sarker
- W. L. Gore & Associates, Inc., Flagstaff, AZ, 86004/Cambridge, MA, 02142, USA
| | - Nathan Friedman
- W. L. Gore & Associates, Inc., Flagstaff, AZ, 86004/Cambridge, MA, 02142, USA
| | | | - Roger D Kamm
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
20
|
Allwardt V, Ainscough AJ, Viswanathan P, Sherrod SD, McLean JA, Haddrick M, Pensabene V. Translational Roadmap for the Organs-on-a-Chip Industry toward Broad Adoption. Bioengineering (Basel) 2020; 7:E112. [PMID: 32947816 PMCID: PMC7552662 DOI: 10.3390/bioengineering7030112] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/09/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022] Open
Abstract
Organs-on-a-Chip (OOAC) is a disruptive technology with widely recognized potential to change the efficiency, effectiveness, and costs of the drug discovery process; to advance insights into human biology; to enable clinical research where human trials are not feasible. However, further development is needed for the successful adoption and acceptance of this technology. Areas for improvement include technological maturity, more robust validation of translational and predictive in vivo-like biology, and requirements of tighter quality standards for commercial viability. In this review, we reported on the consensus around existing challenges and necessary performance benchmarks that are required toward the broader adoption of OOACs in the next five years, and we defined a potential roadmap for future translational development of OOAC technology. We provided a clear snapshot of the current developmental stage of OOAC commercialization, including existing platforms, ancillary technologies, and tools required for the use of OOAC devices, and analyze their technology readiness levels. Using data gathered from OOAC developers and end-users, we identified prevalent challenges faced by the community, strategic trends and requirements driving OOAC technology development, and existing technological bottlenecks that could be outsourced or leveraged by active collaborations with academia.
Collapse
Affiliation(s)
- Vanessa Allwardt
- Center for Innovative Technology, Department of Chemistry, Vanderbilt University, Nashville, TN 37212, USA; (V.A.); (S.D.S.); (J.A.M.)
| | | | - Priyalakshmi Viswanathan
- Medicines Discovery Catapult, Alderley Park, Alderley Edge, Macclesfield SK10 4TG, UK; (P.V.); (M.H.)
| | - Stacy D. Sherrod
- Center for Innovative Technology, Department of Chemistry, Vanderbilt University, Nashville, TN 37212, USA; (V.A.); (S.D.S.); (J.A.M.)
| | - John A. McLean
- Center for Innovative Technology, Department of Chemistry, Vanderbilt University, Nashville, TN 37212, USA; (V.A.); (S.D.S.); (J.A.M.)
- Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, USA
| | - Malcolm Haddrick
- Medicines Discovery Catapult, Alderley Park, Alderley Edge, Macclesfield SK10 4TG, UK; (P.V.); (M.H.)
| | - Virginia Pensabene
- School of Electronic and Electrical Engineering, School of Medicine, Leeds Institute of Medical Research at St. James’s, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
21
|
Bordoni B. The Shape and Function of Solid Fascias Depend on the Presence of Liquid Fascias. Cureus 2020; 12:e6939. [PMID: 32190491 PMCID: PMC7067346 DOI: 10.7759/cureus.6939] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 02/10/2020] [Indexed: 12/21/2022] Open
Abstract
Scientific research is not a showcase of his own talent or own resources, it is a chance to improve common knowledge on certain topics for the collective well-being. A researcher should use multidisciplinarity to observe a phenomenon in its entirety and not only its alignment of thought, federations, committees, and knowledge; to get to understand it is necessary to exploit more tools and more disciplines. The article discusses the importance of the fluids (or liquid fascia) in maintaining the shape and function of the human body, as, currently, many texts forget how much body fluids are fundamental for understanding structural dynamics (bones and muscles, fibrils, and cells). By revisiting the current literature, the text wishes to highlight how the liquid fascia determines body adaptation in the presence of mechanical stress. Without fluids, there would be no body shape that we know.
Collapse
Affiliation(s)
- Bruno Bordoni
- Physical Medicine and Rehabilitation, Foundation Don Carlo Gnocchi, Milan, ITA
| |
Collapse
|