1
|
Yang J, Kim K, Liu Y, Luo X, Ma C, Man W, Zhao Y, Cao Z, Hu P, Chen J, Wang Y, Sun X, Zhao L, Wang G, Yang K, Wang X. 3D bioprinted dynamic bioactive living construct enhances mechanotransduction-assisted rapid neural network self-organization for spinal cord injury repair. Bioact Mater 2025; 46:531-554. [PMID: 39886605 PMCID: PMC11780150 DOI: 10.1016/j.bioactmat.2024.12.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/17/2024] [Accepted: 12/27/2024] [Indexed: 02/01/2025] Open
Abstract
Biomimetic neural substitutes, constructed through the bottom-up assembly of cell-matrix modulus via 3D bioprinting, hold great promise for neural regeneration. However, achieving precise control over the fate of neural stem cells (NSCs) to ensure biological functionality remains challenging. Cell behaviors are closely linked to cellular dynamics and cell-matrix mechanotransduction within a 3D microenvironment. To address this, a dynamic bioactive bioink is designed to provide adaptable biomechanics and instructive biochemical cues, specifically tailored for the fate commitment of NSCs, through incorporating reversible Schiff-base bonds and bioactive motifs, N-cadherin-mimicking and BDNF-mimicking peptides. We demonstrate that the dynamic properties of 3D bioprinted living fibers alleviate the mechanical confinement on NSCs and significantly enhance their mechanosensing, spreading, migration, and matrix remodeling within the 3D matrix. Additionally, the inclusion of N-cadherin-mimicking and BDNF-mimicking peptides further enhances cells' ability to sense and respond to mechanical and neurotrophic cues provided by the surrounding matrix, which accelerates the self-organization of a functional neural network within the 3D bioprinted construct, leading to significant motor and sensory function recovery in a rat complete spinal cord injury model. This work underscores the critical role of precisely designing cell-instructive bioinks for the advanced functionality of 3D bioprinted living constructs in neural regeneration.
Collapse
Affiliation(s)
- Jia Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Kunkoo Kim
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Yaosai Liu
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Xiaobin Luo
- Department of Orthopedics, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Chao Ma
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Weitao Man
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Yating Zhao
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Zheng Cao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
- Center for Biomaterials and Regenerative Medicine, Wuzhen Laboratory, Tongxiang 314500, China
| | - Peilun Hu
- Department of Orthopedics, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Junlin Chen
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Yu Wang
- Department of Orthopedics, Peking University First Hospital, Beijing 100034, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Guihuai Wang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Kaiyuan Yang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
2
|
Xu H, Zhang S, Song K, Yang H, Yin J, Huang Y. Droplet-based 3D bioprinting for drug delivery and screening. Adv Drug Deliv Rev 2025; 217:115486. [PMID: 39667692 DOI: 10.1016/j.addr.2024.115486] [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: 08/30/2024] [Revised: 12/01/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Recently, the conventional criterion of "one-size-fits-all" is not qualified for each individual patient, requiring precision medicine for enhanced therapeutic effects. Besides, drug screening is a high-cost and time-consuming process which requires innovative approaches to facilitate drug development rate. Benefiting from consistent technical advances in 3D bioprinting techniques, droplet-based 3D bioprinting techniques have been broadly utilized in pharmaceutics due to the noncontact printing mechanism and precise control on the deposition position of droplets. More specifically, cell-free/cell-laden bioinks which are deposited for the fabrication of drug carriers/3D tissue constructs have been broadly utilized for precise drug delivery and high throughput drug screening, respectively. This review summarizes the mechanism of various droplet-based 3D bioprinting techniques and the most up-to-date applications in drug delivery and screening and discusses the potential improvements of droplet-based 3D bioprinting techniques from both technical and material aspects. Through technical innovations, materials development, and the assistance from artificial intelligence, the formation process of drug carriers will be more stable and accurately controlled guaranteeing precise drug delivery. Meanwhile, the shape fidelity and uniformity of the printed tissue models will be significantly improved ensuring drug screening efficiency and efficacy.
Collapse
Affiliation(s)
- Heqi Xu
- The State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, China
| | - Shaokun Zhang
- The State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, China
| | | | - Huayong Yang
- The State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, China
| | - Jun Yin
- The State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, China.
| | - Yong Huang
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
3
|
Zhu S, Zhou Z, Chen X, Zhu W, Yang M, Yu M, Sun J, Zuo Y, He J, Pan H, Liu H. High mechanical performance and multifunctional degraded fucoidan-derived bioink for 3D bioprinting. Carbohydr Polym 2025; 348:122805. [PMID: 39562080 DOI: 10.1016/j.carbpol.2024.122805] [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: 07/20/2024] [Revised: 09/06/2024] [Accepted: 09/24/2024] [Indexed: 11/21/2024]
Abstract
While 3D bioprinting serves as a powerful tool in the field of tissue engineering, there is still a lack of natural biomaterial inks that simultaneously combine high mechanical performance with multiple biofunctionalities. Here, a single-component natural bioink with high strength and multi-biofunctionality was developed through the simple degradation and methacrylation of natural fucoidan. Hydrothermal degradation significantly decreased the natural fucoidan solution's viscosity by 99.9 %, meeting the necessary viscosity for Digital Light Processing (DLP) 3D printing. Meanwhile, various biofunctionalities of low molecular weight fucoidan obtained through degradation, such as antimicrobial and antioxidant properties, were developed. The resulting bioink exhibited good mechanical performance (compression modulus of 311 kPa), antimicrobial properties (antibacterial rates of 95.5 % and 97.9 % against E. coli and S. aureus, respectively), and antioxidant properties (intracellular ROS inhibition rates of 94.7 %). Using DLP 3D bioprinting, all printed products showed high shape fidelity with exceptional viability and activity of the encapsulated cells. Due to the unique sulfate structure resembling the natural components of chondroitin sulfate, the in vivo tests revealed its efficacy in promoting cartilage defect repair. In conclusion, the novel bioink blending high mechanical performance with multiple biofunctionalities, shows great potential in the 3D printing of tissue and organ regeneration.
Collapse
Affiliation(s)
- Shuai Zhu
- College of Material Science and Engineering, Hunan University, Changsha 410082, China
| | - Zheng Zhou
- College of Biology, Hunan University, Changsha 410082, China.
| | - Xin Chen
- College of Material Science and Engineering, Hunan University, Changsha 410082, China
| | - Wenxiang Zhu
- College of Material Science and Engineering, Hunan University, Changsha 410082, China
| | - Mengni Yang
- College of Material Science and Engineering, Hunan University, Changsha 410082, China
| | - Mengyi Yu
- College of Material Science and Engineering, Hunan University, Changsha 410082, China
| | - Jingjing Sun
- College of Biology, Hunan University, Changsha 410082, China
| | - You Zuo
- College of Biology, Hunan University, Changsha 410082, China
| | - Jiaqian He
- College of Biology, Hunan University, Changsha 410082, China
| | - Haobo Pan
- Shenzhen Key Laboratory of Marine Biomaterials, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Hairong Liu
- College of Material Science and Engineering, Hunan University, Changsha 410082, China.
| |
Collapse
|
4
|
Halper J. Narrative Review and Guide: State of the Art and Emerging Opportunities of Bioprinting in Tissue Regeneration and Medical Instrumentation. Bioengineering (Basel) 2025; 12:71. [PMID: 39851345 PMCID: PMC11760465 DOI: 10.3390/bioengineering12010071] [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: 12/03/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/26/2025] Open
Abstract
Three-dimensional printing was introduced in the 1980s, though bioprinting started developing a few years later. Today, 3D bioprinting is making inroads in medical fields, including the production of biomedical supplies intended for internal use, such as biodegradable staples. Medical bioprinting enables versatility and flexibility on demand and is able to modify and individualize production using several established printing methods. A great selection of biomaterials and bioinks is available, including natural, synthetic, and mixed options; they are biocompatible and non-toxic. Many bioinks are biodegradable and they accommodate cells so upon implantation, they integrate within the new environment. Bioprinting is suitable for printing tissues using living or viable components, such as collagen scaffolding, cartilage components, and cells, and also for printing parts of structures, such as teeth, using artificial man-made materials that will become embedded in vivo. Bioprinting is an integral part of tissue engineering and regenerative medicine. The addition of newly developed smart biomaterials capable of incorporating dynamic changes in shape depending on the nature of stimuli led to the addition of the fourth dimension of time in the form of changing shape to the three static dimensions. Four-dimensional bioprinting is already making significant inroads in tissue engineering and regenerative medicine, including new ways to create dynamic tissues. Its future lies in constructing partial or whole organ generation.
Collapse
Affiliation(s)
- Jaroslava Halper
- Department of Pathology, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
5
|
Moss SP, Bakirci E, Feinberg AW. Engineering the 3D structure of organoids. Stem Cell Reports 2025; 20:102379. [PMID: 39706178 PMCID: PMC11784486 DOI: 10.1016/j.stemcr.2024.11.009] [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: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/23/2024] Open
Abstract
Organoids form through the sel f-organizing capabilities of stem cells to produce a variety of differentiated cell and tissue types. Most organoid models, however, are limited in terms of the structure and function of the tissues that form, in part because it is difficult to regulate the cell type, arrangement, and cell-cell/cell-matrix interactions within these systems. In this article, we will discuss the engineering approaches to generate more complex organoids with improved function and translational relevance, as well as their advantages and disadvantages. Additionally, we will explore how biofabrication strategies can manipulate the cell composition, 3D organization, and scale-up of organoids, thus improving their utility for disease modeling, drug screening, and regenerative medicine applications.
Collapse
Affiliation(s)
- Samuel P Moss
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Ezgi Bakirci
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Adam W Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA; Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Yang J, Li W, Zhang Z, Xu Z, Zhu W, Wang J, Wang W. Development and Applications of Organoids in Gynecological Diseases. Stem Cell Rev Rep 2024:10.1007/s12015-024-10833-0. [PMID: 39666266 DOI: 10.1007/s12015-024-10833-0] [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] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Organoids are rapidly self-organizing 3D in vitro cultures derived from pluripotent stem cells (PSCs) or adult stem cells (ASCs) that possess disease-like characteristics with high success rates. Due to their ability to retain tissue structure, biological phenotypes, and genetic information, they have been utilized as a novel in vitro model for disease research. In recent years, scientists have established self-organizing 3D organoids for human endometrium, fallopian tubes, ovaries, and cervix by culturing stem cells with cytokines in 3D scaffolds. The integration of organoids with animal models, organ-on-a-chip systems, and 3D printing technologies offers a novel preclinical model for exploring disease mechanisms and developing treatments. This review elaborate on the recent research progress of stem cells-formed organoids in the field of gynecology from the aspects of constructing gynecological disease organoids, drug screening and new drug development, simulation modeling, allogeneic transplantation, regenerative medicine and personalized treatment."
Collapse
Affiliation(s)
- Jian Yang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wenwen Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zihan Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhonglei Xu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wenjing Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Anhui Women and Children's Medical Center, Hefei, Anhui, China
| | - Wenyan Wang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
7
|
Tonk M, Gupta V, Dhwaj A, Sachdeva M. Current developments and advancements of 3-dimensional printing in personalized medication and drug screening. Drug Metab Pers Ther 2024; 39:167-182. [PMID: 39331538 DOI: 10.1515/dmpt-2024-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/06/2024] [Indexed: 09/29/2024]
Abstract
INTRODUCTION 3-Dimensional printing (3DP) is an additive manufacturing (AM) technique that is expanding quickly because of its low cost and excellent efficiency. The 3D printing industry grew by 19.5 % in 2021 in spite of the COVID-19 epidemic, and by 2026, the worldwide market is expected to be valued up to 37.2 billion US dollars. CONTENT Science Direct, Scopus, MEDLINE, EMBASE, PubMed, DOAJ, and other academic databases provide evidence of the increased interest in 3DP technology and innovative drug delivery approaches in recent times. SUMMARY In this review four main 3DP technologies that are appropriate for pharmaceutical applications: extrusion-based, powder-based, liquid-based, and sheet lamination-based systems are discussed. This study is focused on certain 3DP technologies that may be used to create dosage forms, pharmaceutical goods, and other items with broad regulatory acceptance and technological viability for use in commercial manufacturing. It also discusses pharmaceutical applications of 3DP in drug delivery and drug screening. OUTLOOK The pharmaceutical sector has seen the prospect of 3D printing in risk assessment, medical personalisation, and the manufacture of complicated dose formulas at a reasonable cost. AM has great promise to revolutionise the manufacturing and use of medicines, especially in the field of personalized medicine. The need to understand more about the potential applications of 3DP in medical and pharmacological contexts has grown over time.
Collapse
Affiliation(s)
- Megha Tonk
- Raj Kumar Goel Institute of Technology (Pharmacy), Ghaziabad, Uttar Pradesh, India
| | - Vishal Gupta
- Raj Kumar Goel Institute of Technology (Pharmacy), Ghaziabad, Uttar Pradesh, India
| | | | - Monika Sachdeva
- Raj Kumar Goel Institute of Technology (Pharmacy), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
8
|
Yang J, Fischer NG, Ye Z. Revolutionising oral organoids with artificial intelligence. BIOMATERIALS TRANSLATIONAL 2024; 5:372-389. [PMID: 39872928 PMCID: PMC11764189 DOI: 10.12336/biomatertransl.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/20/2024] [Accepted: 11/01/2024] [Indexed: 01/30/2025]
Abstract
The convergence of organoid technology and artificial intelligence (AI) is poised to revolutionise oral healthcare. Organoids - three-dimensional structures derived from human tissues - offer invaluable insights into the complex biology of diseases, allowing researchers to effectively study disease mechanisms and test therapeutic interventions in environments that closely mimic in vivo conditions. In this review, we first present the historical development of organoids and delve into the current types of oral organoids, focusing on their use in disease models, regeneration and microbiome intervention. We then compare single-source and multi-lineage oral organoids and assess the latest progress in bioprinted, vascularised and neural-integrated organoids. In the next part of the review, we highlight significant advancements in AI, emphasising how AI algorithms may potentially promote organoid development for early disease detection and diagnosis, personalised treatment, disease prediction and drug screening. However, our main finding is the identification of remaining challenges, such as data integration and the critical need for rigorous validation of AI algorithms to ensure their clinical reliability. Our main viewpoint is that current AI-enabled oral organoids are still limited in applications but, as we look to the future, we offer insights into the potential transformation of AI-integrated oral organoids in oral disease diagnosis, oral microbial interactions and drug discoveries. By synthesising these components, this review aims to provide a comprehensive perspective on the current state and future implications of AI-enabled oral organoids, emphasising their role in advancing oral healthcare and improving patient outcomes.
Collapse
Affiliation(s)
- Jiawei Yang
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Nicholas G. Fischer
- MDRCBB, Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, MN, USA
| | - Zhou Ye
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
9
|
Al Monla R, Daien V, Michon F. Advanced bioengineering strategies broaden the therapeutic landscape for corneal failure. Front Bioeng Biotechnol 2024; 12:1480772. [PMID: 39605752 PMCID: PMC11598527 DOI: 10.3389/fbioe.2024.1480772] [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: 08/14/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
The cornea acts as the eye foremost protective layer and is essential for its focusing power. Corneal blindness may arise from physical trauma or conditions like dystrophies, keratitis, keratoconus, or ulceration. While conventional treatments involve medical therapies and donor allografts-sometimes supplemented with keratoprostheses-these options are not suitable for all corneal defects. Consequently, the development of bioartificial corneal tissue has emerged as a critical research area, aiming to address the global shortage of human cornea donors. Bioengineered corneas hold considerable promise as substitutes, with the potential to replace either specific layers or the entire thickness of damaged corneas. This review first delves into the structural anatomy of the human cornea, identifying key attributes necessary for successful corneal tissue bioengineering. It then examines various corneal pathologies, current treatments, and their limitations. Finally, the review outlines the primary approaches in corneal tissue engineering, exploring cell-free, cell-based, and scaffold-based options as three emerging strategies to address corneal failure.
Collapse
Affiliation(s)
- Reem Al Monla
- Institute for Neurosciences of Montpellier, INSERM, University of Montpellier, Montpellier, France
| | - Vincent Daien
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
- Sydney Medical School, The Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Frederic Michon
- Institute for Neurosciences of Montpellier, INSERM, University of Montpellier, Montpellier, France
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
| |
Collapse
|
10
|
Elkordy AA, Hill D, Attia M, Chaw CS. Liposomes and Their Therapeutic Applications in Enhancing Psoriasis and Breast Cancer Treatments. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1760. [PMID: 39513840 PMCID: PMC11547384 DOI: 10.3390/nano14211760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Psoriasis and breast cancer are two examples of diseases where associated inflammatory pathways within the body's immune system are implicated. Psoriasis is a complex, chronic and incurable inflammatory skin disorder that is primarily recognized by thick, scaly plaques on the skin. The most noticeable pathophysiological effect of psoriasis is the abnormal proliferation of keratinocytes. Breast cancer is currently the most diagnosed cancer and the leading cause of cancer-related death among women globally. While treatments targeting the primary tumor have significantly improved, preventing metastasis with systemic treatments is less effective. Nanocarriers such as liposomes and lipid nanoparticles have emerged as promising drug delivery systems for drug targeting and specificity. Advances in technologies and drug combinations have emerged to develop more efficient lipid nanocarriers to include more than one drug in combinational therapy to enhance treatment outcomes and/or relief symptoms for better patients' quality of life. Although there are FDA-approved liposomes with anti-cancer drugs for breast cancer, there are still unmet clinical needs to reduce the side effects associated with those nanomedicines. Hence, combinational nano-therapy may eliminate some of the issues and challenges. Furthermore, there are no nanomedicines yet clinically available for psoriasis. Hence, this review will focus on liposomes encapsulated single and/or combinational therapy to augment treatment outcomes with an emphasis on the effectiveness of combinational therapy within liposomal-based nanoparticulate drug delivery systems to tackle psoriasis and breast cancer. This review will also include an overview of both diseases, challenges in delivering drug therapy and the roles of nanomedicines as well as psoriasis and breast cancer models used for testing therapeutic interventions to pave the way for effective in vivo testing prior to the clinical trials.
Collapse
Affiliation(s)
- Amal Ali Elkordy
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| | - David Hill
- School of Nursing and Health Sciences, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK;
| | - Mohamed Attia
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| | - Cheng Shu Chaw
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| |
Collapse
|
11
|
Ucci A, Giacchi L, Rucci N. Primary Bone Tumors and Breast Cancer-Induced Bone Metastases: In Vivo Animal Models and New Alternative Approaches. Biomedicines 2024; 12:2451. [PMID: 39595017 PMCID: PMC11591690 DOI: 10.3390/biomedicines12112451] [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: 07/03/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Bone is the preferential site of metastasis for the most common tumors, including breast cancer. On the other hand, osteosarcoma is the primary bone cancer that most commonly occurs and causes bone cancer-related deaths in children. Several treatment strategies have been developed so far, with little or no efficacy for patient survival and with the development of side effects. Therefore, there is an urgent need to develop more effective therapies for bone primary tumors and bone metastatic disease. This almost necessarily requires the use of in vivo animal models that better mimic human pathology and at the same time follow the ethical principles for the humane use of animal testing. In this review we aim to illustrate the main and more suitable in vivo strategies employed to model bone metastases and osteosarcoma. We will also take a look at the recent technologies implemented for a partial replacement of animal testing.
Collapse
Affiliation(s)
| | | | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (A.U.); (L.G.)
| |
Collapse
|
12
|
Mancuso S, Bhalerao A, Cucullo L. Advances and Challenges of Bioassembly Strategies in Neurovascular In Vitro Modeling: An Overview of Current Technologies with a Focus on Three-Dimensional Bioprinting. Int J Mol Sci 2024; 25:11000. [PMID: 39456783 PMCID: PMC11506837 DOI: 10.3390/ijms252011000] [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: 09/18/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Bioassembly encompasses various techniques such as bioprinting, microfluidics, organoids, and self-assembly, enabling advances in tissue engineering and regenerative medicine. Advancements in bioassembly technologies have enabled the precise arrangement and integration of various cell types to more closely mimic the complexity functionality of the neurovascular unit (NVU) and that of other biodiverse multicellular tissue structures. In this context, bioprinting offers the ability to deposit cells in a spatially controlled manner, facilitating the construction of interconnected networks. Scaffold-based assembly strategies provide structural support and guidance cues for cell growth, enabling the formation of complex bio-constructs. Self-assembly approaches utilize the inherent properties of cells to drive the spontaneous organization and interaction of neuronal and vascular components. However, recreating the intricate microarchitecture and functional characteristics of a tissue/organ poses additional challenges. Advancements in bioassembly techniques and materials hold great promise for addressing these challenges. The further refinement of bioprinting technologies, such as improved resolution and the incorporation of multiple cell types, can enhance the accuracy and complexity of the biological constructs; however, developing bioinks that support the growth of cells, viability, and functionality while maintaining compatibility with the bioassembly process remains an unmet need in the field, and further advancements in the design of bioactive and biodegradable scaffolds will aid in controlling cell adhesion, differentiation, and vascularization within the engineered tissue. Additionally, integrating advanced imaging and analytical techniques can provide real-time monitoring and characterization of bioassembly, aiding in quality control and optimization. While challenges remain, ongoing research and technological advancements propel the field forward, paving the way for transformative developments in neurovascular research and tissue engineering. This work provides an overview of the advancements, challenges, and future perspectives in bioassembly for fabricating neurovascular constructs with an add-on focus on bioprinting technologies.
Collapse
Affiliation(s)
- Salvatore Mancuso
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA; (S.M.); (A.B.)
| | - Aditya Bhalerao
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA; (S.M.); (A.B.)
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, 586 Pioneer Dr, 460 O’Dowd Hall, Rochester, MI 48309, USA
| |
Collapse
|
13
|
Park G, Rim YA, Sohn Y, Nam Y, Ju JH. Replacing Animal Testing with Stem Cell-Organoids : Advantages and Limitations. Stem Cell Rev Rep 2024; 20:1375-1386. [PMID: 38639829 PMCID: PMC11319430 DOI: 10.1007/s12015-024-10723-5] [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] [Accepted: 04/08/2024] [Indexed: 04/20/2024]
Abstract
Various groups including animal protection organizations, medical organizations, research centers, and even federal agencies such as the U.S. Food and Drug Administration, are working to minimize animal use in scientific experiments. This movement primarily stems from animal welfare and ethical concerns. However, recent advances in technology and new studies in medicine have contributed to an increase in animal experiments throughout the years. With the rapid increase in animal testing, concerns arise including ethical issues, high cost, complex procedures, and potential inaccuracies.Alternative solutions have recently been investigated to address the problems of animal testing. Some of these technologies are related to stem cell technologies, such as organ-on-a-chip, organoids, and induced pluripotent stem cell models. The aim of the review is to focus on stem cell related methodologies, such as organoids, that can serve as an alternative to animal testing and discuss its advantages and limitations, alongside regulatory considerations.Although stem cell related methodologies has shortcomings, it has potential to replace animal testing. Achieving this requires further research on stem cells, with potential societal and technological benefits.
Collapse
Affiliation(s)
- Guiyoung Park
- School of Biopharmaceutical and Medical Sciences, Health & Wellness College, Sungshin Women's University, 55, Dobong-ro 76ga-gil, Gangbuk-gu, Seoul, Republic of Korea
| | - Yeri Alice Rim
- CiSTEM laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, 4 3, Seoul, 06591, Republic of Korea
- Department of Biomedicine & Health Sciences, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yeowon Sohn
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
| | - Yoojun Nam
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea.
- Yipscell Inc, L2 Omnibus Park, Banpo-dearo 222, Seocho-gu, Seoul, Korea.
| | - Ji Hyeon Ju
- CiSTEM laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, 4 3, Seoul, 06591, Republic of Korea.
- Department of Biomedicine & Health Sciences, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Yipscell Inc, L2 Omnibus Park, Banpo-dearo 222, Seocho-gu, Seoul, Korea.
| |
Collapse
|
14
|
Almeida-Pinto J, Moura BS, Gaspar VM, Mano JF. Advances in Cell-Rich Inks for Biofabricating Living Architectures. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313776. [PMID: 38639337 DOI: 10.1002/adma.202313776] [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: 12/17/2023] [Revised: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Advancing biofabrication toward manufacturing living constructs with well-defined architectures and increasingly biologically relevant cell densities is highly desired to mimic the biofunctionality of native human tissues. The formulation of tissue-like, cell-dense inks for biofabrication remains, however, challenging at various levels of the bioprinting process. Promising advances have been made toward this goal, achieving relatively high cell densities that surpass those found in conventional platforms, pushing the current boundaries closer to achieving tissue-like cell densities. On this focus, herein the overarching challenges in the bioprocessing of cell-rich living inks into clinically grade engineered tissues are discussed, as well as the most recent advances in cell-rich living ink formulations and their processing technologies are highlighted. Additionally, an overview of the foreseen developments in the field is provided and critically discussed.
Collapse
Affiliation(s)
- José Almeida-Pinto
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Beatriz S Moura
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Vítor M Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| |
Collapse
|
15
|
Faeed M, Ghiasvand M, Fareghzadeh B, Taghiyar L. Osteochondral organoids: current advances, applications, and upcoming challenges. Stem Cell Res Ther 2024; 15:183. [PMID: 38902814 PMCID: PMC11191177 DOI: 10.1186/s13287-024-03790-5] [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: 02/05/2024] [Accepted: 06/09/2024] [Indexed: 06/22/2024] Open
Abstract
In the realm of studying joint-related diseases, there is a continuous quest for more accurate and representative models. Recently, regenerative medicine and tissue engineering have seen a growing interest in utilizing organoids as powerful tools for studying complex biological systems in vitro. Organoids, three-dimensional structures replicating the architecture and function of organs, provide a unique platform for investigating disease mechanisms, drug responses, and tissue regeneration. The surge in organoid research is fueled by the need for physiologically relevant models to bridge the gap between traditional cell cultures and in vivo studies. Osteochondral organoids have emerged as a promising avenue in this pursuit, offering a better platform to mimic the intricate biological interactions within bone and cartilage. This review explores the significance of osteochondral organoids and the need for their development in advancing our understanding and treatment of bone and cartilage-related diseases. It summarizes osteochondral organoids' insights and research progress, focusing on their composition, materials, cell sources, and cultivation methods, as well as the concept of organoids on chips and application scenarios. Additionally, we address the limitations and challenges these organoids face, emphasizing the necessity for further research to overcome these obstacles and facilitate orthopedic regeneration.
Collapse
Affiliation(s)
- Maryam Faeed
- Cell and Molecular School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Mahsa Ghiasvand
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute for Stem cell Biology and Technology, ACECR, Tehran, Iran
| | - Bahar Fareghzadeh
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Leila Taghiyar
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute for Stem cell Biology and Technology, ACECR, Tehran, Iran.
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
16
|
Wang F, Song P, Wang J, Wang S, Liu Y, Bai L, Su J. Organoid bioinks: construction and application. Biofabrication 2024; 16:032006. [PMID: 38697093 DOI: 10.1088/1758-5090/ad467c] [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: 11/23/2023] [Accepted: 05/02/2024] [Indexed: 05/04/2024]
Abstract
Organoids have emerged as crucial platforms in tissue engineering and regenerative medicine but confront challenges in faithfully mimicking native tissue structures and functions. Bioprinting technologies offer a significant advancement, especially when combined with organoid bioinks-engineered formulations designed to encapsulate both the architectural and functional elements of specific tissues. This review provides a rigorous, focused examination of the evolution and impact of organoid bioprinting. It emphasizes the role of organoid bioinks that integrate key cellular components and microenvironmental cues to more accurately replicate native tissue complexity. Furthermore, this review anticipates a transformative landscape invigorated by the integration of artificial intelligence with bioprinting techniques. Such fusion promises to refine organoid bioink formulations and optimize bioprinting parameters, thus catalyzing unprecedented advancements in regenerative medicine. In summary, this review accentuates the pivotal role and transformative potential of organoid bioinks and bioprinting in advancing regenerative therapies, deepening our understanding of organ development, and clarifying disease mechanisms.
Collapse
Affiliation(s)
- Fuxiao Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- These authors contributed equally
| | - Peiran Song
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- These authors contributed equally
| | - Jian Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- These authors contributed equally
| | - Sicheng Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200444, People's Republic of China
| | - Yuanyuan Liu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, People's Republic of China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- Wenzhou Institute of Shanghai University, Wenzhou 325000, People's Republic of China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
| |
Collapse
|
17
|
Shrestha S, Lekkala VKR, Acharya P, Kang SY, Vanga MG, Lee MY. Reproducible generation of human liver organoids (HLOs) on a pillar plate platform via microarray 3D bioprinting. LAB ON A CHIP 2024; 24:2747-2761. [PMID: 38660778 PMCID: PMC11605706 DOI: 10.1039/d4lc00149d] [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] [Indexed: 04/26/2024]
Abstract
Human liver organoids (HLOs) hold significant potential for recapitulating the architecture and function of liver tissues in vivo. However, conventional culture methods of HLOs, forming Matrigel domes in 6-/24-well plates, have technical limitations such as high cost and low throughput in organoid-based assays for predictive assessment of compounds in clinical and pharmacological lab settings. To address these issues, we have developed a unique microarray 3D bioprinting protocol of progenitor cells in biomimetic hydrogels on a pillar plate with sidewalls and slits, coupled with a clear bottom, 384-deep well plate for scale-up production of HLOs. Microarray 3D bioprinting, a droplet-based printing technology, was used to generate a large number of small organoids on the pillar plate for predictive hepatotoxicity assays. Foregut cells, differentiated from human iPSCs, were mixed with Matrigel and then printed on the pillar plate rapidly and uniformly, resulting in coefficient of variation (CV) values in the range of 15-18%, without any detrimental effect on cell viability. Despite utilizing 10-50-fold smaller cell culture volume compared to their counterparts in Matrigel domes in 6-/24-well plates, HLOs differentiated on the pillar plate exhibited similar morphology and superior function, potentially due to rapid diffusion of nutrients and oxygen at the small scale. Day 25 HLOs were robust and functional on the pillar plate in terms of their viability, albumin secretion, CYP3A4 activity, and drug toxicity testing, all with low CV values. From three independent trials of in situ assessment, the IC50 values calculated for sorafenib and tamoxifen were 6.2 ± 1.6 μM and 25.4 ± 8.3 μM, respectively. Therefore, our unique 3D bioprinting and miniature organoid culture on the pillar plate could be used for scale-up, reproducible generation of HLOs with minimal manual intervention for high-throughput assessment of compound hepatotoxicity.
Collapse
Affiliation(s)
- Sunil Shrestha
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA.
| | | | - Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA.
| | - Soo-Yeon Kang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA.
| | - Manav Goud Vanga
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA.
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA.
- Bioprinting Laboratories Inc., Dallas, Texas, USA
| |
Collapse
|
18
|
Park S, Cho SW. Bioengineering toolkits for potentiating organoid therapeutics. Adv Drug Deliv Rev 2024; 208:115238. [PMID: 38447933 DOI: 10.1016/j.addr.2024.115238] [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: 09/26/2023] [Revised: 01/28/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Organoids are three-dimensional, multicellular constructs that recapitulate the structural and functional features of specific organs. Because of these characteristics, organoids have been widely applied in biomedical research in recent decades. Remarkable advancements in organoid technology have positioned them as promising candidates for regenerative medicine. However, current organoids still have limitations, such as the absence of internal vasculature, limited functionality, and a small size that is not commensurate with that of actual organs. These limitations hinder their survival and regenerative effects after transplantation. Another significant concern is the reliance on mouse tumor-derived matrix in organoid culture, which is unsuitable for clinical translation due to its tumor origin and safety issues. Therefore, our aim is to describe engineering strategies and alternative biocompatible materials that can facilitate the practical applications of organoids in regenerative medicine. Furthermore, we highlight meaningful progress in organoid transplantation, with a particular emphasis on the functional restoration of various organs.
Collapse
Affiliation(s)
- Sewon Park
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea; Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
19
|
Saadeldin IM, Ehab S, Noreldin AE, Swelum AAA, Bang S, Kim H, Yoon KY, Lee S, Cho J. Current strategies using 3D organoids to establish in vitro maternal-embryonic interaction. J Vet Sci 2024; 25:e40. [PMID: 38834510 PMCID: PMC11156602 DOI: 10.4142/jvs.24004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/14/2024] [Accepted: 03/28/2024] [Indexed: 06/06/2024] Open
Abstract
IMPORTANCE The creation of robust maternal-embryonic interactions and implantation models is important for comprehending the early stages of embryonic development and reproductive disorders. Traditional two-dimensional (2D) cell culture systems often fail to accurately mimic the highly complex in vivo conditions. The employment of three-dimensional (3D) organoids has emerged as a promising strategy to overcome these limitations in recent years. The advancements in the field of organoid technology have opened new avenues for studying the physiology and diseases affecting female reproductive tract. OBSERVATIONS This review summarizes the current strategies and advancements in the field of 3D organoids to establish maternal-embryonic interaction and implantation models for use in research and personalized medicine in assisted reproductive technology. The concepts of endometrial organoids, menstrual blood flow organoids, placental trophoblast organoids, stem cell-derived blastoids, and in vitro-generated embryo models are discussed in detail. We show the incorportaion of organoid systems and microfluidic technology to enhance tissue performance and precise management of the cellular surroundings. CONCLUSIONS AND RELEVANCE This review provides insights into the future direction of modeling maternal-embryonic interaction research and its combination with other powerful technologies to interfere with this dialogue either by promoting or hindering it for improving fertility or methods for contraception, respectively. The merging of organoid systems with microfluidics facilitates the creation of sophisticated and functional organoid models, enhancing insights into organ development, disease mechanisms, and personalized medical investigations.
Collapse
Affiliation(s)
- Islam Mohamed Saadeldin
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Seif Ehab
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza 11341, Egypt
| | - Ahmed Elsayed Noreldin
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, the Scientific Campus, Damanhour 22511, Egypt
| | - Ayman Abdel-Aziz Swelum
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Seonggyu Bang
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea
| | - Hyejin Kim
- Division in Biomedical Art, Department of Fine Art, Incheon Catholic University Graduate School, Incheon 21986, Korea
| | - Ki Young Yoon
- Department of Companion Animal, Shingu College, Seongnam 13174, Korea
| | - Sanghoon Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Jongki Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
20
|
Shrestha S, Lekkala VKR, Acharya P, Kang SY, Vanga MG, Lee MY. Reproducible generation of human liver organoids (HLOs) on a pillar plate platform via microarray 3D bioprinting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584478. [PMID: 38559126 PMCID: PMC10979895 DOI: 10.1101/2024.03.11.584478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Human liver organoids (HLOs) hold significant potential for recapitulating the architecture and function of liver tissues in vivo. However, conventional culture methods of HLOs, forming Matrigel domes in 6-/24-well plates, have technical limitations such as high cost and low throughput in organoid-based assays for predictive assessment of compounds in clinical and pharmacological lab settings. To address these issues, we have developed a unique microarray 3D bioprinting protocol of progenitor cells in biomimetic hydrogels on a pillar plate with sidewalls and slits, coupled with a clear bottom, 384-deep well plate for scale-up production of HLOs. Microarray 3D bioprinting, a droplet-based printing technology, was used to generate a large number of small organoids on the pillar plate for predictive hepatotoxicity assays. Foregut cells, differentiated from human iPSCs, were mixed with Matrigel and then printed on the pillar plate rapidly and uniformly, resulting in coefficient of variation (CV) values in the range of 15 - 18%, without any detrimental effect on cell viability. Despite utilizing 10 - 50-fold smaller cell culture volume compared to their counterparts in Matrigel domes in 6-/24-well plates, HLOs differentiated on the pillar plate exhibited similar morphology and superior function, potentially due to rapid diffusion of nutrients and oxygen at the small scale. Day 25 HLOs were robust and functional on the pillar plate in terms of their viability, albumin secretion, CYP3A4 activity, and drug toxicity testing, all with low CV values. From three independent trials of in situ assessment, the IC50 values calculated for sorafenib and tamoxifen were 6.2 ± 1.6 μM and 25.4 ± 8.3 μM, respectively. Therefore, our unique 3D bioprinting and miniature organoid culture on the pillar plate could be used for scale-up, reproducible generation of HLOs with minimal manual intervention for high-throughput assessment of compound hepatotoxicity.
Collapse
Affiliation(s)
- Sunil Shrestha
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | | | - Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | - Soo-Yeon Kang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | - Manav Goud Vanga
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas
- Bioprinting Laboratories Inc., Dallas, Texas
| |
Collapse
|
21
|
Chen W, Zhu Y, Liu R, Kong B, Xia N, Zhao Y, Sun L. Screening Therapeutic Effects of MSC-EVs to Acute Lung Injury Model on A Chip. Adv Healthc Mater 2024; 13:e2303123. [PMID: 38084928 DOI: 10.1002/adhm.202303123] [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: 09/17/2023] [Revised: 11/29/2023] [Indexed: 12/19/2023]
Abstract
Acute lung injury (ALI) is a lethal disease with high mortality rate, and its physiologically relevant models that could mimic human disease processes are urgently needed to study pathophysiology and predict drug efficacy. Here, this work presents a novel lipopolysaccharide (LPS) based ALI model on a microfluidic chip that reconstitutes an air-liquid interface lined by human alveolar epithelium and microvascular endothelium for screening the therapeutic effects of mesenchymal stem cells (MSC) derived extracellular vesicles (MSC-EVs) to the biomimetic ALI. The air-liquid interface is established by coculture of alveolar epithelium and microvascular endothelium on the opposite sides of the porous membrane. The functionalized architecture is characterized by integrate cell layers and suitable permeability. Using this biomimetic microsystem, LPS based ALI model is established, which exhibits the disrupted alveolar-capillary barrier, reduced transepithelial/transendothelial electrical resistance (TEER), and impaired expression of junction proteins. As a reliable disease model, this work examines the effects of MSC-EVs, and the data indicate the therapeutic potential of EVs for severe ALI. MSC-EVs can alleviate barrier disruption by restoring both the epithelial and endothelial barrier integrity. They hope this study can become a unique approach to study human pathophysiology of ALI and advance drug development.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Yujuan Zhu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Rui Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Bin Kong
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Nan Xia
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, China
| |
Collapse
|
22
|
Setiawan J, Rizal DM, Sofyantoro F, Priyono DS, Septriani NI, Mafiroh WU, Kotani T, Matozaki T, Putri WA. Bibliometric analysis of organoids in regenerative medicine-related research worldwide over two decades (2002-2022). Regen Med 2024; 19:119-133. [PMID: 38449425 DOI: 10.2217/rme-2023-0176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024] Open
Abstract
Aim: This study aimed to evaluate the trends in organoid culture research within the field of regenerative medicine from 2002 to 2022. Methods: The worldwide distribution of organoid research in regenerative medicine articles indexed in the Scopus database was analyzed. Result: A total of 840 documents were analyzed, averaging 42 publications annually. The USA (n = 296) led in publications, followed by China (n = 127), Japan (n = 91) and the UK (n = 75). Since 2011, research has surged, particularly in China, which emerged as a prominent center. Conclusion: The findings highlight significant growth in organoid research, promising future organ transplantation. Research trends integrate tissue engineering, gene modification and induced pluripotent stem cell technologies, reflecting a move toward personalized medicine.
Collapse
Affiliation(s)
- Jajar Setiawan
- Department of Physiology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Dicky Moch Rizal
- Department of Physiology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Fajar Sofyantoro
- Department of Tropical Biology, Faculty of Biology, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Dwi Sendi Priyono
- Department of Tropical Biology, Faculty of Biology, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Nur Indah Septriani
- Department of Tropical Biology, Faculty of Biology, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Wulan Usfi Mafiroh
- Department of Tropical Biology, Faculty of Biology, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Takenori Kotani
- Division of Molecular and Cellular Signaling, Department of Biochemistry & Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takashi Matozaki
- Division of Molecular and Cellular Signaling, Department of Biochemistry & Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Biosignal Regulation, Department of Biochemistry & Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Wahyu Aristyaning Putri
- Department of Tropical Biology, Faculty of Biology, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
23
|
Mai S, Inkielewicz-Stepniak I. Graphene Oxide Nanoparticles and Organoids: A Prospective Advanced Model for Pancreatic Cancer Research. Int J Mol Sci 2024; 25:1066. [PMID: 38256139 PMCID: PMC10817028 DOI: 10.3390/ijms25021066] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Pancreatic cancer, notorious for its grim 10% five-year survival rate, poses significant clinical challenges, largely due to late-stage diagnosis and limited therapeutic options. This review delves into the generation of organoids, including those derived from resected tissues, biopsies, pluripotent stem cells, and adult stem cells, as well as the advancements in 3D printing. It explores the complexities of the tumor microenvironment, emphasizing culture media, the integration of non-neoplastic cells, and angiogenesis. Additionally, the review examines the multifaceted properties of graphene oxide (GO), such as its mechanical, thermal, electrical, chemical, and optical attributes, and their implications in cancer diagnostics and therapeutics. GO's unique properties facilitate its interaction with tumors, allowing targeted drug delivery and enhanced imaging for early detection and treatment. The integration of GO with 3D cultured organoid systems, particularly in pancreatic cancer research, is critically analyzed, highlighting current limitations and future potential. This innovative approach has the promise to transform personalized medicine, improve drug screening efficiency, and aid biomarker discovery in this aggressive disease. Through this review, we offer a balanced perspective on the advancements and future prospects in pancreatic cancer research, harnessing the potential of organoids and GO.
Collapse
Affiliation(s)
| | - Iwona Inkielewicz-Stepniak
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| |
Collapse
|
24
|
Bhatt S S, Krishna Kumar J, Laya S, Thakur G, Nune M. Scaffold-mediated liver regeneration: A comprehensive exploration of current advances. J Tissue Eng 2024; 15:20417314241286092. [PMID: 39411269 PMCID: PMC11475092 DOI: 10.1177/20417314241286092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/08/2024] [Indexed: 10/19/2024] Open
Abstract
The liver coordinates over 500 biochemical processes crucial for maintaining homeostasis, detoxification, and metabolism. Its specialized cells, arranged in hexagonal lobules, enable it to function as a highly efficient metabolic engine. However, diseases such as cirrhosis, fatty liver disease, and hepatitis present significant global health challenges. Traditional drug development is expensive and often ineffective at predicting human responses, driving interest in advanced in vitro liver models utilizing 3D bioprinting and microfluidics. These models strive to mimic the liver's complex microenvironment, improving drug screening and disease research. Despite its resilience, the liver is vulnerable to chronic illnesses, injuries, and cancers, leading to millions of deaths annually. Organ shortages hinder liver transplantation, highlighting the need for alternative treatments. Tissue engineering, employing polymer-based scaffolds and 3D bioprinting, shows promise. This review examines these innovative strategies, including liver organoids and liver tissue-on-chip technologies, to address the challenges of liver diseases.
Collapse
Affiliation(s)
- Supriya Bhatt S
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Jayanthi Krishna Kumar
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shurthi Laya
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
- Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Goutam Thakur
- Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Manasa Nune
- Manipal Institute of Regenerative Medicine, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
25
|
Martinello T, Akyürek EE, Ventriglia G, Patruno M, Sacchetto R. The dorsal portion of the bovine diaphragm as a useful tissue for producing a 3D muscle scaffold. J Anat 2023; 243:878-885. [PMID: 37322832 PMCID: PMC10557388 DOI: 10.1111/joa.13915] [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: 02/07/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/17/2023] Open
Abstract
Three-dimensional (3D) organoids are an innovative approach to obtain an in vitro model for ex vivo studies to overcome the limitations of monolayer cell culture and reduce the use of animal models. An organoid of skeletal muscle requires the presence of the extracellular matrix to represent a functional muscle in vitro, which is why decellularized tissue is an optimal choice. Various muscles have been considered to produce a muscle organoid, most from rodents or small animals, and only recently some studies have been reported on the muscles of large animals. This work presents a muscular organoid produced from the bovine diaphragm, which has a peculiar multilayered structure with different fibre orientations depending on the considered area. This paper analyses the anatomical structure of the bovine diaphragm, selects the most appropriate portion, and presents a decellularization protocol for a multilayered muscle. In addition, a preliminary test of recellularization with primary bovine myocytes was presented with the future aim of obtaining a 3D muscle allogenic organoid, completely bovine-derived. The results demonstrate that the dorsal portion of bovine diaphragm presents a regular alternation of muscular and fibrous layers and that the complete decellularization does not affect the biocompatibility. These results provide a strong foundation for the potential application of this portion of tissue as a scaffold for in vitro studies of muscle organoids.
Collapse
Affiliation(s)
| | - Eylem Emek Akyürek
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | | | - Marco Patruno
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | - Roberta Sacchetto
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| |
Collapse
|
26
|
Ren B, Chiaravalloti TR, Belony NL, Romero DI, Chai W, Leon C, Wu L, Lamango NS, Offringa IA, Huang Y. Design and Realization of Lung Organoid Cultures for COVID-19 Applications. Biodes Manuf 2023; 6:646-660. [PMID: 38993804 PMCID: PMC11238720 DOI: 10.1007/s42242-023-00255-1] [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: 04/01/2023] [Accepted: 06/25/2023] [Indexed: 07/13/2024]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been spreading globally and threatening public health. Advanced in vitro models that recapitulate the architecture and functioning of specific tissues and organs are in high demand for COVID-19-related pathology studies and drug screening. Three-dimensional (3D) in vitro cultures such as self-assembled and engineered organoid cultures surpass conventional two-dimensional (2D) cultures and animal models with respect to the increased cellular complexity, better human-relevant environment, and reduced cost, thus presenting as promising platforms for understanding viral pathogenesis and developing new therapeutics. This review highlights the recent advances in self-assembled and engineered organoid technologies that are used for COVID-19 studies. The challenges and future perspectives are also discussed.
Collapse
Affiliation(s)
- Bing Ren
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611
| | | | - Nadine L Belony
- College of Pharmacy, University of Florida, Gainesville, FL 32611
| | - Diana I Romero
- College of Pharmacy, University of Florida, Gainesville, FL 32611
| | - Wenxuan Chai
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611
| | - Christopher Leon
- Norris Comprehensive Cancer Center, Departments of Surgery and of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Lizi Wu
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32611
| | - Nazarius S Lamango
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307
| | - Ite A Offringa
- Norris Comprehensive Cancer Center, Departments of Surgery and of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089
| | - Yong Huang
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611
- Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611
| |
Collapse
|
27
|
Fang Y, Ji M, Wu B, Xu X, Wang G, Zhang Y, Xia Y, Li Z, Zhang T, Sun W, Xiong Z. Engineering Highly Vascularized Bone Tissues by 3D Bioprinting of Granular Prevascularized Spheroids. ACS APPLIED MATERIALS & INTERFACES 2023; 15:43492-43502. [PMID: 37691550 DOI: 10.1021/acsami.3c08550] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The convergence of 3D bioprinting with powerful manufacturing capability and cellular self-organization that can reproduce intricate tissue microarchitecture and function is a promising direction toward building functional tissues and has yet to be demonstrated. Here, we develop a granular aggregate-prevascularized (GAP) bioink for engineering highly vascularized bone tissues by capitalizing on the condensate-mimicking, self-organization, and angiogenic properties of prevascularized mesenchymal spheroids. The GAP bioink utilizes prevascularized aggregates as building blocks, which are embedded densely in extracellular matrices conducive to spontaneous self-organization. We printed various complex structures with high cell density (∼1.5 × 108 cells/cm3), viability (∼80%), and shape fidelity using GAP bioink. After printing, the prevascularized mesenchymal spheroids developed an interconnected vascular network through angiogenic sprouting. We printed highly vascularized bone tissues using GAP bioink and found that prevascularized spheroids were more conducive to osteogenesis and angiogenesis. We envision that the design of the GAP bioink could be further integrated with human-induced pluripotent stem cell-derived organoids, which opens new avenues to create patient-specific vascularized tissues for therapeutic applications..
Collapse
Affiliation(s)
- Yongcong Fang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, P. R. China
- Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing 100084, P. R. China
| | - Mengke Ji
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, P. R. China
- Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing 100084, P. R. China
| | - Bingyan Wu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, P. R. China
- Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing 100084, P. R. China
| | - Xinxin Xu
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, P. R. China
| | - Ge Wang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, P. R. China
- Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing 100084, P. R. China
| | - Yanmei Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, P. R. China
- Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing 100084, P. R. China
| | - Yingkai Xia
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, P. R. China
- Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing 100084, P. R. China
| | - Zhe Li
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, P. R. China
- Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing 100084, P. R. China
| | - Ting Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, P. R. China
- Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing 100084, P. R. China
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, P. R. China
- Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing 100084, P. R. China
- Department of Mechanical Engineering, Drexel University, Philadelphia, Pennsylvania 19104, United States of America
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, P. R. China
- Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing 100084, P. R. China
| |
Collapse
|
28
|
Lotto J, Stephan TL, Hoodless PA. Fetal liver development and implications for liver disease pathogenesis. Nat Rev Gastroenterol Hepatol 2023; 20:561-581. [PMID: 37208503 DOI: 10.1038/s41575-023-00775-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 05/21/2023]
Abstract
The metabolic, digestive and homeostatic roles of the liver are dependent on proper crosstalk and organization of hepatic cell lineages. These hepatic cell lineages are derived from their respective progenitors early in organogenesis in a spatiotemporally controlled manner, contributing to the liver's specialized and diverse microarchitecture. Advances in genomics, lineage tracing and microscopy have led to seminal discoveries in the past decade that have elucidated liver cell lineage hierarchies. In particular, single-cell genomics has enabled researchers to explore diversity within the liver, especially early in development when the application of bulk genomics was previously constrained due to the organ's small scale, resulting in low cell numbers. These discoveries have substantially advanced our understanding of cell differentiation trajectories, cell fate decisions, cell lineage plasticity and the signalling microenvironment underlying the formation of the liver. In addition, they have provided insights into the pathogenesis of liver disease and cancer, in which developmental processes participate in disease emergence and regeneration. Future work will focus on the translation of this knowledge to optimize in vitro models of liver development and fine-tune regenerative medicine strategies to treat liver disease. In this Review, we discuss the emergence of hepatic parenchymal and non-parenchymal cells, advances that have been made in in vitro modelling of liver development and draw parallels between developmental and pathological processes.
Collapse
Affiliation(s)
- Jeremy Lotto
- Terry Fox Laboratory, BC Cancer, Vancouver, BC, Canada
- Cell and Developmental Biology Program, University of British Columbia, Vancouver, BC, Canada
| | - Tabea L Stephan
- Terry Fox Laboratory, BC Cancer, Vancouver, BC, Canada
- Cell and Developmental Biology Program, University of British Columbia, Vancouver, BC, Canada
| | - Pamela A Hoodless
- Terry Fox Laboratory, BC Cancer, Vancouver, BC, Canada.
- Cell and Developmental Biology Program, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
29
|
Mitrovic J, Richey G, Kim S, Guler MO. Peptide Hydrogels and Nanostructures Controlling Biological Machinery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:11935-11945. [PMID: 37589176 PMCID: PMC10469456 DOI: 10.1021/acs.langmuir.3c01269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Indexed: 08/18/2023]
Abstract
Peptides are versatile building blocks for the fabrication of various nanostructures that result in the formation of hydrogels and nanoparticles. Precise chemical functionalization promotes discrete structure formation, causing controlled bioactivity and physical properties for functional materials development. The conjugation of small molecules on amino acid side chains determines their intermolecular interactions in addition to their intrinsic peptide characteristics. Molecular information affects the peptide structure, formation, and activity. In this Perspective, peptide building blocks, nanostructure formation mechanisms, and the properties of these peptide materials are discussed with the results of recent publications. Bioinstructive and stimuli-responsive peptide materials have immense impacts on the nanomedicine field including drug delivery, cellular engineering, regenerative medicine, and biomedicine.
Collapse
Affiliation(s)
- Jovana Mitrovic
- The Pritzker School of Molecular
Engineering, The University of Chicago, Chicago, Illinois 60637 United States
| | - Gabriella Richey
- The Pritzker School of Molecular
Engineering, The University of Chicago, Chicago, Illinois 60637 United States
| | - Sarah Kim
- The Pritzker School of Molecular
Engineering, The University of Chicago, Chicago, Illinois 60637 United States
| | - Mustafa O. Guler
- The Pritzker School of Molecular
Engineering, The University of Chicago, Chicago, Illinois 60637 United States
| |
Collapse
|
30
|
Li W, Liu Z, Tang F, Jiang H, Zhou Z, Hao X, Zhang JM. Application of 3D Bioprinting in Liver Diseases. MICROMACHINES 2023; 14:1648. [PMID: 37630184 PMCID: PMC10457767 DOI: 10.3390/mi14081648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/03/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023]
Abstract
Liver diseases are the primary reason for morbidity and mortality in the world. Owing to a shortage of organ donors and postoperative immune rejection, patients routinely suffer from liver failure. Unlike 2D cell models, animal models, and organoids, 3D bioprinting can be successfully employed to print living tissues and organs that contain blood vessels, bone, and kidney, heart, and liver tissues and so on. 3D bioprinting is mainly classified into four types: inkjet 3D bioprinting, extrusion-based 3D bioprinting, laser-assisted bioprinting (LAB), and vat photopolymerization. Bioinks for 3D bioprinting are composed of hydrogels and cells. For liver 3D bioprinting, hepatic parenchymal cells (hepatocytes) and liver nonparenchymal cells (hepatic stellate cells, hepatic sinusoidal endothelial cells, and Kupffer cells) are commonly used. Compared to conventional scaffold-based approaches, marked by limited functionality and complexity, 3D bioprinting can achieve accurate cell settlement, a high resolution, and more efficient usage of biomaterials, better mimicking the complex microstructures of native tissues. This method will make contributions to disease modeling, drug discovery, and even regenerative medicine. However, the limitations and challenges of this method cannot be ignored. Limitation include the requirement of diverse fabrication technologies, observation of drug dynamic response under perfusion culture, the resolution to reproduce complex hepatic microenvironment, and so on. Despite this, 3D bioprinting is still a promising and innovative biofabrication strategy for the creation of artificial multi-cellular tissues/organs.
Collapse
Affiliation(s)
- Wenhui Li
- Department of Radiology, Yancheng Third People’s Hospital, Affiliated Hospital 6 of Nantong University, Yancheng 224000, China
| | - Zhaoyue Liu
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
| | - Fengwei Tang
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
| | - Hao Jiang
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
| | - Zhengyuan Zhou
- Nanjing Hangdian Intelligent Manufacturing Technology Co., Ltd., Nanjing 210014, China
| | - Xiuqing Hao
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
| | - Jia Ming Zhang
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
- Nanjing Hangdian Intelligent Manufacturing Technology Co., Ltd., Nanjing 210014, China
- Yangtze River Delta Intelligent Manufacturing Innovation Center, Nanjing 210014, China
| |
Collapse
|
31
|
Santos AK, Scalzo S, de Souza RTV, Santana PHG, Marques BL, Oliveira LF, Filho DM, Kihara AH, da Costa Santiago H, Parreira RC, Birbrair A, Ulrich H, Resende RR. Strategic use of organoids and organs-on-chip as biomimetic tools. Semin Cell Dev Biol 2023; 144:3-10. [PMID: 36192310 DOI: 10.1016/j.semcdb.2022.09.010] [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: 06/14/2022] [Revised: 09/17/2022] [Accepted: 09/17/2022] [Indexed: 11/30/2022]
Abstract
Organoid development and organ-on-a-chip are technologies based on differentiating stem cells, forming 3D multicellular structures resembling organs and tissues in vivo. Hence, both can be strategically used for disease modeling, drug screening, and host-pathogen studies. In this context, this review highlights the significant advancements in the area, providing technical approaches to organoids and organ-on-a-chip that best imitate in vivo physiology.
Collapse
Affiliation(s)
- Anderson K Santos
- Department of Pediatrics, Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Sérgio Scalzo
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | - Bruno L Marques
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Lucas F Oliveira
- Departamento de Fisiologia, Instituto de Ciências Biológicas, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Daniel M Filho
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Alexandre Hiroaki Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Helton da Costa Santiago
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Alexander Birbrair
- Departmento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Henning Ulrich
- Departmento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinópolis, Brazil.
| |
Collapse
|
32
|
Tian CM, Yang MF, Xu HM, Zhu MZ, Yue NN, Zhang Y, Shi RY, Yao J, Wang LS, Liang YJ, Li DF. Stem cell-derived intestinal organoids: a novel modality for IBD. Cell Death Discov 2023; 9:255. [PMID: 37479716 PMCID: PMC10362068 DOI: 10.1038/s41420-023-01556-1] [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: 04/28/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/23/2023] Open
Abstract
The organoids represent one of the greatest revolutions in the biomedical field in the past decade. This three-dimensional (3D) micro-organ cultured in vitro has a structure highly similar to that of the tissue and organ. Using the regeneration ability of stem cells, a 3D organ-like structure called intestinal organoids is established, which can mimic the characteristics of real intestinal organs, including morphology, function, and personalized response to specific stimuli. Here, we discuss current stem cell-based organ-like 3D intestinal models, including understanding the molecular pathophysiology, high-throughput screening drugs, drug efficacy testing, toxicological evaluation, and organ-based regeneration of inflammatory bowel disease (IBD). We summarize the advances and limitations of the state-of-the-art reconstruction platforms for intestinal organoids. The challenges, advantages, and prospects of intestinal organs as an in vitro model system for precision medicine are also discussed. Key applications of stem cell-derived intestinal organoids. Intestinal organoids can be used to model infectious diseases, develop new treatments, drug screens, precision medicine, and regenerative medicine.
Collapse
Affiliation(s)
- Cheng-Mei Tian
- Department of Gastroenterology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China
- Department of Emergency, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China
| | - Mei-Feng Yang
- Department of Hematology, Yantian District People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 51000, China
| | - Min-Zheng Zhu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 51000, China
| | - Ning-Ning Yue
- Department of Gastroenterology, Shenzhen People's Hospital The Second Clinical Medical College, Jinan University, Shenzhen, 518020, Guangdong, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, 516000, Guangdong, China
| | - Rui-Yue Shi
- Department of Gastroenterology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China.
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China.
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China.
| | - Yu-Jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen, 518020, Guangdong, China.
| | - De-Feng Li
- Department of Gastroenterology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China.
| |
Collapse
|
33
|
Doganay MT, Chelliah CJ, Tozluyurt A, Hujer AM, Obaro SK, Gurkan U, Patel R, Bonomo RA, Draz M. 3D Printed Materials for Combating Antimicrobial Resistance. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2023; 67:371-398. [PMID: 37790286 PMCID: PMC10545363 DOI: 10.1016/j.mattod.2023.05.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Three-dimensional (3D) printing is a rapidly growing technology with a significant capacity for translational applications in both biology and medicine. 3D-printed living and non-living materials are being widely tested as a potential replacement for conventional solutions for testing and combating antimicrobial resistance (AMR). The precise control of cells and their microenvironment, while simulating the complexity and dynamics of an in vivo environment, provides an excellent opportunity to advance the modeling and treatment of challenging infections and other health conditions. 3D-printing models the complicated niches of microbes and host-pathogen interactions, and most importantly, how microbes develop resistance to antibiotics. In addition, 3D-printed materials can be applied to testing and delivering antibiotics. Here, we provide an overview of 3D printed materials and biosystems and their biomedical applications, focusing on ever increasing AMR. Recent applications of 3D printing to alleviate the impact of AMR, including developed bioprinted systems, targeted bacterial infections, and tested antibiotics are presented.
Collapse
Affiliation(s)
- Mert Tunca Doganay
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Cyril John Chelliah
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Abdullah Tozluyurt
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Andrea M Hujer
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | | | - Umut Gurkan
- Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology and Division of Public Health, Infectious Diseases, and Occupational medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Robert A Bonomo
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
- Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES) Cleveland, OH, USA
| | - Mohamed Draz
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
- Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH 44106, USA
| |
Collapse
|
34
|
Juraski AC, Sharma S, Sparanese S, da Silva VA, Wong J, Laksman Z, Flannigan R, Rohani L, Willerth SM. 3D bioprinting for organ and organoid models and disease modeling. Expert Opin Drug Discov 2023; 18:1043-1059. [PMID: 37431937 DOI: 10.1080/17460441.2023.2234280] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/05/2023] [Indexed: 07/12/2023]
Abstract
INTRODUCTION 3D printing, a versatile additive manufacturing technique, has diverse applications ranging from transportation, rapid prototyping, clean energy, and medical devices. AREAS COVERED The authors focus on how 3D printing technology can enhance the drug discovery process through automating tissue production that enables high-throughput screening of potential drug candidates. They also discuss how the 3D bioprinting process works and what considerations to address when using this technology to generate cell laden constructs for drug screening as well as the outputs from such assays necessary for determining the efficacy of potential drug candidates. They focus on how bioprinting how has been used to generate cardiac, neural, and testis tissue models, focusing on bio-printed 3D organoids. EXPERT OPINION The next generation of 3D bioprinted organ model holds great promises for the field of medicine. In terms of drug discovery, the incorporation of smart cell culture systems and biosensors into 3D bioprinted models could provide highly detailed and functional organ models for drug screening. By addressing current challenges of vascularization, electrophysiological control, and scalability, researchers can obtain more reliable and accurate data for drug development, reducing the risk of drug failures during clinical trials.
Collapse
Affiliation(s)
- Amanda C Juraski
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria BC, Canada
- Department of Chemical Engineering, Polytechnic School, University of Sao Paulo, Sao Paulo, Brazil
| | - Sonali Sharma
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Sydney Sparanese
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver BC, Canada
| | - Victor A da Silva
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria BC, Canada
| | - Julie Wong
- Department of Urologic Sciences, University of British Columbia, Vancouver BC, Canada
| | - Zachary Laksman
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Ryan Flannigan
- Department of Urologic Sciences, University of British Columbia, Vancouver BC, Canada
| | - Leili Rohani
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie M Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria BC, Canada
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| |
Collapse
|
35
|
Ansaf RB, Ziebart R, Gudapati H, Simoes Torigoe RM, Victorelli S, Passos J, Wyles SP. 3D bioprinting-a model for skin aging. Regen Biomater 2023; 10:rbad060. [PMID: 37501679 PMCID: PMC10369216 DOI: 10.1093/rb/rbad060] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/16/2023] [Accepted: 06/04/2023] [Indexed: 07/29/2023] Open
Abstract
Human lifespan continues to extend as an unprecedented number of people reach their seventh and eighth decades of life, unveiling chronic conditions that affect the older adult. Age-related skin conditions include senile purpura, seborrheic keratoses, pemphigus vulgaris, bullous pemphigoid, diabetic foot wounds and skin cancer. Current methods of drug testing prior to clinical trials require the use of pre-clinical animal models, which are often unable to adequately replicate human skin response. Therefore, a reliable model for aged human skin is needed. The current challenges in developing an aged human skin model include the intrinsic variability in skin architecture from person to person. An ideal skin model would incorporate innate functionality such as sensation, vascularization and regeneration. The advent of 3D bioprinting allows us to create human skin equivalent for use as clinical-grade surgical graft, for drug testing and other needs. In this review, we describe the process of human skin aging and outline the steps to create an aged skin model with 3D bioprinting using skin cells (i.e. keratinocytes, fibroblasts and melanocytes). We also provide an overview of current bioprinted skin models, associated limitations and direction for future research.
Collapse
Affiliation(s)
- Ryeim B Ansaf
- Department of Biology, Colorado State University Pueblo, Pueblo, CO 81001, USA
| | - Rachel Ziebart
- Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
| | | | | | - Stella Victorelli
- Mayo Clinic Department of Physiology and Biomedical Engineering, Rochester, MN 55905, USA
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Rochester, MN 55905, USA
| | - Joao Passos
- Mayo Clinic Department of Physiology and Biomedical Engineering, Rochester, MN 55905, USA
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Rochester, MN 55905, USA
| | | |
Collapse
|
36
|
Lokai T, Albin B, Qubbaj K, Tiwari AP, Adhikari P, Yang IH. A review on current brain organoid technologies from a biomedical engineering perspective. Exp Neurol 2023; 367:114461. [PMID: 37295544 DOI: 10.1016/j.expneurol.2023.114461] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/24/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
Brain organoids are 3D cytoarchitectures resembling the embryonic human brain. This review focuses on current advancements in biomedical engineering methods to develop organoids such as pluripotent stem cells assemblies, quickly aggregated floating culture, hydrogel suspension, microfluidic systems (both photolithography and 3D printing), and brain organoids-on-a-chip. These methods have the potential to create a large impact on neurological disorder studies by creating a model of the human brain investigating pathogenesis and drug screening for individual patients. 3D brain organoid cultures mimic not only features of patients' unknown drug reactions, but also early human brain development at cellular, structural, and functional levels. The challenge of current brain organoids lies in the formation of distinct cortical neuron layers, gyrification, and the establishment of complex neuronal circuitry, as they are critically specialized, developmental aspects. Furthermore, recent advances such as vascularization and genome engineering are in development to overcome the barrier of neuronal complexity. Future technology of brain organoids is needed to improve tissue cross-communication, body axis simulation, cell patterning signals, and spatial-temporal control of differentiation, as engineering methods discussed in this review are rapidly evolving.
Collapse
Affiliation(s)
- Taylor Lokai
- Center for Biomedical Engineering and Science, Mechanical Engineering and Engineering Science, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Bayne Albin
- Center for Biomedical Engineering and Science, Mechanical Engineering and Engineering Science, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Khayzaran Qubbaj
- Center for Biomedical Engineering and Science, Mechanical Engineering and Engineering Science, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Arjun Prasad Tiwari
- Center for Biomedical Engineering and Science, Mechanical Engineering and Engineering Science, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Prashant Adhikari
- Center for Biomedical Engineering and Science, Mechanical Engineering and Engineering Science, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - In Hong Yang
- Center for Biomedical Engineering and Science, Mechanical Engineering and Engineering Science, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA.
| |
Collapse
|
37
|
Mi X, Su Z, Yue X, Ren Y, Yang X, Qiang L, Kong W, Ma Z, Zhang C, Wang J. 3D bioprinting tumor models mimic the tumor microenvironment for drug screening. Biomater Sci 2023; 11:3813-3827. [PMID: 37052182 DOI: 10.1039/d3bm00159h] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Cancer is a severe threat to human life and health and represents the main cause of death globally. Drug therapy is one of the primary means of treating cancer; however, most anticancer medications do not proceed beyond preclinical testing because the conditions of actual human tumors are not effectively mimicked by traditional tumor models. Hence, bionic in vitro tumor models must be developed to screen for anticancer drugs. Three-dimensional (3D) bioprinting technology can produce structures with built-in spatial and chemical complexity and models with accurately controlled structures, a homogeneous size and morphology, less variation across batches, and a more realistic tumor microenvironment (TME). This technology can also rapidly produce such models for high-throughput anticancer medication testing. This review describes 3D bioprinting methods, the use of bioinks in tumor models, and in vitro tumor model design strategies for building complex tumor microenvironment features using biological 3D printing technology. Moreover, the application of 3D bioprinting in vitro tumor models in drug screening is also discussed.
Collapse
Affiliation(s)
- Xuelian Mi
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Zhi Su
- School of Kinesiology, Shanghai University of Sport, 399 Chang Hai Road, Shanghai, 200438, China
| | - Xiaokun Yue
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Ya Ren
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Xue Yang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Lei Qiang
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 611756, China
| | - Weiqing Kong
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, Shandong Province, 266000, China
| | - Zhenjiang Ma
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Changru Zhang
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Jinwu Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
38
|
Zhang S, Chen X, Shan M, Hao Z, Zhang X, Meng L, Zhai Z, Zhang L, Liu X, Wang X. Convergence of 3D Bioprinting and Nanotechnology in Tissue Engineering Scaffolds. Biomimetics (Basel) 2023; 8:biomimetics8010094. [PMID: 36975324 PMCID: PMC10046132 DOI: 10.3390/biomimetics8010094] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Three-dimensional (3D) bioprinting has emerged as a promising scaffold fabrication strategy for tissue engineering with excellent control over scaffold geometry and microstructure. Nanobiomaterials as bioinks play a key role in manipulating the cellular microenvironment to alter its growth and development. This review first introduces the commonly used nanomaterials in tissue engineering scaffolds, including natural polymers, synthetic polymers, and polymer derivatives, and reveals the improvement of nanomaterials on scaffold performance. Second, the 3D bioprinting technologies of inkjet-based bioprinting, extrusion-based bioprinting, laser-assisted bioprinting, and stereolithography bioprinting are comprehensively itemized, and the advantages and underlying mechanisms are revealed. Then the convergence of 3D bioprinting and nanotechnology applications in tissue engineering scaffolds, such as bone, nerve, blood vessel, tendon, and internal organs, are discussed. Finally, the challenges and perspectives of convergence of 3D bioprinting and nanotechnology are proposed. This review will provide scientific guidance to develop 3D bioprinting tissue engineering scaffolds by nanotechnology.
Collapse
Affiliation(s)
- Shike Zhang
- Henan Innovation Center for Functional Polymer Membrane Materials, Henan Key Laboratory of Advanced Nylon Materials and Application, School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xin Chen
- National Engineering Research Center of Wheat and Corn Further Processing, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Mengyao Shan
- Henan Innovation Center for Functional Polymer Membrane Materials, Henan Key Laboratory of Advanced Nylon Materials and Application, School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Zijuan Hao
- Henan Innovation Center for Functional Polymer Membrane Materials, Xinxiang 453000, China
| | - Xiaoyang Zhang
- Henan Innovation Center for Functional Polymer Membrane Materials, Henan Key Laboratory of Advanced Nylon Materials and Application, School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Lingxian Meng
- Henan Innovation Center for Functional Polymer Membrane Materials, Henan Key Laboratory of Advanced Nylon Materials and Application, School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Zhen Zhai
- Henan Innovation Center for Functional Polymer Membrane Materials, Henan Key Laboratory of Advanced Nylon Materials and Application, School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Linlin Zhang
- Henan Innovation Center for Functional Polymer Membrane Materials, Henan Key Laboratory of Advanced Nylon Materials and Application, School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xuying Liu
- Henan Innovation Center for Functional Polymer Membrane Materials, Henan Key Laboratory of Advanced Nylon Materials and Application, School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xianghong Wang
- Henan Innovation Center for Functional Polymer Membrane Materials, Henan Key Laboratory of Advanced Nylon Materials and Application, School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
- Correspondence: ; Tel.: +86-371-67739217
| |
Collapse
|
39
|
Adhikari J, Roy A, Chanda A, D A G, Thomas S, Ghosh M, Kim J, Saha P. Effects of surface patterning and topography on the cellular functions of tissue engineered scaffolds with special reference to 3D bioprinting. Biomater Sci 2023; 11:1236-1269. [PMID: 36644788 DOI: 10.1039/d2bm01499h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The extracellular matrix (ECM) of the tissue organ exhibits a topography from the nano to micrometer range, and the design of scaffolds has been inspired by the host environment. Modern bioprinting aims to replicate the host tissue environment to mimic the native physiological functions. A detailed discussion on the topographical features controlling cell attachment, proliferation, migration, differentiation, and the effect of geometrical design on the wettability and mechanical properties of the scaffold are presented in this review. Moreover, geometrical pattern-mediated stiffness and pore arrangement variations for guiding cell functions have also been discussed. This review also covers the application of designed patterns, gradients, or topographic modulation on 3D bioprinted structures in fabricating the anisotropic features. Finally, this review accounts for the tissue-specific requirements that can be adopted for topography-motivated enhancement of cellular functions during the fabrication process with a special thrust on bioprinting.
Collapse
Affiliation(s)
- Jaideep Adhikari
- School of Advanced Materials, Green Energy and Sensor Systems, Indian Institute of Engineering Science and Technology, Shibpur, Howrah 711103, India
| | - Avinava Roy
- Department of Metallurgy and Materials Engineering, Indian Institute of Engineering Science and Technology, Shibpur, Howrah 711103, India
| | - Amit Chanda
- Department of Mechanical Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Gouripriya D A
- Centre for Interdisciplinary Sciences, JIS Institute of Advanced Studies and Research (JISIASR) Kolkata, JIS University, GP Block, Salt Lake, Sector-5, West Bengal 700091, India.
| | - Sabu Thomas
- School of Chemical Sciences, MG University, Kottayam 686560, Kerala, India
| | - Manojit Ghosh
- Department of Metallurgy and Materials Engineering, Indian Institute of Engineering Science and Technology, Shibpur, Howrah 711103, India
| | - Jinku Kim
- Department of Bio and Chemical Engineering, Hongik University, Sejong, 30016, South Korea.
| | - Prosenjit Saha
- Centre for Interdisciplinary Sciences, JIS Institute of Advanced Studies and Research (JISIASR) Kolkata, JIS University, GP Block, Salt Lake, Sector-5, West Bengal 700091, India.
| |
Collapse
|
40
|
An Automated High-Throughput Screening (HTS) Spotter for 3D Tumor Spheroid Formation. Int J Mol Sci 2023; 24:ijms24021006. [PMID: 36674523 PMCID: PMC9867480 DOI: 10.3390/ijms24021006] [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: 11/18/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Three-dimensional (3D) culture platforms have been adopted in a high-throughput screening (HTS) system to mimic in vivo physiological microenvironments. The automated dispenser has been established commercially to enable spotting or distributing non-viscous or viscous biomaterials onto microplates. However, there are still challenges to the precise and accurate dispensation of cells embedded in hydrogels such as Alginate- and Matrigel-extracellular matrices. We developed and improved an automated contact-free dispensing machine, the ASFA SPOTTER (V5 and V6), which is compatible with 96- and 384-pillar/well plates and 330- and 532-micropillar/well chips for the support of 3D spheroid/organoid models using bioprinting techniques. This enables the distribution of non-viscous and viscous biosamples, including chemical drugs and cancer cells, for large-scale drug screening at high speed and small volumes (20 to 4000 nanoliters) with no damage to cells. The ASFA SPOTTER (V5 and V6) utilizes a contact-free method that minimizes cross-contamination for the dispensation of encapsulated tissue cells with highly viscous scaffolds (over 70%). In particular, the SPOTTER V6 does not require a washing process and offers the advantage of almost no dead volume (defined as additional required sample volume, including a pre-shot and flushing shot for dispensing). It can be successfully applied for the achievement of an organoid culture in automation, with rapid and easy operation, as well as miniaturization for high-throughput screening. In this study, we report the advantages of the ASFA SPOTTER, which distributes standard-sized cell spots with hydrogels onto a 384-pillar/well plate with a fast dispensing speed, small-scale volume, accuracy, and precision.
Collapse
|
41
|
Bioengineering Liver Organoids for Diseases Modelling and Transplantation. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120796. [PMID: 36551002 PMCID: PMC9774794 DOI: 10.3390/bioengineering9120796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Organoids as three-dimension (3D) cellular organizations partially mimic the physiological functions and micro-architecture of native tissues and organs, holding great potential for clinical applications. Advances in the identification of essential factors including physical cues and biochemical signals for controlling organoid development have contributed to the success of growing liver organoids from liver tissue and stem/progenitor cells. However, to recapitulate the physiological properties and the architecture of a native liver, one has to generate liver organoids that contain all the major liver cell types in correct proportions and relative 3D locations as found in a native liver. Recent advances in stem-cell-, biomaterial- and engineering-based approaches have been incorporated into conventional organoid culture methods to facilitate the development of a more sophisticated liver organoid culture resembling a near to native mini-liver in a dish. However, a comprehensive review on the recent advancement in the bioengineering liver organoid is still lacking. Here, we review the current liver organoid systems, focusing on the construction of the liver organoid system with various cell sources, the roles of growth factors for engineering liver organoids, as well as the recent advances in the bioengineering liver organoid disease models and their biomedical applications.
Collapse
|
42
|
Chakraborty J, Chawla S, Ghosh S. Developmental biology-inspired tissue engineering by combining organoids and 3D bioprinting. Curr Opin Biotechnol 2022; 78:102832. [PMID: 36332345 DOI: 10.1016/j.copbio.2022.102832] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/01/2022] [Accepted: 10/02/2022] [Indexed: 12/14/2022]
Abstract
Very few tissue-engineered constructs could achieve the desired results in human clinical trials. The main reason is their inability to recapitulate the cellular conformation, biological, and mechanical functions of the native tissue. Here, we highlight the future avenues of tissue regeneration combining developmental biology, organoids, and 3D bioprinting. A deep mechanistic insight into the embryonic level and recapitulating them would be the most promising strategy in next-generation tissue engineering. Rather than focusing on the adult tissue features, the latest developmental re-engineering strategies replicate the developmental phases of tissue development. Integrating developmental re-engineering with 3D bioprinting can regulate several signaling pathways. This would further help to fabricate mini-organ constructs for transplantation or in vitro screening of drugs using an organ-on-a-chip platform.
Collapse
Affiliation(s)
- Juhi Chakraborty
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Shikha Chawla
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Sourabh Ghosh
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India.
| |
Collapse
|
43
|
Wen L, Tang F. Organoid research on human early development and beyond. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:512-523. [PMID: 37724162 PMCID: PMC10471100 DOI: 10.1515/mr-2022-0028] [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: 08/31/2022] [Accepted: 10/03/2022] [Indexed: 09/20/2023]
Abstract
The organoid field has been developing rapidly during the last decade. Organoids for human pre-, peri- and post-implantation development have opened an avenue to study these biological processes in vitro, which have been hampered by lack of accessible research models for long term. The technologies of four fields, single cell omics sequencing, genome editing and lineage tracing, microfluidics and tissue engineering, have fueled the rapid development of the organoid field. In this review, we will discuss the organoid research on human early development as well as future directions of the organoid field combining with other powerful technologies.
Collapse
Affiliation(s)
- Lu Wen
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, P. R. China
| | - Fuchou Tang
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, P. R. China
| |
Collapse
|
44
|
Petrosyan A, Montali F, Peloso A, Citro A, Byers LN, La Pointe C, Suleiman M, Marchetti A, Mcneill EP, Speer AL, Ng WH, Ren X, Bussolati B, Perin L, Di Nardo P, Cardinale V, Duisit J, Monetti AR, Savino JR, Asthana A, Orlando G. Regenerative medicine technologies applied to transplant medicine. An update. Front Bioeng Biotechnol 2022; 10:1015628. [PMID: 36263358 PMCID: PMC9576214 DOI: 10.3389/fbioe.2022.1015628] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Regenerative medicine (RM) is changing how we think and practice transplant medicine. In regenerative medicine, the aim is to develop and employ methods to regenerate, restore or replace damaged/diseased tissues or organs. Regenerative medicine investigates using tools such as novel technologies or techniques, extracellular vesicles, cell-based therapies, and tissue-engineered constructs to design effective patient-specific treatments. This review illustrates current advancements in regenerative medicine that may pertain to transplant medicine. We highlight progress made and various tools designed and employed specifically for each tissue or organ, such as the kidney, heart, liver, lung, vasculature, gastrointestinal tract, and pancreas. By combing both fields of transplant and regenerative medicine, we can harbor a successful collaboration that would be beneficial and efficacious for the repair and design of de novo engineered whole organs for transplantations.
Collapse
Affiliation(s)
- Astgik Petrosyan
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Filippo Montali
- Department of General Surgery, di Vaio Hospital, Fidenza, Italy
| | - Andrea Peloso
- Visceral Surgery Division, University Hospitals of Geneva, Geneva, Switzerland
| | - Antonio Citro
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Lori N. Byers
- Wake Forest School of Medicine, Winston Salem, NC, United States
| | | | - Mara Suleiman
- Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alice Marchetti
- Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Eoin P. Mcneill
- Department of Pediatric Surgery, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, United States
| | - Allison L Speer
- Department of Pediatric Surgery, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, United States
| | - Wai Hoe Ng
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Xi Ren
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Laura Perin
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Paolo Di Nardo
- Centro Interdipartimentale per la Medicina Rigenerativa (CIMER), Università Degli Studi di Roma Tor Vergata, Rome, Italy
| | - Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Jerome Duisit
- Department of Plastic, Reconstructive and Aesthetic Surgery, CHU Rennes, University of Rennes I, Rennes, France
| | | | | | - Amish Asthana
- Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Giuseppe Orlando
- Wake Forest School of Medicine, Winston Salem, NC, United States
| |
Collapse
|
45
|
Lukomskyj AO, Rao N, Yan L, Pye JS, Li H, Wang B, Li JJ. Stem Cell-Based Tissue Engineering for the Treatment of Burn Wounds: A Systematic Review of Preclinical Studies. Stem Cell Rev Rep 2022; 18:1926-1955. [PMID: 35150392 PMCID: PMC9391245 DOI: 10.1007/s12015-022-10341-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2022] [Indexed: 02/06/2023]
Abstract
Burn wounds are a devastating type of skin injury leading to severe impacts on both patients and the healthcare system. Current treatment methods are far from ideal, driving the need for tissue engineered solutions. Among various approaches, stem cell-based strategies are promising candidates for improving the treatment of burn wounds. A thorough search of the Embase, Medline, Scopus, and Web of Science databases was conducted to retrieve original research studies on stem cell-based tissue engineering treatments tested in preclinical models of burn wounds, published between January 2009 and June 2021. Of the 347 articles retrieved from the initial database search, 33 were eligible for inclusion in this review. The majority of studies used murine models with a xenogeneic graft, while a few used the porcine model. Thermal burn was the most commonly induced injury type, followed by surgical wound, and less commonly radiation burn. Most studies applied stem cell treatment immediately post-burn, with final endpoints ranging from 7 to 90 days. Mesenchymal stromal cells (MSCs) were the most common stem cell type used in the included studies. Stem cells from a variety of sources were used, most commonly from adipose tissue, bone marrow or umbilical cord, in conjunction with an extensive range of biomaterial scaffolds to treat the skin wounds. Overall, the studies showed favourable results of skin wound repair in animal models when stem cell-based tissue engineering treatments were applied, suggesting that such strategies hold promise as an improved therapy for burn wounds.
Collapse
Affiliation(s)
- Alissa Olga Lukomskyj
- Kolling Institute, Faculty of Medicine and Health, University of Sydney, St Leonards, NSW, 2065, Australia
| | - Nikitha Rao
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Lei Yan
- Department of Orthopedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China
| | - Jasmine Sarah Pye
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Haiyan Li
- Chemical and Environmental Engineering, School of Engineering, RMIT University, Melbourne, VIC, 3000, Australia
| | - Bin Wang
- Department of Orthopedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China.
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 315000, China.
| | - Jiao Jiao Li
- Kolling Institute, Faculty of Medicine and Health, University of Sydney, St Leonards, NSW, 2065, Australia.
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| |
Collapse
|
46
|
Parihar A, Pandita V, Khan R. 3D printed human organoids: High throughput system for drug screening and testing in current COVID-19 pandemic. Biotechnol Bioeng 2022; 119:2669-2688. [PMID: 35765706 DOI: 10.1002/bit.28166] [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: 11/05/2021] [Revised: 04/27/2022] [Accepted: 06/24/2022] [Indexed: 11/07/2022]
Abstract
In the current pandemic, scenario the world is facing a huge shortage of effective drugs and other prophylactic medicine to treat patients which created havoc in several countries with poor resources. With limited demand and supply of effective drugs, researchers rushed to repurpose the existing approved drugs for the treatment of COVID-19. The process of drug screening and testing is very costly and requires several steps for validation and treatment efficacy evaluation ranging from in-vitro to in-vivo setups. After these steps, a clinical trial is mandatory for the evaluation of treatment efficacy and side effects in humans. These processes enhance the overall cost and sometimes the lead molecule show adverse effects in humans and the trial ends up in the final stages. Recently with the advent of 3D organoid culture which mimics the human tissue exactly the process of drug screening and testing can be done in a faster and cost-effective manner. Further 3D organoids prepared from stems cells taken from individuals can be beneficial for personalized drug therapy which could save millions of lives. This review discussed approaches and techniques for the synthesis of 3D-printed human organoids for drug screening. The key findings of the usage of organoids for personalized medicine for the treatment of COVID-19 have been discussed. In the end, the key challenges for the wide applicability of human organoids for drug screening with prospects of future orientation have been included. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Arpana Parihar
- Industrial Waste Utilization, Nano and Biomaterials, CSIR-Advanced Materials and Processes Research Institute (AMPRI), Hoshangabad Road, Bhopal, 462026, MP, India
| | - Vasundhara Pandita
- Department of Biosciences, Barkatullah University, Habib Ganj, Bhopal, Madhya Pradesh, 462026, India
| | - Raju Khan
- Industrial Waste Utilization, Nano and Biomaterials, CSIR-Advanced Materials and Processes Research Institute (AMPRI), Hoshangabad Road, Bhopal, 462026, MP, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
47
|
Chia SPS, Kong SLY, Pang JKS, Soh BS. 3D Human Organoids: The Next "Viral" Model for the Molecular Basis of Infectious Diseases. Biomedicines 2022; 10:1541. [PMID: 35884846 PMCID: PMC9312734 DOI: 10.3390/biomedicines10071541] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/07/2023] Open
Abstract
The COVID-19 pandemic has driven the scientific community to adopt an efficient and reliable model that could keep up with the infectious disease arms race. Coinciding with the pandemic, three dimensional (3D) human organoids technology has also gained traction in the field of infectious disease. An in vitro construct that can closely resemble the in vivo organ, organoid technology could bridge the gap between the traditional two-dimensional (2D) cell culture and animal models. By harnessing the multi-lineage characteristic of the organoid that allows for the recapitulation of the organotypic structure and functions, 3D human organoids have emerged as an essential tool in the field of infectious disease research. In this review, we will be providing a comparison between conventional systems and organoid models. We will also be highlighting how organoids played a role in modelling common infectious diseases and molecular mechanisms behind the pathogenesis of causative agents. Additionally, we present the limitations associated with the current organoid models and innovative strategies that could resolve these shortcomings.
Collapse
Affiliation(s)
- Shirley Pei Shan Chia
- Disease Modeling and Therapeutics Laboratory, ASTAR Institute of Molecular and Cell Biology, Singapore 138673, Singapore; (S.P.S.C.); (S.L.Y.K.); (J.K.S.P.)
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Sharleen Li Ying Kong
- Disease Modeling and Therapeutics Laboratory, ASTAR Institute of Molecular and Cell Biology, Singapore 138673, Singapore; (S.P.S.C.); (S.L.Y.K.); (J.K.S.P.)
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Jeremy Kah Sheng Pang
- Disease Modeling and Therapeutics Laboratory, ASTAR Institute of Molecular and Cell Biology, Singapore 138673, Singapore; (S.P.S.C.); (S.L.Y.K.); (J.K.S.P.)
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Boon-Seng Soh
- Disease Modeling and Therapeutics Laboratory, ASTAR Institute of Molecular and Cell Biology, Singapore 138673, Singapore; (S.P.S.C.); (S.L.Y.K.); (J.K.S.P.)
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| |
Collapse
|
48
|
Staros R, Michalak A, Rusinek K, Mucha K, Pojda Z, Zagożdżon R. Perspectives for 3D-Bioprinting in Modeling of Tumor Immune Evasion. Cancers (Basel) 2022; 14:cancers14133126. [PMID: 35804898 PMCID: PMC9265021 DOI: 10.3390/cancers14133126] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/31/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023] Open
Abstract
In a living organism, cancer cells function in a specific microenvironment, where they exchange numerous physical and biochemical cues with other cells and the surrounding extracellular matrix (ECM). Immune evasion is a clinically relevant phenomenon, in which cancer cells are able to direct this interchange of signals against the immune effector cells and to generate an immunosuppressive environment favoring their own survival. A proper understanding of this phenomenon is substantial for generating more successful anticancer therapies. However, classical cell culture systems are unable to sufficiently recapture the dynamic nature and complexity of the tumor microenvironment (TME) to be of satisfactory use for comprehensive studies on mechanisms of tumor immune evasion. In turn, 3D-bioprinting is a rapidly evolving manufacture technique, in which it is possible to generate finely detailed structures comprised of multiple cell types and biomaterials serving as ECM-analogues. In this review, we focus on currently used 3D-bioprinting techniques, their applications in the TME research, and potential uses of 3D-bioprinting in modeling of tumor immune evasion and response to immunotherapies.
Collapse
Affiliation(s)
- Rafał Staros
- Department of Immunology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-006 Warsaw, Poland; (R.S.); (K.M.)
| | - Agata Michalak
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Institute of Oncology, 02-781 Warsaw, Poland; (A.M.); (K.R.); (Z.P.)
| | - Kinga Rusinek
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Institute of Oncology, 02-781 Warsaw, Poland; (A.M.); (K.R.); (Z.P.)
| | - Krzysztof Mucha
- Department of Immunology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-006 Warsaw, Poland; (R.S.); (K.M.)
| | - Zygmunt Pojda
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Institute of Oncology, 02-781 Warsaw, Poland; (A.M.); (K.R.); (Z.P.)
| | - Radosław Zagożdżon
- Department of Immunology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-006 Warsaw, Poland; (R.S.); (K.M.)
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Institute of Oncology, 02-781 Warsaw, Poland; (A.M.); (K.R.); (Z.P.)
- Department of Clinical Immunology, Medical University of Warsaw, 02-006 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-502-14-72; Fax: +48-22-502-21-59
| |
Collapse
|
49
|
Ro J, Kim J, Cho YK. Recent advances in spheroid-based microfluidic models to mimic the tumour microenvironment. Analyst 2022; 147:2023-2034. [PMID: 35485712 DOI: 10.1039/d2an00172a] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Three-dimensional (3D) multicellular spheroid models can recapitulate the human tumour microenvironment with more accuracy than conventional cell culture models, as they include complex architectural structures and dynamic cellular interactions. Among the diverse platforms for spheroid formation, microfluidic platforms have been extensively applied to study spheroids because they can mimic the in vivo microenvironment. This review provides an overview of the advantages of 3D spheroid cultures with a summary of the recent applications for tumour microenvironment-focused cellular interactions, as well as the studies on spheroids and external stimuli. These 3D tumour spheroid-based microfluidic devices will provide a platform for a better understanding of cellular and external interactions, as well as the discovery of cancer therapeutics.
Collapse
Affiliation(s)
- Jooyoung Ro
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Korea. .,Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Korea
| | - Junyoung Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Korea.
| | - Yoon-Kyoung Cho
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Korea. .,Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, Korea
| |
Collapse
|
50
|
Hsu YT, Ho MH, Lee SP, Kao CY. Preparation of Biomimetic 3D Gastric Model with Photo-Curing Resin and Evaluation the Growth of Helicobacter pylori. Polymers (Basel) 2021; 13:polym13203593. [PMID: 34685352 PMCID: PMC8538753 DOI: 10.3390/polym13203593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/12/2021] [Accepted: 10/16/2021] [Indexed: 11/20/2022] Open
Abstract
Three-dimensional (3D) printing technology is now widely used in biomedical developments. Especially, photo-curing systems provide high resolution and precision. The current goal of biomedical 3D printing technology is the printing of human organs, but the current commercial photo-curable materials generally have high mechanical strength that cannot meet the mechanical properties of the object to be printed. In this research, a gastric model was printed using a photo-curing 3D printing technique. To mimic the wrinkle pattern of human gastric tissue, cis-1,4 polyisoprene with different reactive diluents was mixed and identified a formulation that produced a print with human gastric softness. This research discussed the effect of the Young’s modulus of the material and elucidated the relationship between the degree of conversion rate and viscosity. After modifying the cis-1,4 polyisoprene surface from hydrophobic to hydrophilic, we then evaluated its adhesion efficiency for gastric mucin and the gastrointestinal-inhabiting bacterium Helicobacter pylori.
Collapse
Affiliation(s)
- Yu-Tung Hsu
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 10607, Taiwan; (Y.-T.H.); (M.-H.H.)
| | - Ming-Hu Ho
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 10607, Taiwan; (Y.-T.H.); (M.-H.H.)
- R&D Center for Membrane Technology, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
| | - Shiao-Pieng Lee
- Department of Dentistry, Division of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei 11490, Taiwan;
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 11490, Taiwan
| | - Chen-Yu Kao
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
- Biomedical Engineering Research Center, National Defense Medical Center, Taipei 11490, Taiwan
- Correspondence: ; Tel.: +886-2-2730-3676
| |
Collapse
|