1
|
Gnatowski P, Ansariaghmiuni M, Piłat E, Poostchi M, Kucińska-Lipka J, Yazdi MK, Ryl J, Ashrafizadeh M, Mottaghitalab F, Farokhi M, Saeb MR, Bączek T, Chen C, Lu Q. Hydrogel membranes in organ-on-a-chip devices: A review. Colloids Surf B Biointerfaces 2025; 251:114591. [PMID: 40054047 DOI: 10.1016/j.colsurfb.2025.114591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/31/2025] [Accepted: 02/20/2025] [Indexed: 04/15/2025]
Abstract
Organ-on-a-chip (OoC) devices represent advanced in vitro models enabling to mimic the human tissue architecture function and physiology, providing a promising alternative to the traditional animal testing methods. These devices combine the microfluidics with soft materials, specifically hydrogel membranes (HMs) for mimicking the extracellular matrix (ECM) and biological barriers, such as the blood-brain barrier (BBB). Hydrogels are ideal biomaterials for OoC systems because of their tunable properties, biocompatibility, biodegradability, and microscale self-assembly. The integration of HMs with OoC devices provides an effective way to develop dynamic, biologically relevant environments for supporting living cells targeted at drug discovery, disease modeling, and personalized medicine. Recent advancements in fabrication technologies such as additive manufacturing (3D printing), photolithography, and bioprinting have additionally advanced development of such systems. This review aims to outline the role of HMs in OoC platforms, highlighting their material properties, self-assembly behavior, and also challenges associated with their fabrication. Additionally, we visualize and discuss the latest progress made in utilizing HMs for applications in tissue engineering, drug development, and biosensing, with a focus on their interface dynamics and structural self-organization. The future perspective on OoC technology has also been patterned in order to provide a broader image on integration of OoC and HMs with personalized medicine and advanced drug delivery systems.
Collapse
Affiliation(s)
- Przemysław Gnatowski
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Narutowicza St. 11/12, Gdańsk 80-233, Poland; Department of Environmental Toxicology, Faculty of Health Sciences with the Institute of Maritime and Tropical Medicine, Medical University of Gdańsk, Dębowa 23A, Gdańsk 80-204, Poland
| | - Maryam Ansariaghmiuni
- Polymer Research Laboratory, Department of Chemistry, Sharif University of Technology, Tehran 11365-9516, Iran
| | - Edyta Piłat
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Narutowicza St. 11/12, Gdańsk 80-233, Poland
| | - Maryam Poostchi
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Narutowicza St. 11/12, Gdańsk 80-233, Poland
| | - Justyna Kucińska-Lipka
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Narutowicza St. 11/12, Gdańsk 80-233, Poland
| | - Mohsen Khodadadi Yazdi
- Division of Electrochemistry and Surface Physical Chemistry, Faculty of Applied Physics and Mathematics, Gdańsk University of Technology, Narutowicza 11/12, Gdańsk 80-233, Poland; Advanced Materials Center, Gdańsk University of Technology, Narutowicza 11/12, Gdańsk 80-233, Poland
| | - Jacek Ryl
- Division of Electrochemistry and Surface Physical Chemistry, Faculty of Applied Physics and Mathematics, Gdańsk University of Technology, Narutowicza 11/12, Gdańsk 80-233, Poland; Advanced Materials Center, Gdańsk University of Technology, Narutowicza 11/12, Gdańsk 80-233, Poland
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Fatemeh Mottaghitalab
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Reza Saeb
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, J. Hallera 107, Gdańsk 80-416, Poland.
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, J. Hallera 107, Gdańsk 80-416, Poland.
| | - Chu Chen
- Department of Cardiology, Cardiac Arrhythmia Center, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Qi Lu
- Department of Cardiology, Cardiac Arrhythmia Center, Affiliated Hospital of Nantong University, Nantong, 226001, China.
| |
Collapse
|
2
|
Liu L, Ding Z, Huang Y, Zou J. Magnesium Ion/Gallic Acid MOF-Laden Multifunctional Acellular Matrix Hydrogels for Diabetic Wound Healing. ACS APPLIED BIO MATERIALS 2025. [PMID: 40317115 DOI: 10.1021/acsabm.4c01979] [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: 05/07/2025]
Abstract
The main objective for diabetic wound treatment is the design of a functional dressing that scavenges free radicals, alleviates inflammation, and is antibacterial while promoting neovascularization. Herein, a multifunctional acellular matrix hydrogel was prepared with the antimicrobial peptide jelleine-1 and a magnesium ion/gallic acid metal framework to exhibit antioxidant, anti-inflammatory, and proangiogenesis effects in diabetic wounds. The prepared hydrogel termed Gel-J-MOF efficiently released gallic acid in the acidic microenvironment of the diabetic wound, scavenged excess free radicals in vitro, and effectively reduced the levels of inflammation by regulating M2 macrophage polarization in vivo. The antimicrobial peptide jelleine-1 in the composite hydrogel effectively inhibited S. aureus and E. coli in vitro, promoting a suitable microenvironment for wound healing. In the later stage of wound healing, the composite hydrogel stimulated angiogenesis, accelerating the re-epithelialization and collagen deposition in the wound. In conclusion, this multifunctional composite hydrogel provides a regulated microenvironment for treating diabetic wounds and, therefore, has significant potential application promise in the treatment of chronic diabetic wounds.
Collapse
Affiliation(s)
- Lutong Liu
- Department of General Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, China
- Beijing Allgens Medical Science and Technology Co., Ltd., Beijing 100176, China
| | - Zhaozhao Ding
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China
| | - Yong Huang
- Department of General Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, China
| | - Junwei Zou
- Department of General Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, China
| |
Collapse
|
3
|
Addario G, Moroni L, Mota C. Kidney Fibrosis In Vitro and In Vivo Models: Path Toward Physiologically Relevant Humanized Models. Adv Healthc Mater 2025; 14:e2403230. [PMID: 39906010 PMCID: PMC11973949 DOI: 10.1002/adhm.202403230] [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: 08/27/2024] [Revised: 01/14/2025] [Indexed: 02/06/2025]
Abstract
Chronic kidney disease (CKD) affects over 10% of the global population and is a leading cause of mortality. Kidney fibrosis, a key endpoint of CKD, disrupts nephron tubule anatomy and filtration function, and disease pathomechanisms are not fully understood. Kidney fibrosis is currently investigated with in vivo models, that gradually support the identification of possible mechanisms of fibrosis, but with limited translational research, as they do not fully recapitulate human kidney physiology, metabolism, and molecular pathways. In vitro 2D cell culture models are currently used, as a starting point in disease modeling and pharmacology, however, they lack the 3D kidney architecture complexity and functions. The failure of several therapies and drugs in clinical trials highlights the urgent need for advanced 3D in vitro models. This review discusses the urinary system's anatomy, associated diseases, and diagnostic methods, including biomarker analysis and tissue biopsy. It evaluates 2D and in vivo models, highlighting their limitations. The review explores the state-of-the-art 3D-humanized in vitro models, such as 3D cell aggregates, on-chip models, biofabrication techniques, and hybrid models, which aim to mimic kidney morphogenesis and functions. These advanced models hold promise for translating new therapies and drugs for kidney fibrosis into clinics.
Collapse
Affiliation(s)
- Gabriele Addario
- Department of Complex Tissue RegenerationMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityER Maastricht6229The Netherlands
| | - Lorenzo Moroni
- Department of Complex Tissue RegenerationMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityER Maastricht6229The Netherlands
| | - Carlos Mota
- Department of Complex Tissue RegenerationMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityER Maastricht6229The Netherlands
| |
Collapse
|
4
|
Banigo AT, Nauta L, Zoetebier B, Karperien M. Hydrogel-Based Bioinks for Coaxial and Triaxial Bioprinting: A Review of Material Properties, Printing Techniques, and Applications. Polymers (Basel) 2025; 17:917. [PMID: 40219306 PMCID: PMC11991663 DOI: 10.3390/polym17070917] [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: 02/10/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Three-dimensional bioprinting technology has emerged as a rapidly advancing multidisciplinary field with significant potential for tissue engineering applications. This technology enables the formation of complex tissues and organs by utilizing hydrogels, with or without cells, as scaffolds or structural supports. Among various bioprinting methods, advanced bioprinting using coaxial and triaxial nozzles stands out as a promising technique. Coaxial bioprinting technique simultaneously deposits two material streams through a coaxial nozzle, enabling controlled formation of an outer shell and inner core construct. In contrast, triaxial bioprinting utilizes three material streams namely the outer shell, inner shell and inner core to fabricate more complex constructs. Despite the growing interest in 3D bioprinting, the development of suitable cell-laden bioinks for creating complex tissues remains unclear. To address this gap, a systematic review was conducted using the preferred reporting items for systematic reviews and meta-analyses (PRISMA) flowchart, collecting 1621 papers from various databases, including Web of Science, PUBMED, SCOPUS, and Springer Link. After careful selection, 85 research articles focusing on coaxial and triaxial bioprinting were included in the review. Specifically, 77 research articles concentrated on coaxial bioprinting and 11 focused on triaxial bioprinting, with 3 covering both techniques. The search, conducted between 1 April and 30 September 2023, had no restrictions on publication date, and no meta-analyses were carried out due to the heterogeneity of studies. The primary objective of this review is to assess and identify the most commonly occurring cell-laden bioinks critical for successful advancements in bioprinting technologies. Specifically, the review focuses on delineating the commonly explored bioinks utilized in coaxial and triaxial bioprinting approaches. It focuses on evaluating the inherent merits of these bioinks, systematically comparing them while emphasizing their classifications, essential attributes, properties, and potential limitations within the domain of tissue engineering. Additionally, the review considers the applications of these bioinks, offering comprehensive insights into their efficacy and utility in the field of bioprinting technology. Overall, this review provides a comprehensive overview of some conditions of the relevant hydrogel bioinks used for coaxial and triaxial bioprinting of tissue constructs. Future research directions aimed at advancing the field are also briefly discussed.
Collapse
Affiliation(s)
| | | | | | - Marcel Karperien
- Department of Developmental BioEngineering, Faculty of Science and Technology and TechMed Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands; (A.T.B.); (L.N.); (B.Z.)
| |
Collapse
|
5
|
Kiranmai G, Chameettachal S, Sriya Y, Duin S, Lode A, Gelinsky M, Akkineni AR, Pati F. Recent trends in the development of in vitro3D kidney models. Biofabrication 2025; 17:022010. [PMID: 39993331 DOI: 10.1088/1758-5090/adb999] [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: 08/23/2024] [Accepted: 02/24/2025] [Indexed: 02/26/2025]
Abstract
The kidneys are vital for maintaining bodily homeostasis and are susceptible to various diseases that disrupt their function. Traditionally, research on kidney diseases has relied on animal models and simplistic two-dimensional cell cultures, which do not fully replicate human tissue pathology. To address this, recent advances focus on developing advanced 3D biomimeticin vitromodels using human-derived cells. These models mimic healthy and diseased kidney tissues with specificity, replicating key elements like glomerular and tubular structures through tissue engineering. By closely mimicking human physiology, they provide a promising platform for studying renal disorders, drug-induced nephrotoxicity, and evaluating new therapies. However, the challenges include optimizing scalability, reproducibility, and long-term stability to enhance reliability in research and clinical applications. This review highlights the transformative potential of 3D biomimeticin vitrokidney models in advancing biomedical research and clinical applications. By focusing on human-specific cell cultures and tissue engineering techniques, these models aim to overcome the limitations of conventional animal models and simplistic 2D cell cultures. The review discusses in detail the various types of biomimetic kidney models currently under development, their specific applications, and the innovative approaches used to construct them. It also addresses the challenges and limitations associated with these models for their widespread adoption and reliability in research settings.
Collapse
Affiliation(s)
- Gaddam Kiranmai
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502285, India
| | - Shibu Chameettachal
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502285, India
| | - Yeleswarapu Sriya
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502285, India
| | - Sarah Duin
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, Dresden 01307, Germany
| | - Anja Lode
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, Dresden 01307, Germany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, Dresden 01307, Germany
| | - Ashwini Rahul Akkineni
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, Dresden 01307, Germany
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502285, India
| |
Collapse
|
6
|
Pal V, Gupta D, Liu S, Namli I, Rizvi SHA, Yilmaz YO, Haugh L, Gerhard EM, Ozbolat IT. Interparticle Crosslinked Ion-responsive Microgels for 3D and 4D (Bio)printing Applications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635095. [PMID: 39975099 PMCID: PMC11838323 DOI: 10.1101/2025.01.28.635095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Microgels offer unique advantages over bulk hydrogels due to their improved diffusion limits for oxygen and nutrients. Particularly, stimuli-responsive microgels with inherently bioactive and self-supporting properties emerge as highly promising biomaterials. This study unveils the development of interparticle-crosslinked, self-supporting, ion-responsive microgels tailored for 3D and 4D (bio)printing applications. A novel strategy was proposed to develop microgels that enabled interparticle crosslinking, eliminating the need for filler hydrogels and preserving essential microscale void spaces to support cell migration and vascularization. Additionally, these microgels possessed unique, ion-responsive shrinking behavior primarily by the Hofmeister effect, reversible upon the removal of the stimulus. Two types of microgels, spherical (µS) and random-shaped (µR), were fabricated, with µR exhibiting superior mechanical properties and higher packing density. Fabricated microgel-based constructs supported angiogenesis with tunable vessel size based on interstitial void spaces while demonstrating excellent shear-thinning and self-healing properties and high print fidelity. Various bioprinting techniques were employed and validated using these microgels, including extrusion-based, embedded, intraembedded, and aspiration-assisted bioprinting, facilitating the biofabrication of scalable constructs. Multi-material 4D printing was achieved by combining ion-responsive microgels with non-responsive microgels, enabling programmable shape transformations upon exposure to ionic solutions. Utilizing 4D printing, complex, dynamic structures were generated such as coiling filaments, grippers, and folding sheets, providing a foundation for the development of advanced tissue models and devices for regenerative medicine and soft robotics, respectively.
Collapse
|
7
|
Liu J, Song Q, Yin W, Li C, An N, Le Y, Wang Q, Feng Y, Hu Y, Wang Y. Bioactive scaffolds for tissue engineering: A review of decellularized extracellular matrix applications and innovations. EXPLORATION (BEIJING, CHINA) 2025; 5:20230078. [PMID: 40040827 PMCID: PMC11875452 DOI: 10.1002/exp.20230078] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/12/2024] [Indexed: 03/06/2025]
Abstract
Decellularized extracellular matrix (dECM) offers a three-dimensional, non-immunogenic scaffold, enriched with bioactive components, making it a suitable candidate for tissue regeneration. Although dECM-based scaffolds have been successfully implemented in preclinical and clinical settings within tissue engineering and regenerative medicine, the mechanisms of tissue remodeling and functional restoration are not fully understood. This review critically assesses the state-of-the-art in dECM scaffolds, including decellularization techniques for various tissues, quality control and cross-linking. It highlights the functional properties of dECM components and their latest applications in multiorgan tissue engineering and biomedicine. Additionally, the review addresses current challenges and limitations of decellularized scaffolds and offers perspectives on future directions in the field.
Collapse
Affiliation(s)
- Juan Liu
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Key Laboratory of Digital Intelligence HepatologyMinistry of EducationSchool of Clinical MedicineTsinghua UniversityBeijingChina
| | - Qingru Song
- Clinical Translational Science CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
| | - Wenzhen Yin
- Clinical Translational Science CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
| | - Chen Li
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- College of Chemistry and Life SciencesBeijing University of TechnologyBeijingChina
| | - Ni An
- Clinical Translational Science CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
| | - Yinpeng Le
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Institute of Smart Biomedical MaterialsSchool of Materials Science and EngineeringZhejiang Sci‐Tech UniversityHangzhouPeople's Republic of China
| | - Qi Wang
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Department of Hepatobiliary and Pancreatic SurgeryThe First Hospital of Jilin UniversityJilin UniversityChangchunChina
| | - Yutian Feng
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
| | - Yuelei Hu
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Department of Hepatobiliary and Pancreatic SurgeryThe First Hospital of Jilin UniversityJilin UniversityChangchunChina
| | - Yunfang Wang
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Key Laboratory of Digital Intelligence HepatologyMinistry of EducationSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Clinical Translational Science CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
| |
Collapse
|
8
|
Derman ID, Moses JC, Rivera T, Ozbolat IT. Understanding the cellular dynamics, engineering perspectives and translation prospects in bioprinting epithelial tissues. Bioact Mater 2025; 43:195-224. [PMID: 39386221 PMCID: PMC11462153 DOI: 10.1016/j.bioactmat.2024.09.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/04/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
The epithelium is one of the important tissues in the body as it plays a crucial barrier role serving as a gateway into and out of the body. Most organs in the body contain an epithelial tissue component, where the tightly connected, organ-specific epithelial cells organize into cysts, invaginations, or tubules, thereby performing distinct to endocrine or exocrine secretory functions. Despite the significance of epithelium, engineering functional epithelium in vitro has remained a challenge due to it is special architecture, heterotypic composition of epithelial tissues, and most importantly, difficulty in attaining the apico-basal and planar polarity of epithelial cells. Bioprinting has brought a paradigm shift in fabricating such apico-basal polarized tissues. In this review, we provide an overview of epithelial tissues and provide insights on recapitulating their cellular arrangement and polarization to achieve epithelial function. We describe the different bioprinting techniques that have been successful in engineering polarized epithelium, which can serve as in vitro models for understanding homeostasis and studying diseased conditions. We also discuss the different attempts that have been investigated to study these 3D bioprinted engineered epithelium for preclinical use. Finally, we highlight the challenges and the opportunities that need to be addressed for translation of 3D bioprinted epithelial tissues towards paving way for personalized healthcare in the future.
Collapse
Affiliation(s)
- Irem Deniz Derman
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
| | - Joseph Christakiran Moses
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
| | - Taino Rivera
- Biomedical Engineering Department, Penn State University, University Park, PA, 16802, USA
| | - Ibrahim T. Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Biomedical Engineering Department, Penn State University, University Park, PA, 16802, USA
- Materials Research Institute, Penn State University, University Park, PA, 16802, USA
- Cancer Institute, Penn State University, University Park, PA, 16802, USA
- Neurosurgery Department, Penn State University, University Park, PA, 16802, USA
- Department of Medical Oncology, Cukurova University, Adana, 01330, Turkey
| |
Collapse
|
9
|
Lu J, Shi X, Zhou Z, Lu N, Chu G, Jin H, Zhu L, Chen A. Enhancing Fracture Healing with 3D Bioprinted Hif1a-Overexpressing BMSCs Hydrogel: A Novel Approach to Accelerated Bone Repair. Adv Healthc Mater 2025; 14:e2402415. [PMID: 39580668 DOI: 10.1002/adhm.202402415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/17/2024] [Indexed: 11/26/2024]
Abstract
Addressing the urgent need for effective fracture treatments, this study investigates the efficacy of a 3D bioprinted biomimetic hydrogel, enriched with bone marrow mesenchymal stem cells (BMSCs) and targeted hypoxia-inducible factor 1 alpha (Hif1a) gene activation, in enhancing fracture healing. A photocross-linkable bioink, gelatin methacryloyl bone matrix anhydride (GBMA) is developed, and selected its 5% concentration for bioink formulation. Rat BMSCs are isolated and combined with GBMA to create the GBMA@BMSCs bioink. This bioink is then used in 3D bioprinting to fabricate a hydrogel for application in a rat femoral fracture model. Through transcriptome sequencing, WGCNA, and Venn analysis, the hypoxia-inducible factor Hif1a is identified as a critical gene in the fracture healing process. In vitro studies showed that Hif1a promoted BMSC proliferation, chondrogenic differentiation, and cartilage matrix stability. The in vivo application of the GBMA@BMSCs hydrogel with Hif1a overexpression significantly accelerated fracture healing, evidenced by early and enhanced cartilage callus formation. The study demonstrates that 3D bioprinting of GBMA@BMSCs hydrogel, particularly with Hif1a-enhanced BMSCs, offers a promising approach for rapid and effective fracture repair.
Collapse
Affiliation(s)
- Jiajia Lu
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, P. R. China
- Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200001, P. R. China
| | - Xiaojian Shi
- Department of Orthopedic Trauma, Haimen People's Hospital of Jiangsu Province, Haimen, 226100, P. R. China
| | - Zhibin Zhou
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang, 110016, P. R. China
| | - Nan Lu
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, P. R. China
| | - Guangxin Chu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, 110016, China
| | - Hai Jin
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, 110016, China
| | - Lei Zhu
- Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200001, P. R. China
| | - Aimin Chen
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, P. R. China
| |
Collapse
|
10
|
Brouki Milan P, Masoumi F, Biazar E, Zare Jalise S, Mehrabi A. Exploiting the Potential of Decellularized Extracellular Matrix (ECM) in Tissue Engineering: A Review Study. Macromol Biosci 2025; 25:e2400322. [PMID: 39412772 DOI: 10.1002/mabi.202400322] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/03/2024] [Indexed: 01/14/2025]
Abstract
While significant progress has been made in creating polymeric structures for tissue engineering, the therapeutic application of these scaffolds remains challenging owing to the intricate nature of replicating the conditions of native organs and tissues. The use of human-derived biomaterials for therapeutic purposes closely imitates the properties of natural tissue, thereby assisting in tissue regeneration. Decellularized extracellular matrix (dECM) scaffolds derived from natural tissues have become popular because of their unique biomimetic properties. These dECM scaffolds can enhance the body's ability to heal itself or be used to generate new tissues for restoration, expanding beyond traditional tissue transfers and transplants. Enhanced knowledge of how ECM scaffold materials affect the microenvironment at the injury site is expected to improve clinical outcomes. In this review, recent advancements in dECM scaffolds are explored and relevant perspectives are offered, highlighting the development and application of these scaffolds in tissue engineering for various organs, such as the skin, nerve, bone, heart, liver, lung, and kidney.
Collapse
Affiliation(s)
- Peiman Brouki Milan
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, 144-961-4535, Iran
| | - Farimah Masoumi
- School of Medicine, Tonekabon Branch, Islamic Azad University, Tonekabon, 468-416-1167, Iran
| | - Esmaeil Biazar
- Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, 468-416-1167, Iran
| | - Saeedeh Zare Jalise
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, 371-364-9373, Iran
| | - Arezou Mehrabi
- School of Medicine, Tonekabon Branch, Islamic Azad University, Tonekabon, 468-416-1167, Iran
| |
Collapse
|
11
|
Veser C, Carlier A, Dubois V, Mihăilă SM, Swapnasrita S. Embracing sex-specific differences in engineered kidney models for enhanced biological understanding of kidney function. Biol Sex Differ 2024; 15:99. [PMID: 39623463 PMCID: PMC11613810 DOI: 10.1186/s13293-024-00662-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 10/16/2024] [Indexed: 12/06/2024] Open
Abstract
In vitro models serve as indispensable tools for advancing our understanding of biological processes, elucidating disease mechanisms, and establishing screening platforms for drug discovery. Kidneys play an instrumental role in the transport and elimination of drugs and toxins. Nevertheless, despite the well-documented inter-individual variability in kidney function and the multifaceted nature of renal diseases-spanning from their origin, trigger and which segment of the kidney is affected-to presentation, progression and prognosis, few studies take into consideration the variable of sex. Notably, the inherent disparities between female and male biology warrants a more comprehensive representation within in vitro models of the kidney. The omission of sex as a fundamental biological variable carries the substantial risk of overlooking sex-specific mechanisms implicated in health and disease, along with potential differences in drug responsiveness and toxicity profiles between sexes. This review emphasizes the importance of incorporating cellular, biological and functional sex-specific features of renal activity in health and disease in in vitro models. For that, we thoroughly document renal sex-specific features and propose a strategic experimental framework to integrate sex-based differences into human kidney in vitro models by outlining critical design criteria to elucidate sex-based features at cellular and tissue levels. The goal is to enhance the accuracy of models to unravel renal mechanisms, and improve our understanding of their impact on drug efficacy and safety profiles, paving the way for a more comprehensive understanding of patient-specific treatment modalities.
Collapse
Affiliation(s)
- Charlotte Veser
- Utrecht Institute for Pharmaceutical Sciences, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Aurélie Carlier
- MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands
| | - Vanessa Dubois
- Basic and Translational Endocrinology (BaTE), Department of Basic and Applied Medical Sciences, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Silvia M Mihăilă
- Utrecht Institute for Pharmaceutical Sciences, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands.
| | - Sangita Swapnasrita
- MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands.
| |
Collapse
|
12
|
Li F, XinHuang, Wang R, Li Y, Wu L, Qiao X, Zhong Y, Gong G, Huang W. Collagen-based materials in male genitourinary diseases and tissue regeneration. COLLAGEN AND LEATHER 2024; 6:36. [DOI: 10.1186/s42825-024-00185-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 01/03/2025]
Abstract
AbstractMale genitourinary dysfunction causes serious physical or mental distress, such as infertility and psychological harm, which leads to impaired quality of life. Current conventional treatments involving drug therapy, surgical repair, and tissue grafting have a limited effect on recovering the function and fertility of the genitourinary organs. To address these limitations, various biomaterials have been explored, with collagen-based materials increasingly gaining attention for reconstructing the male genitourinary system due to their superior biocompatibility, biodegradability, low antigenicity, biomimetic 3D matrix characteristics, hemostatic efficacy, and tissue regeneration capabilities. This review covers the recent biomedical applications of collagen-based materials including treatment of erectile dysfunction, premature ejaculation, penile girth enlargement, prostate cancer, Peyronie's disease, chronic kidney disease, etc. Although there are relatively few clinical trials, the promising results of the existing studies on animal models reveal a bright future for collagen-based materials in the treatment of male genitourinary diseases.
Graphic Abstract
Collapse
|
13
|
Addario G, Fernández‐Pérez J, Formica C, Karyniotakis K, Herkens L, Djudjaj S, Boor P, Moroni L, Mota C. 3D Humanized Bioprinted Tubulointerstitium Model to Emulate Renal Fibrosis In Vitro. Adv Healthc Mater 2024; 13:e2400807. [PMID: 39152919 PMCID: PMC11582511 DOI: 10.1002/adhm.202400807] [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: 03/01/2024] [Revised: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Chronic kidney disease (CKD) leads to a gradual loss of kidney function, with fibrosis as pathological endpoint, which is characterized by extracellular matrix (ECM) deposition and remodeling. Traditionally, in vivo models are used to study interstitial fibrosis, through histological characterization of biopsy tissue. However, ethical considerations and the 3Rs (replacement, reduction, and refinement) regulations emphasizes the need for humanized 3D in vitro models. This study introduces a bioprinted in vitro model which combines primary human cells and decellularized and partially digested extracellular matrix (ddECM). A protocol was established to decellularize kidney pig tissue and the ddECM was used to encapsulate human renal cells. To investigate fibrosis progression, cells were treated with transforming growth factor beta 1 (TGF-β1), and the mechanical properties of the ddECM hydrogel were modulated using vitamin B2 crosslinking. The bioprinting perfusable model replicates the renal tubulointerstitium. Results show an increased Young's modulus over time, together with the increase of ECM components and cell dedifferentiation toward myofibroblasts. Multiple fibrotic genes resulted upregulated, and the model closely resembled fibrotic human tissue in terms of collagen deposition. This 3D bioprinted model offers a more physiologically relevant platform for studying kidney fibrosis, potentially improving disease progression research and high-throughput drug screening.
Collapse
Affiliation(s)
- Gabriele Addario
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ETThe Netherlands
| | - Julia Fernández‐Pérez
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ETThe Netherlands
| | - Chiara Formica
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ETThe Netherlands
| | | | - Lea Herkens
- Institute of PathologyRWTH University of Aachen52074AachenGermany
| | - Sonja Djudjaj
- Institute of PathologyRWTH University of Aachen52074AachenGermany
| | - Peter Boor
- Institute of PathologyRWTH University of Aachen52074AachenGermany
- Electron Microscopy FacilityRWTH University of Aachen52074AachenGermany
| | - Lorenzo Moroni
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ETThe Netherlands
| | - Carlos Mota
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ETThe Netherlands
| |
Collapse
|
14
|
Wang Y, Pang F, Lai S, Cai R, Lai C, Yu Z, Zhu Y, Wu M, Zhang H, Kong C. Numerical Analysis of the Cell Droplet Loading Process in Cell Printing. MICROMACHINES 2024; 15:1335. [PMID: 39597146 PMCID: PMC11596778 DOI: 10.3390/mi15111335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
Cell printing is a promising technology in tissue engineering, with which the complex three-dimensional tissue constructs can be formed by sequentially printing the cells layer by layer. Though some cell printing experiments with commercial inkjet printers show the possibility of this idea, there are some problems, such as cell damage due the mechanical impact during cell direct writing, which include two processes of cell ejection and cell landing. Cell damage observed during the bioprinting process is often simply attributed to interactions between cells and substrate. However, in reality, cell damage can also arise from complex mechanical effects caused by collisions between cell droplets during continuous printing processes. The objective of this research is to numerically simulate the collision effects between continuously printed cell droplets within the bioprinting process, with a particular focus on analyzing the consequent cell droplet deformation and stress distribution. The influence of gravity force was ignored, cell droplet landing was divided into four phases, the first phase is cell droplet free falling at a certain velocity; the second phase is the collision between the descending cell droplet and the pre-existing cell droplets that have been previously printed onto the substrate. This collision results in significant deformation of the cell membranes of both cell droplets in contact; the third phase is the cell droplet hitting a rigid body substrate; the fourth phase is the cell droplet being bounced. We conducted a qualitative analysis of the stress and strain of cell droplets during the cell printing process to evaluate the influence of different parameters on the printing effect. The results indicate that an increase in jet velocity leads to an increase in stress on cell droplets, thereby increasing the probability of cell damage. Adding cell droplet layers on the substrate can effectively reduce the impact force caused by collisions. Smaller droplets are more susceptible to rupture at higher velocities. These findings provide a scientific basis for optimizing cell printing parameters.
Collapse
Affiliation(s)
- Yankun Wang
- School of Automobile and Transportation Engineering, Guangdong Polytechnic Normal University, Guangzhou 510665, China; (Y.W.); (F.P.); (S.L.); (Y.Z.); (M.W.); (H.Z.)
| | - Fagui Pang
- School of Automobile and Transportation Engineering, Guangdong Polytechnic Normal University, Guangzhou 510665, China; (Y.W.); (F.P.); (S.L.); (Y.Z.); (M.W.); (H.Z.)
| | - Shushan Lai
- School of Automobile and Transportation Engineering, Guangdong Polytechnic Normal University, Guangzhou 510665, China; (Y.W.); (F.P.); (S.L.); (Y.Z.); (M.W.); (H.Z.)
| | - Renye Cai
- School of Automobile and Transportation Engineering, Guangdong Polytechnic Normal University, Guangzhou 510665, China; (Y.W.); (F.P.); (S.L.); (Y.Z.); (M.W.); (H.Z.)
| | - Chenxiang Lai
- Guangzhou Metro Design & Research Institute Co., Ltd., Guangzhou 510010, China
| | - Zexin Yu
- Institute for Manufacturing Technologies of Ceramic Components and Composites (IMTCCC), University of Stuttgart, Allmandring 7b, 70569 Stuttgart, Germany;
| | - Yiwei Zhu
- School of Automobile and Transportation Engineering, Guangdong Polytechnic Normal University, Guangzhou 510665, China; (Y.W.); (F.P.); (S.L.); (Y.Z.); (M.W.); (H.Z.)
| | - Min Wu
- School of Automobile and Transportation Engineering, Guangdong Polytechnic Normal University, Guangzhou 510665, China; (Y.W.); (F.P.); (S.L.); (Y.Z.); (M.W.); (H.Z.)
| | - Heng Zhang
- School of Automobile and Transportation Engineering, Guangdong Polytechnic Normal University, Guangzhou 510665, China; (Y.W.); (F.P.); (S.L.); (Y.Z.); (M.W.); (H.Z.)
| | - Chunyu Kong
- School of Automobile and Transportation Engineering, Guangdong Polytechnic Normal University, Guangzhou 510665, China; (Y.W.); (F.P.); (S.L.); (Y.Z.); (M.W.); (H.Z.)
| |
Collapse
|
15
|
Rayat Pisheh H, Haghdel M, Jahangir M, Hoseinian MS, Rostami Yasuj S, Sarhadi Roodbari A. Effective and new technologies in kidney tissue engineering. Front Bioeng Biotechnol 2024; 12:1476510. [PMID: 39479295 PMCID: PMC11521926 DOI: 10.3389/fbioe.2024.1476510] [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: 08/05/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024] Open
Abstract
Kidney disease encompasses a wide spectrum of conditions, ranging from simple infections to chronic kidney disease. When the kidneys are unable to filter blood and remove waste products, these abnormalities can lead to kidney failure. In severe cases of kidney failure, kidney transplantation is considered the only definitive treatment. Worldwide, the World Health Organization (WHO) repeatedly emphasizes the importance of organ donation and increasing transplantation rates. Many countries implement national programs to promote the culture of organ donation and improve patient access to kidney transplantation. The extent to which this procedure is performed varies across countries and is influenced by several factors, including the volume of organ donation, medical infrastructure, access to technology and health policies. However, a kidney transplant comes with challenges and problems that impact its success. Kidney tissue engineering is a new approach that shows promise for repairing and replacing damaged kidney tissue. This article reviews recent advances in kidney tissue engineering, focusing on engineered structures such as hydrogels, electrospinning, 3D bioprinting, and microfluidic systems. By mimicking the extracellular environment of the kidney, these structures provide suitable conditions for the growth and development of kidney cells. The role of these structures in the formation of blood vessels, the mimicry of kidney functions and the challenges in this field were also discussed. The results of this study show that kidney tissue engineering has high potential for treating kidney diseases and reducing the need for kidney transplantation. However, to achieve clinical application of this technology, further research is required to improve the biocompatibility, vascularization and long-term performance of engineered tissues.
Collapse
Affiliation(s)
- Hossein Rayat Pisheh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mobin Haghdel
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboube Jahangir
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Monireh Sadat Hoseinian
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shaghayegh Rostami Yasuj
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Sarhadi Roodbari
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
16
|
Cui X, Jiao J, Yang L, Wang Y, Jiang W, Yu T, Li M, Zhang H, Chao B, Wang Z, Wu M. Advanced tumor organoid bioprinting strategy for oncology research. Mater Today Bio 2024; 28:101198. [PMID: 39205873 PMCID: PMC11357813 DOI: 10.1016/j.mtbio.2024.101198] [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: 04/10/2024] [Revised: 07/14/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Bioprinting is a groundbreaking technology that enables precise distribution of cell-containing bioinks to construct organoid models that accurately reflect the characteristics of tumors in vivo. By incorporating different types of tumor cells into the bioink, the heterogeneity of tumors can be replicated, enabling studies to simulate real-life situations closely. Precise reproduction of the arrangement and interactions of tumor cells using bioprinting methods provides a more realistic representation of the tumor microenvironment. By mimicking the complexity of the tumor microenvironment, the growth patterns and diffusion of tumors can be demonstrated. This approach can also be used to evaluate the response of tumors to drugs, including drug permeability and cytotoxicity, and other characteristics. Therefore, organoid models can provide a more accurate oncology research and treatment simulation platform. This review summarizes the latest advancements in bioprinting to construct tumor organoid models. First, we describe the bioink used for tumor organoid model construction, followed by an introduction to various bioprinting methods for tumor model formation. Subsequently, we provide an overview of existing bioprinted tumor organoid models.
Collapse
Affiliation(s)
- Xiangran Cui
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Jianhang Jiao
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Lili Yang
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Yang Wang
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Weibo Jiang
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Tong Yu
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Mufeng Li
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Han Zhang
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Bo Chao
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| | - Zhonghan Wang
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
- Orthopaedic Research Institute of Jilin Province, Changchun, 130041, PR China
| | - Minfei Wu
- Department of Orthopedics, The Second Hospital of Jilin University Changchun, 130041, PR China
| |
Collapse
|
17
|
Wang Y, Duan Y, Yang B, Li Y. Nanocomposite Hydrogel Bioinks for 3D Bioprinting of Tumor Models. Biomacromolecules 2024; 25:5288-5299. [PMID: 39083715 DOI: 10.1021/acs.biomac.4c00671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
In vitro tumor models were successfully constructed by 3D bioprinting; however, bioinks with proper viscosity, good biocompatibility, and tunable biophysical and biochemical properties are highly desirable for tumor models that closely recapitulated the main features of native tumors. Here, we developed a nanocomposite hydrogel bioink that was used to construct ovarian and colon cancer models by 3D bioprinting. The nanocomposite bioink was composed of aldehyde-modified cellulose nanocrystals (aCNCs), aldehyde-modified hyaluronic acid (aHA), and gelatin. The hydrogels possessed tunable gelation time, mechanical properties, and printability by controlling the ratio between aCNCs and gelatin. In addition, ovarian and colorectal cancer cells embedded in hydrogels showed high survival rates and rapid growth. By the combination of 3D bioprinting, ovarian and colorectal tumor models were constructed in vitro and used for drug screening. The results showed that gemcitabine had therapeutic effects on ovarian tumor cells. However, the ovarian tumor model showed drug resistance for oxaliplatin treatment.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Yixiong Duan
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130061, China
| | - Yunfeng Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130061, China
| |
Collapse
|
18
|
Chen A, Wang W, Mao Z, He Y, Chen S, Liu G, Su J, Feng P, Shi Y, Yan C, Lu J. Multimaterial 3D and 4D Bioprinting of Heterogenous Constructs for Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307686. [PMID: 37737521 DOI: 10.1002/adma.202307686] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/06/2023] [Indexed: 09/23/2023]
Abstract
Additive manufacturing (AM), which is based on the principle of layer-by-layer shaping and stacking of discrete materials, has shown significant benefits in the fabrication of complicated implants for tissue engineering (TE). However, many native tissues exhibit anisotropic heterogenous constructs with diverse components and functions. Consequently, the replication of complicated biomimetic constructs using conventional AM processes based on a single material is challenging. Multimaterial 3D and 4D bioprinting (with time as the fourth dimension) has emerged as a promising solution for constructing multifunctional implants with heterogenous constructs that can mimic the host microenvironment better than single-material alternatives. Notably, 4D-printed multimaterial implants with biomimetic heterogenous architectures can provide a time-dependent programmable dynamic microenvironment that can promote cell activity and tissue regeneration in response to external stimuli. This paper first presents the typical design strategies of biomimetic heterogenous constructs in TE applications. Subsequently, the latest processes in the multimaterial 3D and 4D bioprinting of heterogenous tissue constructs are discussed, along with their advantages and challenges. In particular, the potential of multimaterial 4D bioprinting of smart multifunctional tissue constructs is highlighted. Furthermore, this review provides insights into how multimaterial 3D and 4D bioprinting can facilitate the realization of next-generation TE applications.
Collapse
Affiliation(s)
- Annan Chen
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Wanying Wang
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
| | - Zhengyi Mao
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Yunhu He
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Shiting Chen
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Guo Liu
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Jin Su
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Pei Feng
- State Key Laboratory of High-Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Yusheng Shi
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Chunze Yan
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Jian Lu
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- Department of Materials Science and Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Hong Kong Branch of National Precious Metals Material Engineering Research, Center (NPMM), City University of Hong Kong, Kowloon, Hong Kong, 999077, China
| |
Collapse
|
19
|
Zhang J, Suttapreyasri S, Leethanakul C, Samruajbenjakun B. Fabrication of vascularized tissue-engineered bone models using triaxial bioprinting. J Biomed Mater Res A 2024; 112:1093-1106. [PMID: 38411369 DOI: 10.1002/jbm.a.37694] [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: 09/15/2023] [Revised: 01/01/2024] [Accepted: 02/14/2024] [Indexed: 02/28/2024]
Abstract
Bone tissue is a highly vascularized tissue. When constructing tissue-engineered bone models, both the osteogenic and angiogenic capabilities of the construct should be carefully considered. However, fabricating a vascularized tissue-engineered bone to promote vascular formation and bone generation, while simultaneously establishing nutrition channels to facilitate nutrient exchange within the constructs, remains a significant challenge. Triaxial bioprinting, which not only allows the independent encapsulation of different cell types while simultaneously forming nutrient channels, could potentially emerge as a strategy for fabricating vascularized tissue-engineered bone. Moreover, bioinks should also be applied in combination to promote both osteogenesis and angiogenesis. In this study, employing triaxial bioprinting, we used a blend bioink of gelatin methacryloyl (GelMA), sodium alginate (Alg), and different concentrations of nano beta-tricalcium phosphate (nano β-TCP) encapsulated MC3T3-E1 preosteoblasts as the outer layer, a mixed bioink of GelMA and Alg loaded with human umbilical vein endothelial cells (HUVEC) as the middle layer, and gelatin as a sacrificial material to form nutrient channels in the inner layer to fabricate vascularized bone constructs simulating the microenvironment for bone and vascular tissues. The results showed that the addition of nano β-TCP could adjust the mechanical, swelling, and degradation properties of the constructs. Biological assessments revealed the cell viability of constructs containing different concentrations of nano β-TCP was higher than 90% on day 7, The cell-laden constructs containing 3% (w/v) nano β-TCP exhibited better osteogenic (higher Alkaline phosphatase activity and larger Osteocalcin positive area) and angiogenic (the gradual increased CD31 positive area) potential. Therefore, using triaxial bioprinting technology and employing GelMA, Alg, and nano β-TCP as bioink components could fabricate vascularized bone tissue constructs, offering a novel strategy for vascularized bone tissue engineering.
Collapse
Affiliation(s)
- Junbiao Zhang
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Songkhla, Thailand
- Guiyang Hospital of Stomatology, Guiyang, People's Republic of China
| | - Srisurang Suttapreyasri
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Thailand
| | - Chidchanok Leethanakul
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Songkhla, Thailand
| | - Bancha Samruajbenjakun
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
20
|
Wang G, Wang Y, Lu G, Dong S, Tang R, Zhao Y, Nie J, Zhu X. Continuous and Controllable Preparation of Sodium Alginate Hydrogel Tubes Guided by the Soft Cap Inspired by the Apical Growth of the Plant. ACS APPLIED MATERIALS & INTERFACES 2024; 16:29600-29609. [PMID: 38832656 DOI: 10.1021/acsami.4c00655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Hydrogel tubes made of sodium alginate (SA) have potential applications in drug delivery, soft robots, biomimetic blood vessels, tissue stents, and other fields. However, the continuous preparation of hollow SA hydrogel tubes with good stability and size control remains a huge challenge for chemists, material scientists, and medical practitioners. Inspired by the plant apical growth strategy, a new method named soft cap-guided growth was proposed to produce SA hydrogel tubes. Due to the introduction of inert low gravity substances, such as air and heptane, into the extrusion needle in front of calcium chloride solution to form a soft cap, the SA hydrogel tubes with controllable sizes were fabricated rapidly and continuously without using a template through a negative gravitropism mechanism. The SA hydrogel tubes had good tensile strength, high burst pressure, and good cell compatibility. In addition, hydrogel tubes with complex patterns were conveniently created by controlling the motion path of a soft cap, such as a rotating SA bath or magnetic force. Our research provided a simple innovative technique to steer the growth of hydrogel tubes, which made it possible to mass produce hydrogel tubes with controllable sizes and programmable patterns.
Collapse
Affiliation(s)
- Guohua Wang
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Yicheng Wang
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Guoqiang Lu
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Shiyu Dong
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Ruifen Tang
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Yingying Zhao
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Jun Nie
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Xiaoqun Zhu
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| |
Collapse
|
21
|
Ortega-Yago A, Ferràs-Tarragó J, de la Calva-Ceinos C, Baeza-Oliete J, Angulo-Sánchez MA, Baixauli-García I, Arguelles-Linares F, Amaya-Valero JV, Baixauli-García F, Medina-Bessó P. [Translated article] Mechanical resistance of polylactic acid bone matrices developed by 3D printing for the reconstruction of bone defects. Rev Esp Cir Ortop Traumatol (Engl Ed) 2024; 68:T262-T270. [PMID: 38253238 DOI: 10.1016/j.recot.2024.01.016] [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: 12/15/2022] [Accepted: 02/01/2023] [Indexed: 01/24/2024] Open
Abstract
INTRODUCTION Bone defects are one of the main limitations in orthopaedic surgery and traumatology. For this reason, multiple bone replacement systems have been developed, either by prosthetic implant or by substitution with osteoforming substances, whose limitations are their survival and lack of structurality, respectively. The objective of this work is the generation of a new material for the creation of biologically active structures that have sufficient tensile strength to maintain the structure during remodelling. MATERIAL AND METHODS A new filament based on the fusion of natural polylactide acid (PLA) powder was designed for the generation of pieces by means of fused deposition modelling (FDM) on which to carry out tensile mechanical tests of osteosynthesis material. A total of 13 groups with different cortical thickness, filling and layer height were carried out, with 10 tensile tests in each group, defining the tensile breaking limit for each group. The regression lines for each group and their mechanical resistance to traction on the filament used were determined. RESULTS The filament ratio per contact surface unit with the osteosynthesis used was the main determinant of the mechanical resistance to traction, either at the expense of the increase in cortical thickness or by the increase in the percentage of cancellous bone filling. Layer height had a minor effect on tensile strength. The regression value was high for cortical thickness and cancellous filling, being elements with a predictable biomechanical behaviour. CONCLUSIONS The new methodology allows the creation of personalised neutral and implantable PLA bone matrices for the reconstruction of large bone defects by means of 3D printing by FDM with a mechanical resistance to traction greater than that of current biological support structures.
Collapse
Affiliation(s)
- A Ortega-Yago
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - J Ferràs-Tarragó
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Departamento de Fisiología, Universidad de Valencia, Valencia, Spain.
| | - C de la Calva-Ceinos
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - J Baeza-Oliete
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - M A Angulo-Sánchez
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - I Baixauli-García
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - F Arguelles-Linares
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - J V Amaya-Valero
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - F Baixauli-García
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - P Medina-Bessó
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
22
|
Quinteira R, Gimondi S, Monteiro NO, Sobreiro-Almeida R, Lasagni L, Romagnani P, Neves NM. Decellularized kidney extracellular matrix-based hydrogels for renal tissue engineering. Acta Biomater 2024; 180:295-307. [PMID: 38642787 DOI: 10.1016/j.actbio.2024.04.026] [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: 09/06/2023] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
Kidney regeneration is hindered by the limited pool of intrinsic reparative cells. Advanced therapies targeting renal regeneration have the potential to alleviate the clinical and financial burdens associated with kidney disease. Delivery systems for cells, extracellular vesicles, or growth factors aimed at enhancing regeneration can benefit from vehicles enabling targeted delivery and controlled release. Hydrogels, optimized to carry biological cargo while promoting regeneration, have emerged as promising candidates for this purpose. This study aims to develop a hydrogel from decellularized kidney extracellular matrix (DKECM) and explore its biocompatibility as a biomaterial for renal regeneration. The resulting hydrogel crosslinks with temperature and exhibits a high concentration of extracellular matrix. The decellularization process efficiently removes detergent residues, yielding a pathogen-free biomaterial that is non-hemolytic and devoid of α-gal epitope. Upon interaction with macrophages, the hydrogel induces differentiation into both pro-inflammatory and anti-inflammatory phenotypes, suggesting an adequate balance to promote biomaterial functionality in vivo. Renal progenitor cells encapsulated in the DKECM hydrogel demonstrate higher viability and proliferation than in commercial collagen-I hydrogels, while also expressing tubular cells and podocyte markers in long-term culture. Overall, the injectable biomaterial derived from porcine DKECM is anticipated to elicit minimal host reaction while fostering progenitor cell bioactivity, offering a potential avenue for enhancing renal regeneration in clinical settings. STATEMENT OF SIGNIFICANCE: The quest to improve treatments for kidney disease is crucial, given the challenges faced by patients on dialysis or waiting for transplants. Exciting new therapies combining biomaterials with cells can revolutionize kidney repair. In this study, researchers created a hydrogel from pig kidney. This gel could be used to deliver cells and other substances that help in kidney regeneration. Despite coming from pigs, it's safe for use in humans, with no harmful substances and reduced risk of immune reactions. Importantly, it promotes a balanced healing response in the body. This research not only advances our knowledge of kidney repair but also offers hope for more effective treatments for kidney diseases.
Collapse
Affiliation(s)
- Rita Quinteira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Sara Gimondi
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Nelson O Monteiro
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rita Sobreiro-Almeida
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Laura Lasagni
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Paola Romagnani
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134 Florence, Italy; Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Nuno M Neves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
23
|
Ortega-Yago A, Ferràs-Tarragó J, de la Calva-Ceinos C, Baeza-Oliete J, Angulo-Sánchez MA, Baixauli-García I, Arguelles-Linares F, Amaya-Valero JV, Baixauli-García F, Medina-Bessó P. Mechanical resistance of polylactic acid bone matrices developed by 3D printing for the reconstruction of bone defects. Rev Esp Cir Ortop Traumatol (Engl Ed) 2024; 68:262-270. [PMID: 36754255 DOI: 10.1016/j.recot.2023.02.001] [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: 12/15/2022] [Revised: 01/25/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
INTRODUCTION Bone defects are one of the main limitations in orthopedic surgery and traumatology. For this reason, multiple bone replacement systems have been developed, either by prosthetic implant or by substitution with osteoforming substances, whose limitations are their survival and lack of structurality, respectively. The objective of this work is the generation of a new material for the creation of biologically active structures that have sufficient tensile strength to maintain the structure during remodeling. MATERIAL AND METHODS A new filament based on the fusion of natural polylactide acid (PLA) powder was designed for the generation of pieces by means of fused deposition modeling (FDM) on which to carry out tensile mechanical tests of osteosynthesis material. A total of 13 groups with different cortical thickness, filling and layer height were carried out, with 10 tensile tests in each group, defining the tensile breaking limit for each group. The regression lines for each group and their mechanical resistance to traction on the filament used were determined. RESULTS The filament ratio per contact surface unit with the osteosynthesis used was the main determinant of the mechanical resistance to traction, either at the expense of the increase in cortical thickness or by the increase in the percentage of cancellous bone filling. Layer height had a minor effect on tensile strength. The regression value was high for cortical thickness and cancellous filling, being elements with a predictable biomechanical behavior. CONCLUSIONS The new methodology allows the creation of personalized neutral and implantable PLA bone matrices for the reconstruction of large bone defects by means of 3D printing by FDM with a mechanical resistance to traction greater than that of current biological support structures.
Collapse
Affiliation(s)
- A Ortega-Yago
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, España; Departamento de Fisiología, Universidad de Valencia, Valencia, España
| | - J Ferràs-Tarragó
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, España; Departamento de Fisiología, Universidad de Valencia, Valencia, España.
| | - C de la Calva-Ceinos
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, España; Departamento de Fisiología, Universidad de Valencia, Valencia, España
| | - J Baeza-Oliete
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, España; Departamento de Fisiología, Universidad de Valencia, Valencia, España
| | - M A Angulo-Sánchez
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, España; Departamento de Fisiología, Universidad de Valencia, Valencia, España
| | - I Baixauli-García
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, España; Departamento de Fisiología, Universidad de Valencia, Valencia, España
| | - F Arguelles-Linares
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, España; Departamento de Fisiología, Universidad de Valencia, Valencia, España
| | - J V Amaya-Valero
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, España; Departamento de Fisiología, Universidad de Valencia, Valencia, España
| | - F Baixauli-García
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, España; Departamento de Fisiología, Universidad de Valencia, Valencia, España
| | - P Medina-Bessó
- Departamento de Cirugía Ortopédica y Traumatología, Hospital Universitario y Politécnico La Fe, Valencia, España; Departamento de Fisiología, Universidad de Valencia, Valencia, España
| |
Collapse
|
24
|
Huang W, Chen YY, He FF, Zhang C. Revolutionizing nephrology research: expanding horizons with kidney-on-a-chip and beyond. Front Bioeng Biotechnol 2024; 12:1373386. [PMID: 38605984 PMCID: PMC11007038 DOI: 10.3389/fbioe.2024.1373386] [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: 01/19/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Organs-on-a-chip (OoC) is a microengineered three-dimensional cell culture system developed for decades. Utilizing microfluidic technology, OoC cultivates cells on perfusable channels to construct in vitro organ models, enabling the simulation of organ-level functions under physiological and pathophysiological conditions. The superior simulation capabilities compared to traditional animal experiments and two-dimensional cell cultures, making OoC a valuable tool for in vitro research. Recently, the application of OoC has extended to the field of nephrology, where it replicates various functional units, including glomerulus-on-a-chip, proximal tubule-on-a-chip, distal tubule-on-a-chip, collecting duct-on-a-chip, and even the entire nephron-on-a-chip to precisely emulate the structure and function of nephrons. Moreover, researchers have integrated kidney models into multi-organ systems, establishing human body-on-a-chip platforms. In this review, the diverse functional kidney units-on-a-chip and their versatile applications are outlined, such as drug nephrotoxicity screening, renal development studies, and investigations into the pathophysiological mechanisms of kidney diseases. The inherent advantages and current limitations of these OoC models are also examined. Finally, the synergy of kidney-on-a-chip with other emerging biomedical technologies are explored, such as bioengineered kidney and bioprinting, and a new insight for chip-based renal replacement therapy in the future are prospected.
Collapse
Affiliation(s)
| | | | | | - Chun Zhang
- *Correspondence: Fang-Fang He, ; Chun Zhang,
| |
Collapse
|
25
|
Górnicki T, Lambrinow J, Golkar-Narenji A, Data K, Domagała D, Niebora J, Farzaneh M, Mozdziak P, Zabel M, Antosik P, Bukowska D, Ratajczak K, Podhorska-Okołów M, Dzięgiel P, Kempisty B. Biomimetic Scaffolds-A Novel Approach to Three Dimensional Cell Culture Techniques for Potential Implementation in Tissue Engineering. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:531. [PMID: 38535679 PMCID: PMC10974775 DOI: 10.3390/nano14060531] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/28/2024] [Accepted: 03/14/2024] [Indexed: 01/06/2025]
Abstract
Biomimetic scaffolds imitate native tissue and can take a multidimensional form. They are biocompatible and can influence cellular metabolism, making them attractive bioengineering platforms. The use of biomimetic scaffolds adds complexity to traditional cell cultivation methods. The most commonly used technique involves cultivating cells on a flat surface in a two-dimensional format due to its simplicity. A three-dimensional (3D) format can provide a microenvironment for surrounding cells. There are two main techniques for obtaining 3D structures based on the presence of scaffolding. Scaffold-free techniques consist of spheroid technologies. Meanwhile, scaffold techniques contain organoids and all constructs that use various types of scaffolds, ranging from decellularized extracellular matrix (dECM) through hydrogels that are one of the most extensively studied forms of potential scaffolds for 3D culture up to 4D bioprinted biomaterials. 3D bioprinting is one of the most important techniques used to create biomimetic scaffolds. The versatility of this technique allows the use of many different types of inks, mainly hydrogels, as well as cells and inorganic substances. Increasing amounts of data provide evidence of vast potential of biomimetic scaffolds usage in tissue engineering and personalized medicine, with the main area of potential application being the regeneration of skin and musculoskeletal systems. Recent papers also indicate increasing amounts of in vivo tests of products based on biomimetic scaffolds, which further strengthen the importance of this branch of tissue engineering and emphasize the need for extensive research to provide safe for humansbiomimetic tissues and organs. In this review article, we provide a review of the recent advancements in the field of biomimetic scaffolds preceded by an overview of cell culture technologies that led to the development of biomimetic scaffold techniques as the most complex type of cell culture.
Collapse
Affiliation(s)
- Tomasz Górnicki
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (J.L.); (M.Z.); (P.D.)
| | - Jakub Lambrinow
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (J.L.); (M.Z.); (P.D.)
| | - Afsaneh Golkar-Narenji
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27607, USA; (P.M.)
| | - Krzysztof Data
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (K.D.); (D.D.); (J.N.)
| | - Dominika Domagała
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (K.D.); (D.D.); (J.N.)
| | - Julia Niebora
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (K.D.); (D.D.); (J.N.)
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 6193673111, Iran;
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27607, USA; (P.M.)
| | - Maciej Zabel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (J.L.); (M.Z.); (P.D.)
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (P.A.); (K.R.)
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland;
| | - Kornel Ratajczak
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (P.A.); (K.R.)
| | - Marzenna Podhorska-Okołów
- Division of Ultrastructure Research, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (J.L.); (M.Z.); (P.D.)
| | - Bartosz Kempisty
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (K.D.); (D.D.); (J.N.)
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (P.A.); (K.R.)
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27613, USA
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 602 00 Brno, Czech Republic
| |
Collapse
|
26
|
Guimaraes APP, Calori IR, Stilhano RS, Tedesco AC. Renal proximal tubule-on-a-chip in PDMS: fabrication, functionalization, and RPTEC:HUVEC co-culture evaluation. Biofabrication 2024; 16:025024. [PMID: 38408383 DOI: 10.1088/1758-5090/ad2d2f] [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: 11/14/2023] [Accepted: 02/26/2024] [Indexed: 02/28/2024]
Abstract
'On-a-chip' technology advances the development of physiologically relevant organ-mimicking architecture by integrating human cells into three-dimensional microfluidic devices. This method also establishes discrete functional units, faciliting focused research on specific organ components. In this study, we detail the development and assessment of a convoluted renal proximal tubule-on-a-chip (PT-on-a-chip). This platform involves co-culturing Renal Proximal Tubule Epithelial Cells (RPTEC) and Human Umbilical Vein Endothelial Cells (HUVEC) within a polydimethylsiloxane microfluidic device, crafted through a combination of 3D printing and molding techniques. Our PT-on-a-chip significantly reduced high glucose level, exhibited albumin uptake, and simulated tubulopathy induced by amphotericin B. Remarkably, the RPTEC:HUVEC co-culture exhibited efficient cell adhesion within 30 min on microchannels functionalized with plasma, 3-aminopropyltriethoxysilane, and type-I collagen. This approach significantly reduced the required incubation time for medium perfusion. In comparison, alternative methods such as plasma and plasma plus polyvinyl alcohol were only effective in promoting cell attachment to flat surfaces. The PT-on-a-chip holds great promise as a valuable tool for assessing the nephrotoxic potential of new drug candidates, enhancing our understanding of drug interactions with co-cultured renal cells, and reducing the need for animal experimentation, promoting the safe and ethical development of new pharmaceuticals.
Collapse
Affiliation(s)
- Ana Paula Pereira Guimaraes
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering- Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Ribeirão Preto 14040-901, Brazil
| | - Italo Rodrigo Calori
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering- Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Ribeirão Preto 14040-901, Brazil
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Labs, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, Oxford, MS 38677, United States of America
| | - Roberta Sessa Stilhano
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, Brazil
| | - Antonio Claudio Tedesco
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering- Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Ribeirão Preto 14040-901, Brazil
| |
Collapse
|
27
|
Jin Y, Sun Q, Ma R, Li R, Qiao R, Li J, Wang L, Hu Y. The trend of allogeneic tendon decellularization: literature review. Cell Tissue Bank 2024; 25:357-367. [PMID: 37355504 DOI: 10.1007/s10561-023-10097-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 06/14/2023] [Indexed: 06/26/2023]
Abstract
Tendon injuries repair is a significant burden for orthopaedic surgeons. Finding a proper graft material to repair tendon is one of the main challenges in orthopaedics, for which the requirement of substitute for tendon repair would be different for each clinical application. Among biological scaffolds, the use of decellularized tendon increasingly represents an interesting approach to treat tendon injuries and several articles have investigated the approaches of tendon decellularization. To understand the outcomes of the the approaches of tendon decellularization on effect of tendon transplantation, a literature review was performed. This review was conducted by searching in Pubmed and Embase and 64 studies were included in this study. The findings revealed that the common approaches to decellularize tendon include chemical, physical, and enzymatic decellularization methods or their combination. With the development of tissue engineering, researchers also put forward new theories such as automatic acellular machine, 3D printing technology to manufacture acellular scaffold.
Collapse
Affiliation(s)
- Yangyang Jin
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Qi Sun
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Rongxing Ma
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Ruifeng Li
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Ruiqi Qiao
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Jikai Li
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Limin Wang
- Beijing Wonderful Medical Biomaterials Co., Ltd., Beijing, China
| | - Yongcheng Hu
- Department of Bone and Soft Tissue Oncology, Tianjin Hospital, 406 Jiefang Southern Road, Tianjin, 300000, China.
| |
Collapse
|
28
|
Patrocinio D, Galván-Chacón V, Gómez-Blanco JC, Miguel SP, Loureiro J, Ribeiro MP, Coutinho P, Pagador JB, Sanchez-Margallo FM. Biopolymers for Tissue Engineering: Crosslinking, Printing Techniques, and Applications. Gels 2023; 9:890. [PMID: 37998980 PMCID: PMC10670821 DOI: 10.3390/gels9110890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Currently, tissue engineering has been dedicated to the development of 3D structures through bioprinting techniques that aim to obtain personalized, dynamic, and complex hydrogel 3D structures. Among the different materials used for the fabrication of such structures, proteins and polysaccharides are the main biological compounds (biopolymers) selected for the bioink formulation. These biomaterials obtained from natural sources are commonly compatible with tissues and cells (biocompatibility), friendly with biological digestion processes (biodegradability), and provide specific macromolecular structural and mechanical properties (biomimicry). However, the rheological behaviors of these natural-based bioinks constitute the main challenge of the cell-laden printing process (bioprinting). For this reason, bioprinting usually requires chemical modifications and/or inter-macromolecular crosslinking. In this sense, a comprehensive analysis describing these biopolymers (natural proteins and polysaccharides)-based bioinks, their modifications, and their stimuli-responsive nature is performed. This manuscript is organized into three sections: (1) tissue engineering application, (2) crosslinking, and (3) bioprinting techniques, analyzing the current challenges and strengths of biopolymers in bioprinting. In conclusion, all hydrogels try to resemble extracellular matrix properties for bioprinted structures while maintaining good printability and stability during the printing process.
Collapse
Affiliation(s)
- David Patrocinio
- CCMIJU, Bioengineering and Health Technologies, Jesus Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain; (D.P.); (V.G.-C.); (J.B.P.)
| | - Victor Galván-Chacón
- CCMIJU, Bioengineering and Health Technologies, Jesus Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain; (D.P.); (V.G.-C.); (J.B.P.)
| | - J. Carlos Gómez-Blanco
- CCMIJU, Bioengineering and Health Technologies, Jesus Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain; (D.P.); (V.G.-C.); (J.B.P.)
| | - Sonia P. Miguel
- CPIRN-IPG, Center of Potential and Innovation of Natural Resources, Polytechnic of Guarda, 6300-559 Guarda, Portugal (M.P.R.)
- CICS-UBI, Health Science Research Center, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Jorge Loureiro
- CPIRN-IPG, Center of Potential and Innovation of Natural Resources, Polytechnic of Guarda, 6300-559 Guarda, Portugal (M.P.R.)
| | - Maximiano P. Ribeiro
- CPIRN-IPG, Center of Potential and Innovation of Natural Resources, Polytechnic of Guarda, 6300-559 Guarda, Portugal (M.P.R.)
- CICS-UBI, Health Science Research Center, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Paula Coutinho
- CPIRN-IPG, Center of Potential and Innovation of Natural Resources, Polytechnic of Guarda, 6300-559 Guarda, Portugal (M.P.R.)
- CICS-UBI, Health Science Research Center, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - J. Blas Pagador
- CCMIJU, Bioengineering and Health Technologies, Jesus Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain; (D.P.); (V.G.-C.); (J.B.P.)
- CIBER CV, Centro de Investigación Biomédica en Red—Enfermedades Cardiovasculares, 28029 Madrid, Spain;
| | - Francisco M. Sanchez-Margallo
- CIBER CV, Centro de Investigación Biomédica en Red—Enfermedades Cardiovasculares, 28029 Madrid, Spain;
- Scientific Direction, Jesus Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain
- TERAV/ISCIII, Red Española de Terapias Avanzadas, Instituto de Salud Carlos III (RICORS, RD21/0017/0029), 28029 Madrid, Spain
| |
Collapse
|
29
|
Xu H, Liu Z, Wei Y, Hu Y, Zhao L, Wang L, Liang Z, Lian X, Chen W, Wang J, Yu Z, Ma X, Huang D. Complexation-Induced Resolution Enhancement Pleiotropic Small Diameter Vascular Constructs with Superior Antibacterial and Angiogenesis Properties. Adv Healthc Mater 2023; 12:e2301809. [PMID: 37571957 DOI: 10.1002/adhm.202301809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/05/2023] [Indexed: 08/13/2023]
Abstract
3D printing has been widely applied for preparing artificial blood vessels, which will bring innovation to cardiovascular disorder intervention. However, the printing resolution and anti-infection properties of small-diameter vessels (Φ < 6 mm) have been challenging in 3D printing. The primary objective of this research is to design a novel coaxial 3D-printing postprocessing method for preparing small-size blood vessels with improved antibacterial and angiogenesis properties. The coaxial printing resolution can be more conveniently improved. Negatively charged polyvinyl alcohol (PVA) and alginate (Alg) interpenetrating networks artificial vessels are immersed in positively charged chitosan (CTS) solution. Rapid dimensional shrinkage takes place on its outer surface through electrostatic interactions. The maximum shrinkage size of wall thickness can reach 61.2%. The vessels demonstrate strong antibacterial properties against Escherichia coli (98.8 ± 0.5%) and Staphylococcus aureus (97.6 ± 1.4%). In rat dorsal skin grafting experiments, Cu2+ can promote angiogenesis by regulating hypoxia-inducible factor-1 pathway. No artificial blood vessel blockage occurs after 5 days of blood circulation in vitro.
Collapse
Affiliation(s)
- Huilun Xu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
| | - Zhengjiang Liu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
| | - Yan Wei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Research Center for Biomaterials, Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Yinchun Hu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Research Center for Biomaterials, Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Liqin Zhao
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Research Center for Biomaterials, Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Longfei Wang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Research Center for Biomaterials, Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Ziwei Liang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Research Center for Biomaterials, Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Xiaojie Lian
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Research Center for Biomaterials, Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Weiyi Chen
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Research Center for Biomaterials, Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Jiucun Wang
- Human Phenome Institute, Fudan University, Shanghai, 200433, P. R. China
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Science, Fudan University, Shanghai, 200433, P. R. China
| | - Zhaoyan Yu
- Shandong Public Health Clinical Center, Shandong University, Jinan, 250000, P. R. China
| | - Xudong Ma
- Cytori Therapeutics LLC, Shanghai, 201802, P. R. China
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Research Center for Biomaterials, Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| |
Collapse
|
30
|
Christou CD, Vasileiadou S, Sotiroudis G, Tsoulfas G. Three-Dimensional Printing and Bioprinting in Renal Transplantation and Regenerative Medicine: Current Perspectives. J Clin Med 2023; 12:6520. [PMID: 37892658 PMCID: PMC10607284 DOI: 10.3390/jcm12206520] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/29/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
For patients with end-stage kidney disease (ESKD), renal transplantation is the treatment of choice, constituting the most common solid organ transplantation. This study aims to provide a comprehensive review regarding the application of three-dimensional (3D) printing and bioprinting in renal transplantation and regenerative medicine. Specifically, we present studies where 3D-printed models were used in the training of surgeons through renal transplantation simulations, in patient education where patients acquire a higher understanding of their disease and the proposed operation, in the preoperative planning to facilitate decision-making, and in fabricating customized, tools and devices. Three-dimensional-printed models could transform how surgeons train by providing surgical rehearsal platforms across all surgical specialties, enabling training with tissue realism and anatomic precision. The use of 3D-printed models in renal transplantations has shown a positive impact on surgical outcomes, including the duration of the operation and the intraoperative blood loss. Regarding 3D bioprinting, the technique has shown promising results, especially in the field of microfluidic devices, with the development of tissue demonstrating proximal tubules, glomerulus, and tubuloinerstitium function, and in renal organoid development. Such models can be applied for renal disease modeling, drug development, and renal regenerative medicine.
Collapse
Affiliation(s)
- Chrysanthos D. Christou
- Department of Transplantation Surgery, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (S.V.); (G.S.); (G.T.)
| | | | | | | |
Collapse
|
31
|
Frankowski J, Kurzątkowska M, Sobczak M, Piotrowska U. Utilization of 3D bioprinting technology in creating human tissue and organoid models for preclinical drug research - State-of-the-art. Int J Pharm 2023; 644:123313. [PMID: 37579828 DOI: 10.1016/j.ijpharm.2023.123313] [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: 05/25/2023] [Revised: 07/28/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
Rapid development of tissue engineering in recent years has increased the importance of three-dimensional (3D) bioprinting technology as novel strategy for fabrication functional 3D tissue and organoid models for pharmaceutical research. 3D bioprinting technology gives hope for eliminating many problems associated with traditional cell culture methods during drug screening. However, there is a still long way to wider clinical application of this technology due to the numerous difficulties associated with development of bioinks, advanced printers and in-depth understanding of human tissue architecture. In this review, the work associated with relatively well-known extrusion-based bioprinting (EBB), jetting-based bioprinting (JBB), and vat photopolymerization bioprinting (VPB) is presented and discussed with the latest advances and limitations in this field. Next we discuss state-of-the-art research of 3D bioprinted in vitro models including liver, kidney, lung, heart, intestines, eye, skin as well as neural and bone tissue that have potential applications in the development of new drugs.
Collapse
Affiliation(s)
- Joachim Frankowski
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Matylda Kurzątkowska
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Marcin Sobczak
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Urszula Piotrowska
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland.
| |
Collapse
|
32
|
Singh NK, Kim JY, Jang J, Kim YK, Cho DW. 3D Cell Printing of Advanced Vascularized Proximal Tubule-on-a-Chip for Drug Induced Nephrotoxicity Advancement. ACS APPLIED BIO MATERIALS 2023; 6:3750-3758. [PMID: 37606916 DOI: 10.1021/acsabm.3c00421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Renal dysfunction due to drug-induced nephrotoxicity (DIN) affects >20% of the adult population worldwide. The vascularized proximal tubule is a complex structure that is often the primary site of drug-induced kidney injury. Herein, a vascularized proximal tubule-on-a-chip (Vas-POAC) was fabricated, demonstrating improved physiological emulation over earlier single-cell proximal tubule models. A perfusable model of vascularized proximal tubules permits the growth and proliferation of renal proximal tubule cells and adjacent endothelial cells under various conditions. An in vitro Vas-POAC showed mature expressions of the tubule and endothelial cell markers in the mature epithelium and endothelium lumens after 7 days of culture. Expression in the mature proximal tubule epithelium resembled the polarized expression of sodium-glucose cotransporter-2 and the de novo synthesis of ECM proteins. These perfusable Vas-POACs display significantly improved functional properties relative to the proximal tubules-on-a-chip (POAC), which lacks vascular components. Furthermore, the developed Vas-POAC model evaluated the cisplatin-induced nephrotoxicity and revealed enhanced drug receptivity compared to POAC. We further evaluated the capability of the developed proximal tubule model to act as a functional platform that targets screening drug doses that can cause renal proximal tubule injury in adults. Thus, our cell-printed models may prove valuable for screening, thoughtful mechanistic investigations of DIN, and discovery of drugs that interfere with tubule formation.
Collapse
Affiliation(s)
- Narendra K Singh
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- Division of Biomaterials and Biomechanics, School of Dentistry, Oregon Health and Science University (OHSU), Portland, Oregon 97201, United States
- Cancer Early Detection Advanced Research Center (CEDAR), OHSU-Knight Cancer Institute, Portland, Oregon 97201, United States
| | - Jae Yun Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Republic of Korea
| | - Yong Kyun Kim
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, St. Vincent's Hospital, Suwon 16247, Republic of Korea
- POSTECH-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
33
|
Ibi Y, Nishinakamura R. Kidney Bioengineering for Transplantation. Transplantation 2023; 107:1883-1894. [PMID: 36717963 DOI: 10.1097/tp.0000000000004526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The kidney is an important organ for maintenance of homeostasis in the human body. As renal failure progresses, renal replacement therapy becomes necessary. However, there is a chronic shortage of kidney donors, creating a major problem for transplantation. To solve this problem, many strategies for the generation of transplantable kidneys are under investigation. Since the first reports describing that nephron progenitors could be induced from human induced pluripotent stem cells, kidney organoids have been attracting attention as tools for studying human kidney development and diseases. Because the kidney is formed through the interactions of multiple renal progenitors, current studies are investigating ways to combine these progenitors derived from human induced pluripotent stem cells for the generation of transplantable kidney organoids. Other bioengineering strategies, such as decellularization and recellularization of scaffolds, 3-dimensional bioprinting, interspecies blastocyst complementation and progenitor replacement, and xenotransplantation, also have the potential to generate whole kidneys, although each of these strategies has its own challenges. Combinations of these approaches will lead to the generation of bioengineered kidneys that are transplantable into humans.
Collapse
Affiliation(s)
- Yutaro Ibi
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | | |
Collapse
|
34
|
Syed Mohamed SMD, Welsh GI, Roy I. Renal tissue engineering for regenerative medicine using polymers and hydrogels. Biomater Sci 2023; 11:5706-5726. [PMID: 37401545 DOI: 10.1039/d3bm00255a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
Chronic Kidney Disease (CKD) is a growing worldwide problem, leading to end-stage renal disease (ESRD). Current treatments for ESRD include haemodialysis and kidney transplantation, but both are deemed inadequate since haemodialysis does not address all other kidney functions, and there is a shortage of suitable donor organs for transplantation. Research in kidney tissue engineering has been initiated to take a regenerative medicine approach as a potential treatment alternative, either to develop effective cell therapy for reconstruction or engineer a functioning bioartificial kidney. Currently, renal tissue engineering encompasses various materials, mainly polymers and hydrogels, which have been chosen to recreate the sophisticated kidney architecture. It is essential to address the chemical and mechanical aspects of the materials to ensure they can support cell development to restore functionality and feasibility. This paper reviews the types of polymers and hydrogels that have been used in kidney tissue engineering applications, both natural and synthetic, focusing on the processing and formulation used in creating bioactive substrates and how these biomaterials affect the cell biology of the kidney cells used.
Collapse
Affiliation(s)
| | - Gavin I Welsh
- Renal Bristol, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS1 3NY, UK
| | - Ipsita Roy
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield S37HQ, UK.
| |
Collapse
|
35
|
Zhang ZJ, Ding LY, Zuo XL, Feng H, Xia Q. A new paradigm in transplant immunology: At the crossroad of synthetic biology and biomaterials. MED 2023:S2666-6340(23)00142-3. [PMID: 37244257 DOI: 10.1016/j.medj.2023.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 02/04/2023] [Accepted: 05/02/2023] [Indexed: 05/29/2023]
Abstract
Solid organ transplant (SOT) recipients require meticulously tailored immunosuppressive regimens to minimize graft loss and mortality. Traditional approaches focus on inhibiting effector T cells, while the intricate and dynamic immune responses mediated by other components remain unsolved. Emerging advances in synthetic biology and material science have provided novel treatment modalities with increased diversity and precision to the transplantation community. This review investigates the active interface between these two fields, highlights how living and non-living structures can be engineered and integrated for immunomodulation, and discusses their potential application in addressing the challenges in SOT clinical practice.
Collapse
Affiliation(s)
- Zi-Jie Zhang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; Shanghai Engineering Research Centre of Transplantation and Immunology, Shanghai 200127, China
| | - Lu-Yue Ding
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiao-Lei Zuo
- Shanghai Engineering Research Centre of Transplantation and Immunology, Shanghai 200127, China; School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao Feng
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; Shanghai Engineering Research Centre of Transplantation and Immunology, Shanghai 200127, China; Shanghai Institute of Transplantation, Shanghai 200127, China; Punan Branch (Shanghai Punan Hospital), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; Shanghai Engineering Research Centre of Transplantation and Immunology, Shanghai 200127, China; Shanghai Institute of Transplantation, Shanghai 200127, China.
| |
Collapse
|
36
|
Nguyen HT, Peirsman A, Tirpakova Z, Mandal K, Vanlauwe F, Maity S, Kawakita S, Khorsandi D, Herculano R, Umemura C, Yilgor C, Bell R, Hanson A, Li S, Nanda HS, Zhu Y, Najafabadi AH, Jucaud V, Barros N, Dokmeci MR, Khademhosseini A. Engineered Vasculature for Cancer Research and Regenerative Medicine. MICROMACHINES 2023; 14:978. [PMID: 37241602 PMCID: PMC10221678 DOI: 10.3390/mi14050978] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023]
Abstract
Engineered human tissues created by three-dimensional cell culture of human cells in a hydrogel are becoming emerging model systems for cancer drug discovery and regenerative medicine. Complex functional engineered tissues can also assist in the regeneration, repair, or replacement of human tissues. However, one of the main hurdles for tissue engineering, three-dimensional cell culture, and regenerative medicine is the capability of delivering nutrients and oxygen to cells through the vasculatures. Several studies have investigated different strategies to create a functional vascular system in engineered tissues and organ-on-a-chips. Engineered vasculatures have been used for the studies of angiogenesis, vasculogenesis, as well as drug and cell transports across the endothelium. Moreover, vascular engineering allows the creation of large functional vascular conduits for regenerative medicine purposes. However, there are still many challenges in the creation of vascularized tissue constructs and their biological applications. This review will summarize the latest efforts to create vasculatures and vascularized tissues for cancer research and regenerative medicine.
Collapse
Affiliation(s)
- Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Arne Peirsman
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Plastic, Reconstructive and Aesthetic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| | - Zuzana Tirpakova
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 04181 Kosice, Slovakia
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Florian Vanlauwe
- Plastic, Reconstructive and Aesthetic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Rondinelli Herculano
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Bioengineering & Biomaterials Group, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil
| | - Christian Umemura
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Can Yilgor
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Remy Bell
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Adrian Hanson
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Shaopei Li
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Himansu Sekhar Nanda
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Biomedical Engineering and Technology Laboratory, PDPM—Indian Institute of Information Technology Design Manufacturing, Jabalpur 482005, Madhya Pradesh, India
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | | | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Natan Barros
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| |
Collapse
|
37
|
Mu X, Gerhard-Herman MD, Zhang YS. Building Blood Vessel Chips with Enhanced Physiological Relevance. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201778. [PMID: 37693798 PMCID: PMC10489284 DOI: 10.1002/admt.202201778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Indexed: 09/12/2023]
Abstract
Blood vessel chips are bioengineered microdevices, consisting of biomaterials, human cells, and microstructures, which recapitulate essential vascular structure and physiology and allow a well-controlled microenvironment and spatial-temporal readouts. Blood vessel chips afford promising opportunities to understand molecular and cellular mechanisms underlying a range of vascular diseases. The physiological relevance is key to these blood vessel chips that rely on bioinspired strategies and bioengineering approaches to translate vascular physiology into artificial units. Here, we discuss several critical aspects of vascular physiology, including morphology, material composition, mechanical properties, flow dynamics, and mass transport, which provide essential guidelines and a valuable source of bioinspiration for the rational design of blood vessel chips. We also review state-of-art blood vessel chips that exhibit important physiological features of the vessel and reveal crucial insights into the biological processes and disease pathogenesis, including rare diseases, with notable implications for drug screening and clinical trials. We envision that the advances in biomaterials, biofabrication, and stem cells improve the physiological relevance of blood vessel chips, which, along with the close collaborations between clinicians and bioengineers, enable their widespread utility.
Collapse
Affiliation(s)
- Xuan Mu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Marie Denise Gerhard-Herman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
38
|
Ma H, Zheng L, Yang S, Cheng YY, Liu T, Wu S, Wang H, Zhang J, Song K. Construction and properties detection of 3D micro-structure scaffolds base on decellularized sheep kidney before and after crosslinking. J Biomater Appl 2023; 37:1593-1604. [PMID: 36919373 DOI: 10.1177/08853282231163758] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Decellularized extracellular matrix is one form of natural material in tissue engineering. The process of dECM retains the tissue microstructure, provides good cell adhesion sites, maintains most of biological signals that promotes the survival and differentiation ability of cells. In this study, sheep kidney was decellularized followed by histochemical staining, elemental analysis and scanning electron microscopy characterizations. The dECM scaffold was prepared with different sequences of freeze drying technology, crosslinking and the water absorption, porosity, mechanical strength with subsequent thermogravimetric analysis, Infrared spectroscopy and biocompatibility tests. Our results indicated that these decellularized treatments of sheep kidney can effectively remove DNA and retain uniform pore size distribution. After crosslinking the scaffold's water absorption decreased from 987.56 ± 40.21% to 934.39 ± 39.61%, the porosity decreased from 89.64 ± 3.2% to 85.09 ± 17.63%, and the compression modulus increased from 304.32 ± 25.43 kPa to 459.53 ± 38.92 kPa, with thermal process the percentage of weight loss decreased from 66.57% to 44.731%, in addition, the composition didn't change significantly, crosslinking could also promote the stability. In terms of biocompatibility, the number of viable cells increased significantly with the days. In conclusion, the crosslinked decellularized sheep kidney extracellular matrix scaffold reduced water absorption and porosity slightly, but has a significant increase in mechanical properties, and presented excellent biocompatibility which are beneficial to cell adhesion, growth and differentiation.
Collapse
Affiliation(s)
- Hailin Ma
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, 12399Dalian University of Technology, Dalian, China
| | - Le Zheng
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, 12399Dalian University of Technology, Dalian, China
| | - Shuangjia Yang
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, 12399Dalian University of Technology, Dalian, China
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, 1994University of Technology Sydney, Sydney, NSW, Australia
| | - Tianqing Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, 12399Dalian University of Technology, Dalian, China
| | - Shuo Wu
- Department of Medical Oncology, Liaoning Cancer Hospital & Institute, 12399Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Hongfei Wang
- Department of Orthopedics, 36674Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jingying Zhang
- The Second Clinical Medical College, 12453Guangdong Medical University, Dongguan, China
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, 12399Dalian University of Technology, Dalian, China
| |
Collapse
|
39
|
Ren B, Jiang Z, Murfee WL, Katz AJ, Siemann D, Huang Y. Realizations of vascularized tissues: From in vitro platforms to in vivo grafts. BIOPHYSICS REVIEWS 2023; 4:011308. [PMID: 36938117 PMCID: PMC10015415 DOI: 10.1063/5.0131972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/07/2023] [Indexed: 03/18/2023]
Abstract
Vascularization is essential for realizing thick and functional tissue constructs that can be utilized for in vitro study platforms and in vivo grafts. The vasculature enables the transport of nutrients, oxygen, and wastes and is also indispensable to organ functional units such as the nephron filtration unit, the blood-air barrier, and the blood-brain barrier. This review aims to discuss the latest progress of organ-like vascularized constructs with specific functionalities and realizations even though they are not yet ready to be used as organ substitutes. First, the human vascular system is briefly introduced and related design considerations for engineering vascularized tissues are discussed. Second, up-to-date creation technologies for vascularized tissues are summarized and classified into the engineering and cellular self-assembly approaches. Third, recent applications ranging from in vitro tissue models, including generic vessel models, tumor models, and different human organ models such as heart, kidneys, liver, lungs, and brain, to prevascularized in vivo grafts for implantation and anastomosis are discussed in detail. The specific design considerations for the aforementioned applications are summarized and future perspectives regarding future clinical applications and commercialization are provided.
Collapse
Affiliation(s)
- Bing Ren
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, USA
| | - Zhihua Jiang
- Department of Surgery, University of Florida, Gainesville, Florida 32610, USA
| | - Walter Lee Murfee
- Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, USA
| | - Adam J. Katz
- Department of Plastic and Reconstructive Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Dietmar Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, Florida 32610, USA
| | - Yong Huang
- Author to whom correspondence should be addressed:
| |
Collapse
|
40
|
Zhu Y, Shi Z, Ding W, Li C. On-chip construction of a fully structured scaffold-free vascularized renal tubule. Biomed Microdevices 2023; 25:8. [PMID: 36826720 DOI: 10.1007/s10544-023-00648-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2023] [Indexed: 02/25/2023]
Abstract
Renal tubule chips have emerged as a promising platform for drug nephrotoxicity testing. However, the reported renal tubule chips hardly replicate the unique structure of renal tubules with thick proximal and distal tubules and a thin loop of Henle. In this study, we developed a fully structured scaffold-free vascularized renal tubule on a microfluidic chip. On the chip, the renal epithelial cell-laden hollow calcium-polymerized alginate tube with thick segments at both ends and a thin middle segment was U-shaped embedded in collagen hydrogel, parallel to the endothelial cell-laden hollow calcium-polymerized alginate tube with uniform tube diameter. After the alginate tubes were on-chip degraded, the renal epithelial cells and endothelial cells automatically attached to the collagen hydrogel and proliferated to form the renal tubule with proximal tubule, loop of Henle and distal tubule as well as peritubular blood vessel. We evaluated the viability of cells on the hollow alginate tubes, characterized the distribution and morphology of cells before and after the degradation of the alginate tube, and confirmed the proliferation of cells and the metabolic function of cells in terms of ATP synthesis, fibronectin secretion and VEGFR2 expression on the chip. The enhanced metabolic functions of renal epithelial cells and endothelial cells were preliminarily demonstrated. This study provides new insights into designing a more biomimetic renal tubule on a microfluidic chip.
Collapse
Affiliation(s)
- Yuntian Zhu
- , Hefei No.1 High School, 230041, Hefei, Anhui, China
| | - Zhengdi Shi
- School of Information Science and Technology, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Weiping Ding
- School of Information Science and Technology, University of Science and Technology of China, 230027, Hefei, Anhui, China.
| | - Chengpan Li
- School of Information Science and Technology, University of Science and Technology of China, 230027, Hefei, Anhui, China. .,Center for Biomedical Imaging, University of Science and Technology of China, 230027, Hefei, Anhui, China.
| |
Collapse
|
41
|
Mohandas S, Gayatri V, Kumaran K, Gopinath V, Paulmurugan R, Ramkumar KM. New Frontiers in Three-Dimensional Culture Platforms to Improve Diabetes Research. Pharmaceutics 2023; 15:pharmaceutics15030725. [PMID: 36986591 PMCID: PMC10056755 DOI: 10.3390/pharmaceutics15030725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Diabetes mellitus is associated with defects in islet β-cell functioning and consequent hyperglycemia resulting in multi-organ damage. Physiologically relevant models that mimic human diabetic progression are urgently needed to identify new drug targets. Three-dimensional (3D) cell-culture systems are gaining a considerable interest in diabetic disease modelling and are being utilized as platforms for diabetic drug discovery and pancreatic tissue engineering. Three-dimensional models offer a marked advantage in obtaining physiologically relevant information and improve drug selectivity over conventional 2D (two-dimensional) cultures and rodent models. Indeed, recent evidence persuasively supports the adoption of appropriate 3D cell technology in β-cell cultivation. This review article provides a considerably updated view of the benefits of employing 3D models in the experimental workflow compared to conventional animal and 2D models. We compile the latest innovations in this field and discuss the various strategies used to generate 3D culture models in diabetic research. We also critically review the advantages and the limitations of each 3D technology, with particular attention to the maintenance of β-cell morphology, functionality, and intercellular crosstalk. Furthermore, we emphasize the scope of improvement needed in the 3D culture systems employed in diabetes research and the promises they hold as excellent research platforms in managing diabetes.
Collapse
Affiliation(s)
- Sundhar Mohandas
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Vijaya Gayatri
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Kriya Kumaran
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Vipin Gopinath
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Centre for Cancer Early Detection, Bio-X Program, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Molecular Oncology Division, Malabar Cancer Centre, Moozhikkara P.O, Thalassery 670103, Kerala, India
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Centre for Cancer Early Detection, Bio-X Program, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Correspondence: (R.P.); (K.M.R.)
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Centre for Cancer Early Detection, Bio-X Program, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Correspondence: (R.P.); (K.M.R.)
| |
Collapse
|
42
|
Salti H, Kramer L, Nelz SC, Lorenz M, Breitrück A, Hofrichter J, Frank M, Schulz K, Mitzner S, Wasserkort R. Decellularization of precision-cut kidney slices-application of physical and chemical methods. Biomed Mater 2023; 18:025004. [PMID: 36599165 DOI: 10.1088/1748-605x/acb02e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 01/04/2023] [Indexed: 01/05/2023]
Abstract
The extracellular matrix (ECM) obtained by decellularization provides scaffolds with the natural complex architecture and biochemical composition of the target organ. Whole kidney decellularization by perfusion uses the vasculature to remove cells leaving a scaffold that can be recellularized with patient-specific cells. However, decellularization and recellularization are highly complex processes that require intensive optimization of various parameters. In pursuit of this, a huge number of animals must be sacrificed. Therefore, we used precision-cut kidney slices (PCKS) as a source of natural scaffolds, which were decellularized by immersion in chemical reagents allowing the examination of more parameters with less animals. However, chemical reagents have a damaging effect on the structure and components of the ECM. Therefore, this study aimed at investigating the effects of physical treatment methods on the effectiveness of PCKS decellularization by immersion in chemical reagents (CHEM). PCKS were treated physically before or during immersion in chemicals (CHEM) with high hydrostatic pressure (HHP), freezing-thawing cycles (FTC) or in an ultrasonic bath system (UBS). Biochemical and DNA quantification as well as structural evaluation with conventional histology and scanning electron microscopy (SEM) were performed. Compared to decellularization by CHEM alone, FTC treatment prior to CHEM was the most effective in reducing DNA while also preserving glycosaminoglycan (GAG) content. Moreover, while UBS resulted in a comparable reduction of DNA, it was the least effective in retaining GAGs. In contrast, despite the pretreatment with HHP with pressures up to 200 MPa, it was the least effective in DNA removal. Histological scoring showed that HHP scaffolds received the best score followed by UBS, FTC and CHEM scaffolds. However further analysis with SEM demonstrated a higher deterioration of the ultrastructure in UBS scaffolds. Altogether, pretreatment with FTC prior to CHEM resulted in a better balance between DNA removal and structural preservation.
Collapse
Affiliation(s)
- Haitham Salti
- Department of Extracorporeal Therapy Systems (EXTHER), Fraunhofer Institute for Cell Therapy and Immunology (IZI), Rostock, Germany
| | - Lea Kramer
- Department of Extracorporeal Therapy Systems (EXTHER), Fraunhofer Institute for Cell Therapy and Immunology (IZI), Rostock, Germany
| | - Sophie-Charlotte Nelz
- Department of Extracorporeal Therapy Systems (EXTHER), Fraunhofer Institute for Cell Therapy and Immunology (IZI), Rostock, Germany
- Division of Nephrology, Department of Internal Medicine, Rostock University Medical Center, Rostock, Germany
| | - Mathias Lorenz
- Wismar University of Applied Sciences, Faculty of Engineering, Wismar, Germany
| | - Anne Breitrück
- Department of Extracorporeal Therapy Systems (EXTHER), Fraunhofer Institute for Cell Therapy and Immunology (IZI), Rostock, Germany
- Division of Nephrology, Department of Internal Medicine, Rostock University Medical Center, Rostock, Germany
| | - Jacqueline Hofrichter
- Department of Extracorporeal Therapy Systems (EXTHER), Fraunhofer Institute for Cell Therapy and Immunology (IZI), Rostock, Germany
- Division of Nephrology, Department of Internal Medicine, Rostock University Medical Center, Rostock, Germany
| | - Marcus Frank
- Medical Biology and Electron Microscopy Centre, Rostock University Medical Center, Rostock, Germany
- Department Life Light & Matter, University of Rostock, Rostock, Germany
| | - Karoline Schulz
- Medical Biology and Electron Microscopy Centre, Rostock University Medical Center, Rostock, Germany
| | - Steffen Mitzner
- Department of Extracorporeal Therapy Systems (EXTHER), Fraunhofer Institute for Cell Therapy and Immunology (IZI), Rostock, Germany
- Division of Nephrology, Department of Internal Medicine, Rostock University Medical Center, Rostock, Germany
| | - Reinhold Wasserkort
- Department of Extracorporeal Therapy Systems (EXTHER), Fraunhofer Institute for Cell Therapy and Immunology (IZI), Rostock, Germany
- Division of Nephrology, Department of Internal Medicine, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
43
|
Wang B, Qinglai T, Yang Q, Li M, Zeng S, Yang X, Xiao Z, Tong X, Lei L, Li S. Functional acellular matrix for tissue repair. Mater Today Bio 2023; 18:100530. [PMID: 36601535 PMCID: PMC9806685 DOI: 10.1016/j.mtbio.2022.100530] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022] Open
Abstract
In view of their low immunogenicity, biomimetic internal environment, tissue- and organ-like physicochemical properties, and functionalization potential, decellularized extracellular matrix (dECM) materials attract considerable attention and are widely used in tissue engineering. This review describes the composition of extracellular matrices and their role in stem-cell differentiation, discusses the advantages and disadvantages of existing decellularization techniques, and presents methods for the functionalization and characterization of decellularized scaffolds. In addition, we discuss progress in the use of dECMs for cartilage, skin, nerve, and muscle repair and the transplantation or regeneration of different whole organs (e.g., kidneys, liver, uterus, lungs, and heart), summarize the shortcomings of using dECMs for tissue and organ repair after refunctionalization, and examine the corresponding future prospects. Thus, the present review helps to further systematize the application of functionalized dECMs in tissue/organ transplantation and keep researchers up to date on recent progress in dECM usage.
Collapse
Affiliation(s)
- Bin Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Tang Qinglai
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Mengmeng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Shiying Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Zian Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xinying Tong
- Department of Hemodialysis, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
44
|
Yang X, Ma Y, Wang X, Yuan S, Huo F, Yi G, Zhang J, Yang B, Tian W. A 3D-Bioprinted Functional Module Based on Decellularized Extracellular Matrix Bioink for Periodontal Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205041. [PMID: 36516309 PMCID: PMC9929114 DOI: 10.1002/advs.202205041] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/15/2022] [Indexed: 05/14/2023]
Abstract
Poor fiber orientation and mismatched bone-ligament interface fusion have plagued the regeneration of periodontal defects by cell-based scaffolds. A 3D bioprinted biomimetic periodontal module is designed with high architectural integrity using a methacrylate gelatin/decellularized extracellular matrix (GelMA/dECM) cell-laden bioink. The module presents favorable mechanical properties and orientation guidance by high-precision topographical cues and provides a biochemical environment conducive to regulating encapsulated cell behavior. The dECM features robust immunomodulatory activity, reducing the release of proinflammatory factors by M1 macrophages and decreasing local inflammation in Sprague Dawley rats. In a clinically relevant critical-size periodontal defect model, the bioprinted module significantly enhances the regeneration of hybrid periodontal tissues in beagles, especially the anchoring structures of the bone-ligament interface, well-aligned periodontal fibers, and highly mineralized alveolar bone. This demonstrates the effectiveness and feasibility of 3D bioprinting combined with a dental follicle-specific dECM bioink for periodontium regeneration, providing new avenues for future clinical practice.
Collapse
Affiliation(s)
- Xueting Yang
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationDepartment of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Yue Ma
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Xiuting Wang
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationDepartment of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Shengmeng Yuan
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationDepartment of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Fangjun Huo
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Genzheng Yi
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationDepartment of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Jingyi Zhang
- Chengdu Shiliankangjian Biotechnology Co., Ltd.Chengdu610041P. R. China
| | - Bo Yang
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationDepartment of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Weidong Tian
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationDepartment of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| |
Collapse
|
45
|
Grebenyuk S, Abdel Fattah AR, Kumar M, Toprakhisar B, Rustandi G, Vananroye A, Salmon I, Verfaillie C, Grillo M, Ranga A. Large-scale perfused tissues via synthetic 3D soft microfluidics. Nat Commun 2023; 14:193. [PMID: 36635264 PMCID: PMC9837048 DOI: 10.1038/s41467-022-35619-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/13/2022] [Indexed: 01/14/2023] Open
Abstract
The vascularization of engineered tissues and organoids has remained a major unresolved challenge in regenerative medicine. While multiple approaches have been developed to vascularize in vitro tissues, it has thus far not been possible to generate sufficiently dense networks of small-scale vessels to perfuse large de novo tissues. Here, we achieve the perfusion of multi-mm3 tissue constructs by generating networks of synthetic capillary-scale 3D vessels. Our 3D soft microfluidic strategy is uniquely enabled by a 3D-printable 2-photon-polymerizable hydrogel formulation, which allows for precise microvessel printing at scales below the diffusion limit of living tissues. We demonstrate that these large-scale engineered tissues are viable, proliferative and exhibit complex morphogenesis during long-term in-vitro culture, while avoiding hypoxia and necrosis. We show by scRNAseq and immunohistochemistry that neural differentiation is significantly accelerated in perfused neural constructs. Additionally, we illustrate the versatility of this platform by demonstrating long-term perfusion of developing neural and liver tissue. This fully synthetic vascularization platform opens the door to the generation of human tissue models at unprecedented scale and complexity.
Collapse
Affiliation(s)
- Sergei Grebenyuk
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.
| | - Abdel Rahman Abdel Fattah
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Manoj Kumar
- Stem Cell Institute Leuven and Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Burak Toprakhisar
- Stem Cell Institute Leuven and Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Gregorius Rustandi
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Anja Vananroye
- Laboratory of Soft Matter, Rheology and Technology, Department of Chemical Engineering, KU Leuven, Leuven, Belgium
| | - Idris Salmon
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Catherine Verfaillie
- Stem Cell Institute Leuven and Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Mark Grillo
- Grillo Consulting Inc., San Francisco, CA, USA
| | - Adrian Ranga
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.
- Leuven Brain Institute, KU Leuven, Leuven, Belgium.
- Leuven Institute for Single Cell Omics, KU Leuven, Leuven, Belgium.
| |
Collapse
|
46
|
Hrynevich A, Li Y, Cedillo-Servin G, Malda J, Castilho M. (Bio)fabrication of microfluidic devices and organs-on-a-chip. 3D Print Med 2023. [DOI: 10.1016/b978-0-323-89831-7.00001-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
47
|
Lacueva-Aparicio A, Lindoso RS, Mihăilă SM, Giménez I. Role of extracellular matrix components and structure in new renal models in vitro. Front Physiol 2022; 13:1048738. [PMID: 36569770 PMCID: PMC9767975 DOI: 10.3389/fphys.2022.1048738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/31/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM), a complex set of fibrillar proteins and proteoglycans, supports the renal parenchyma and provides biomechanical and biochemical cues critical for spatial-temporal patterning of cell development and acquisition of specialized functions. As in vitro models progress towards biomimicry, more attention is paid to reproducing ECM-mediated stimuli. ECM's role in in vitro models of renal function and disease used to investigate kidney injury and regeneration is discussed. Availability, affordability, and lot-to-lot consistency are the main factors determining the selection of materials to recreate ECM in vitro. While simpler components can be synthesized in vitro, others must be isolated from animal or human tissues, either as single isolated components or as complex mixtures, such as Matrigel or decellularized formulations. Synthetic polymeric materials with dynamic and instructive capacities are also being explored for cell mechanical support to overcome the issues with natural products. ECM components can be used as simple 2D coatings or complex 3D scaffolds combining natural and synthetic materials. The goal is to recreate the biochemical signals provided by glycosaminoglycans and other signaling molecules, together with the stiffness, elasticity, segmentation, and dimensionality of the original kidney tissue, to support the specialized functions of glomerular, tubular, and vascular compartments. ECM mimicking also plays a central role in recent developments aiming to reproduce renal tissue in vitro or even in therapeutical strategies to regenerate renal function. Bioprinting of renal tubules, recellularization of kidney ECM scaffolds, and development of kidney organoids are examples. Future solutions will probably combine these technologies.
Collapse
Affiliation(s)
- Alodia Lacueva-Aparicio
- Renal and Cardiovascular Physiopathology (FISIOPREN), Aragon’s Health Sciences Institute, Zaragoza, Spain,Tissue Microenvironment Lab (TME Lab), I3A, University of Zaragoza, Zaragoza, Spain
| | - Rafael Soares Lindoso
- Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Silvia M. Mihăilă
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Ignacio Giménez
- Renal and Cardiovascular Physiopathology (FISIOPREN), Aragon’s Health Sciences Institute, Zaragoza, Spain,Institute for Health Research Aragon (IIS Aragon), Zaragoza, Spain,School of Medicine, University of Zaragoza, Zaragoza, Spain,*Correspondence: Ignacio Giménez,
| |
Collapse
|
48
|
Pichler R, Rizzo L, Tröndle K, Bühler M, Brucker H, Müller AL, Grand K, Farè S, Viau A, Kaminski MM, Kuehn EW, Koch F, Zimmermann S, Koltay P, Lienkamp SS. Tuning the 3D microenvironment of reprogrammed tubule cells enhances biomimetic modeling of polycystic kidney disease. Biomaterials 2022; 291:121910. [DOI: 10.1016/j.biomaterials.2022.121910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/04/2022] [Accepted: 11/06/2022] [Indexed: 11/09/2022]
|
49
|
Khanna A, Oropeza BP, Huang NF. Engineering Spatiotemporal Control in Vascularized Tissues. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9100555. [PMID: 36290523 PMCID: PMC9598830 DOI: 10.3390/bioengineering9100555] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
A major challenge in engineering scalable three-dimensional tissues is the generation of a functional and developed microvascular network for adequate perfusion of oxygen and growth factors. Current biological approaches to creating vascularized tissues include the use of vascular cells, soluble factors, and instructive biomaterials. Angiogenesis and the subsequent generation of a functional vascular bed within engineered tissues has gained attention and is actively being studied through combinations of physical and chemical signals, specifically through the presentation of topographical growth factor signals. The spatiotemporal control of angiogenic signals can generate vascular networks in large and dense engineered tissues. This review highlights the developments and studies in the spatiotemporal control of these biological approaches through the coordinated orchestration of angiogenic factors, differentiation of vascular cells, and microfabrication of complex vascular networks. Fabrication strategies to achieve spatiotemporal control of vascularization involves the incorporation or encapsulation of growth factors, topographical engineering approaches, and 3D bioprinting techniques. In this article, we highlight the vascularization of engineered tissues, with a focus on vascularized cardiac patches that are clinically scalable for myocardial repair. Finally, we discuss the present challenges for successful clinical translation of engineered tissues and biomaterials.
Collapse
Affiliation(s)
| | - Beu P. Oropeza
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Center for Tissue Regeneration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Ngan F. Huang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Center for Tissue Regeneration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Correspondence:
| |
Collapse
|
50
|
Xu H, Su Y, Liao Z, Liu Z, Huang X, Zhao L, Duan R, Hu Y, Wei Y, Lian X, Huang D. Coaxial bioprinting vascular constructs: A review. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|