1
|
Kesharwani A, Tani S, Nishikawa M, Sakai Y, Okada H, Ohba S, Chung UI, Hojo H. Modeling vascular dynamics at the initial stage of endochondral ossification on a microfluidic chip using a human embryonic-stem-cell-derived organoid. Regen Ther 2025; 28:90-100. [PMID: 39703814 PMCID: PMC11655692 DOI: 10.1016/j.reth.2024.11.018] [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: 11/13/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/21/2024] Open
Abstract
Vascular interactions play a crucial role in embryogenesis, including skeletal development. During endochondral ossification, vascular networks are formed as mesenchymal cells condense and later invade skeletal elements to form the bone marrow. We and other groups developed a model of endochondral ossification by implanting human embryonic stem cell (hESC)-derived sclerotome into immunodeficient mice. However, in vitro models of endochondral ossification, particularly vascular interaction with mesenchymal cells at its initial stage, are yet to be established. Therefore, we developed a method to model the initial stage of endochondral ossification using a microfluidic chip-based platform, with a particular focus on the vascular interaction. On the chip, we found that the fibrin gel helped align mCherry-expressing human umbilical vein endothelial cells (HUVECs) better than the collagen-I gel, suggesting that the fibrin gel is more suitable for the formation of a vascular-like network. The perfusability of the vascular-like networks was partially confirmed using fluorescein isothiocyanate (FITC)-dextran and fluorescent microbeads. We then mixed hESC-derived sclerotome with enhanced green fluorescent protein (EGFP)-expressing HUVECs and applied this mixture on the chip. We named this mixture of cells SH organoids. The SH organoids showed superior abilities to maintain the vascular-like network, which was formed by the mCherry-expressing HUVECs, compared with the sclerotome spheroids on the chip. The EGFP-expressing HUVECs migrated from the SH organoid, formed a vascular-like networks, and partially interacted with the mCherry-expressing vascular-like networks on the chip. Histological analysis showed that SRY-box transcription factor 9 (SOX9) and type I collagen were expressed mutually exclusively in the condensed mesenchymal cells and perichondrial-like cells, respectively. This study demonstrates that our SH organoid-on-a-chip method reproduces vascular networks that are formed at the initial stage of endochondral ossification. This model may provide insights into human endochondral ossification and has potential applications in bone disease modeling and drug screening.
Collapse
Affiliation(s)
- Abhiraj Kesharwani
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
| | - Shoichiro Tani
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Masaki Nishikawa
- Department of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
| | - Yasuyuki Sakai
- Department of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
| | - Hiroyuki Okada
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Shinsuke Ohba
- Department of Tissue and Developmental Biology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Ung-il Chung
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hironori Hojo
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
2
|
Lee J, Kim Y, Jung HI, Lim J, Kwak BS. Channel-assembling tumor microenvironment on-chip for evaluating anticancer drug efficacy. J Control Release 2025; 377:376-384. [PMID: 39566854 DOI: 10.1016/j.jconrel.2024.11.030] [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: 09/02/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
Organ-on-a-chip is an advanced system for evaluating drug response in diseases. It simulates the in vivo tumor microenvironment, aiding in the understanding of drug mechanisms and tumor responses. It mimics the structure of the tumor microenvironment and the dynamic conditions within the body. As a result, it holds the potential for applications in precision and personalized medicine. However, there are still limitations in sequential development processes and complex structures, resulting in time-consuming molecular interference during system development. In this study, we developed a channel-assembling tumor microenvironment-on-chip (CATOC) system to overcome these limitations. CATOC was easily segmented into blood vessels and a tumor microenvironment-on-chip, which can be independently developed. The tumor microenvironment-on-chip consists of two independent channels for evaluating drug responses in different types of tumor microenvironments. Each fully developed system was physically interconnected to create a CATOC. Interconnected CATOC was used to validate chemical and targeted anticancer drug responses in different subtypes of the breast tumor microenvironment. We also emphasized the significance of on-chip experiments by observing the drug response of tumor spheroids on CATOC and scaffold-free platforms.
Collapse
Affiliation(s)
- Jaehun Lee
- Yonsei University, School of Mechanical Engineering, 50 Yonsei-ro, Seadaemun-gu, Seoul 03722, Republic of Korea; Dongguk University, College of Medicine, 32 Dongguk-ro, Ilsandong-gu, Goyangsi, Gyeonggi-do 10326, Republic of Korea
| | - Youngwon Kim
- Yonsei University, School of Mechanical Engineering, 50 Yonsei-ro, Seadaemun-gu, Seoul 03722, Republic of Korea; Dongguk University, College of Medicine, 32 Dongguk-ro, Ilsandong-gu, Goyangsi, Gyeonggi-do 10326, Republic of Korea
| | - Hyo-Il Jung
- Yonsei University, School of Mechanical Engineering, 50 Yonsei-ro, Seadaemun-gu, Seoul 03722, Republic of Korea; The DABOM Inc., 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jiseok Lim
- MediSphere Inc., Gyeongsan-si, Gyeongsanbuk-do, Republic of Korea; Yeungnam University, School of Mechanical Engineering, 280 Daehak-ro, Gyeongsan-si, Gyeongsangbuk-do 38541, Republic of Korea.
| | - Bong Seop Kwak
- Dongguk University, College of Medicine, 32 Dongguk-ro, Ilsandong-gu, Goyangsi, Gyeonggi-do 10326, Republic of Korea; MediSphere Inc., Gyeongsan-si, Gyeongsanbuk-do, Republic of Korea.
| |
Collapse
|
3
|
Nguyen HT, Kan EL, Humayun M, Gurvich N, Offeddu GS, Wan Z, Coughlin MF, Renteria DC, Loew A, Wilson S, Zhang C, Vu V, Lee SWL, Tan SL, Barbie D, Hsu J, Gillrie MR, Kamm RD. Patient-specific vascularized tumor model: Blocking monocyte recruitment with multispecific antibodies targeting CCR2 and CSF-1R. Biomaterials 2025; 312:122731. [PMID: 39153324 DOI: 10.1016/j.biomaterials.2024.122731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/19/2024]
Abstract
Tumor-associated inflammation drives cancer progression and therapy resistance, often linked to the infiltration of monocyte-derived tumor-associated macrophages (TAMs), which are associated with poor prognosis in various cancers. To advance immunotherapies, testing on immunocompetent pre-clinical models of human tissue is crucial. We have developed an in vitro model of microvascular networks with tumor spheroids or patient tissues to assess monocyte trafficking into tumors and evaluate immunotherapies targeting the human tumor microenvironment. Our findings demonstrate that macrophages in vascularized breast and lung tumor models can enhance monocyte recruitment via CCL7 and CCL2, mediated by CSF-1R. Additionally, a multispecific antibody targeting CSF-1R, CCR2, and neutralizing TGF-β (CSF1R/CCR2/TGF-β Ab) repolarizes TAMs towards an anti-tumoral M1-like phenotype, reduces monocyte chemoattractant protein secretion, and blocks monocyte migration. This antibody also inhibits monocyte recruitment in patient-specific vascularized tumor models. In summary, this vascularized tumor model recapitulates the monocyte recruitment cascade, enabling functional testing of innovative therapeutic antibodies targeting TAMs in the tumor microenvironment.
Collapse
Affiliation(s)
- Huu Tuan Nguyen
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| | - Ellen L Kan
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Mouhita Humayun
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Nadia Gurvich
- Marengo Therapeutics, 840 Memorial Dr, Cambridge, MA, 02139, USA
| | - Giovanni S Offeddu
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Zhengpeng Wan
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Mark F Coughlin
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Diana C Renteria
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Andreas Loew
- Marengo Therapeutics, 840 Memorial Dr, Cambridge, MA, 02139, USA
| | - Susan Wilson
- Marengo Therapeutics, 840 Memorial Dr, Cambridge, MA, 02139, USA
| | - Christie Zhang
- Marengo Therapeutics, 840 Memorial Dr, Cambridge, MA, 02139, USA
| | - Vivian Vu
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Sharon Wei Ling Lee
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Seng-Lai Tan
- Marengo Therapeutics, 840 Memorial Dr, Cambridge, MA, 02139, USA
| | - David Barbie
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jonathan Hsu
- Marengo Therapeutics, 840 Memorial Dr, Cambridge, MA, 02139, USA
| | - Mark Robert Gillrie
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Department of Medicine, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| | - Roger D Kamm
- Department of Mechanical Engineering and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
4
|
Shih YY, Kao CW, Jhong YR, Chen YA, Chen YW. Synergistic effects of fibrin-enriched adipose decellularized extracellular matrix (AdECM) and microfluidic model on vascularization. RSC Adv 2024; 14:34143-34155. [PMID: 39469019 PMCID: PMC11513771 DOI: 10.1039/d4ra05573j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024] Open
Abstract
Vasculature is essential for maintaining the cellular function and balance of organs and tumors. As a key component of the tumor microenvironment (TME), it significantly influences tumor characteristics. Angiogenesis, heavily influenced by the extracellular matrix (ECM), which acts as a structural scaffold and growth factor reservoir, is regulated by various factors. Notably, adipose tissues and adipose-derived stromal cells contribute angiogenic and anti-apoptotic factors that promote angiogenesis. Sustained vasculature is essential for tissue engineering and ex vivo disease modeling. Lack of shear stress from fluid flow leads to vascular instability and regression. Microfluidic models replicate three-dimensional (3D) cultures from original tissues, encapsulate microenvironmental factors, and maintain consistent fluid flow. In our study, we established decellularized adipose ECM (AdECM) derived from bovine sources and engineered a 3D-printed microfluidic device. We observed significant increases in both the length and diameter of vascular networks after coculturing HUVECs and HDFs in a fibrin gel containing 0.5% AdECM. Additionally, gene expression related to ECM remodeling and angiogenesis was significantly enhanced in vasculature cultivated in fibrin gel containing 0.5% AdECM compared to that in fibrin gel alone. The enhanced vasculogenesis was further amplified and sustained by the 3D microfluidic device placed on a rocker during extended cultivation, primarily through the activation of the PI3K and JAK-mediated pathways. Our ex vivo model with vascularized colon tumoroids revealed that integrating AdECM within a microfluidic device correlates with increased tumoroid growth. Therefore, our study underscores the synergistic impact of AdECM and microfluidic device in promoting and sustaining vasculature. This synergy may have significant implications for tissue regeneration and ex vivo disease modeling, facilitating drug testing and efficacy evaluation.
Collapse
Affiliation(s)
- Yu-Yin Shih
- Research & Development Center for x-Dimensional Extracellular Vesicles, China Medical University Hospital Taichung 404332 Taiwan
| | - Chun-Wei Kao
- Research & Development Center for x-Dimensional Extracellular Vesicles, China Medical University Hospital Taichung 404332 Taiwan
| | - Yi-Rong Jhong
- Research & Development Center for x-Dimensional Extracellular Vesicles, China Medical University Hospital Taichung 404332 Taiwan
| | - Yi-An Chen
- Research & Development Center for x-Dimensional Extracellular Vesicles, China Medical University Hospital Taichung 404332 Taiwan
| | - Yi-Wen Chen
- Research & Development Center for x-Dimensional Extracellular Vesicles, China Medical University Hospital Taichung 404332 Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University Taichung 41354 Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University Taichung 406040 Taiwan
| |
Collapse
|
5
|
Lal R, Singh A, Watts S, Chopra K. Experimental models of Parkinson's disease: Challenges and Opportunities. Eur J Pharmacol 2024; 980:176819. [PMID: 39029778 DOI: 10.1016/j.ejphar.2024.176819] [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: 09/07/2023] [Revised: 05/29/2024] [Accepted: 07/17/2024] [Indexed: 07/21/2024]
Abstract
Parkinson's disease (PD) is a widespread neurodegenerative disorder occurs due to the degradation of dopaminergic neurons present in the substantia nigra pars compacta (SNpc). Millions of people are affected by this devastating disorder globally, and the frequency of the condition increases with the increase in the elderly population. A significant amount of progress has been made in acquiring more knowledge about the etiology and the pathogenesis of PD over the past decades. Animal models have been regarded to be a vital tool for the exploration of complex molecular mechanisms involved in PD. Various animals used as models for disease monitoring include vertebrates (zebrafish, rats, mice, guinea pigs, rabbits and monkeys) and invertebrate models (Drosophila, Caenorhabditis elegans). The animal models most relevant for study of PD are neurotoxin induction-based models (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), 6-Hydroxydopamine (6-OHDA) and agricultural pesticides (rotenone, paraquat), pharmacological models (reserpine or haloperidol treated rats), genetic models (α-synuclein, Leucine-rich repeat kinase 2 (LRRK2), DJ-1, PINK-1 and Parkin). Several non-mammalian genetic models such as zebrafish, Drosophila and Caenorhabditis elegance have also gained popularity in recent years due to easy genetic manipulation, presence of genes homologous to human PD, and rapid screening of novel therapeutic molecules. In addition, in vitro models (SH-SY5Y, PC12, Lund human mesencephalic (LUHMES) cells, Human induced pluripotent stem cell (iPSC), Neural organoids, organ-on-chip) are also currently in trend providing edge in investigating molecular mechanisms involved in PD as they are derived from PD patients. In this review, we explain the current situation and merits and demerits of the various animal models.
Collapse
Affiliation(s)
- Roshan Lal
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India.
| | - Aditi Singh
- TR(i)P for Health Laboratory, Centre for Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City, Sector 81, SAS Nagar, Punjab, 140306, India.
| | - Shivam Watts
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India.
| | - Kanwaljit Chopra
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
6
|
Godier C, Baka Z, Lamy L, Gribova V, Marchal P, Lavalle P, Gaffet E, Bezdetnaya L, Alem H. A 3D Bio-Printed-Based Model for Pancreatic Ductal Adenocarcinoma. Diseases 2024; 12:206. [PMID: 39329875 PMCID: PMC11431387 DOI: 10.3390/diseases12090206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/27/2024] [Accepted: 08/31/2024] [Indexed: 09/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a disease with a very poor prognosis, characterized by incidence rates very close to death rates. Despite the efforts of the scientific community, preclinical models that faithfully recreate the PDAC tumor microenvironment remain limited. Currently, the use of 3D bio-printing is an emerging and promising method for the development of cancer tumor models with reproducible heterogeneity and a precisely controlled structure. This study presents the development of a model using the extrusion 3D bio-printing technique. Initially, a model combining pancreatic cancer cells (Panc-1) and cancer-associated fibroblasts (CAFs) encapsulated in a sodium alginate and gelatin-based hydrogel to mimic the metastatic stage of PDAC was developed and comprehensively characterized. Subsequently, efforts were made to vascularize this model. This study demonstrates that the resulting tumors can maintain viability and proliferate, with cells self-organizing into aggregates with a heterogeneous composition. The utilization of 3D bio-printing in creating this tumor model opens avenues for reproducing tumor complexity in the future, offering a versatile platform for improving anti-cancer therapy models.
Collapse
Affiliation(s)
- Claire Godier
- IJL, CNRS, Université de Lorraine, 54000 Nancy, France; (C.G.); (Z.B.); (E.G.)
| | - Zakaria Baka
- IJL, CNRS, Université de Lorraine, 54000 Nancy, France; (C.G.); (Z.B.); (E.G.)
| | - Laureline Lamy
- CRAN, CNRS, Université de Lorraine, 54506 Vandœuvre-lès-Nancy, France; (L.L.); (L.B.)
- Département Recherche, Institut de Cancérologie de Lorraine (ICL), 6 Avenue de Bourgogne, 54519 Vandœuvre-lès-Nancy, France
| | - Varvara Gribova
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1121, Biomaterials and Bioengineering, 1 rue Eugène Boeckel, 67100 Strasbourg, France; (V.G.); (P.L.)
- Faculté de Chirurgie Dentaire, Université de Strasbourg, 8 rue Sainte Elisabeth, 67000 Strasbourg, France
| | | | - Philippe Lavalle
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1121, Biomaterials and Bioengineering, 1 rue Eugène Boeckel, 67100 Strasbourg, France; (V.G.); (P.L.)
- Faculté de Chirurgie Dentaire, Université de Strasbourg, 8 rue Sainte Elisabeth, 67000 Strasbourg, France
| | - Eric Gaffet
- IJL, CNRS, Université de Lorraine, 54000 Nancy, France; (C.G.); (Z.B.); (E.G.)
| | - Lina Bezdetnaya
- CRAN, CNRS, Université de Lorraine, 54506 Vandœuvre-lès-Nancy, France; (L.L.); (L.B.)
- Département Recherche, Institut de Cancérologie de Lorraine (ICL), 6 Avenue de Bourgogne, 54519 Vandœuvre-lès-Nancy, France
| | - Halima Alem
- IJL, CNRS, Université de Lorraine, 54000 Nancy, France; (C.G.); (Z.B.); (E.G.)
- Institut Universitaire de France, 75000 Paris, France
| |
Collapse
|
7
|
Rizzuti M, Melzi V, Brambilla L, Quetti L, Sali L, Ottoboni L, Meneri M, Ratti A, Verde F, Ticozzi N, Comi GP, Corti S, Abati E. Shaping the Neurovascular Unit Exploiting Human Brain Organoids. Mol Neurobiol 2024; 61:6642-6657. [PMID: 38334812 PMCID: PMC11338975 DOI: 10.1007/s12035-024-03998-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
Brain organoids, three-dimensional cell structures derived from pluripotent stem cells, closely mimic key aspects of the human brain in vitro, providing a powerful tool for studying neurodevelopment and disease. The neuroectodermal induction protocol employed for brain organoid generation primarily gives rise to the neural cellular component but lacks the vital vascular system, which is crucial for the brain functions by regulating differentiation, migration, and circuit formation, as well as delivering oxygen and nutrients. Many neurological diseases are caused by dysfunctions of cerebral microcirculation, making vascularization of human brain organoids an important tool for pathogenetic and translational research. Experimentally, the creation of vascularized brain organoids has primarily focused on the fusion of vascular and brain organoids, on organoid transplantation in vivo, and on the use of microfluidic devices to replicate the intricate microenvironment of the human brain in vitro. This review summarizes these efforts and highlights the importance of studying the neurovascular unit in a forward-looking perspective of leveraging their use for understanding and treating neurological disorders.
Collapse
Affiliation(s)
- Mafalda Rizzuti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Melzi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Brambilla
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Quetti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Sali
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Linda Ottoboni
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Megi Meneri
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Federico Verde
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Nicola Ticozzi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Giacomo Pietro Comi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Abati
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
8
|
Fujimoto K, Kameda Y, Nagano Y, Deguchi S, Yamamoto T, Krol RP, Gee P, Matsumura Y, Okamoto T, Nagao M, Takayama K, Yokokawa R. SARS-CoV-2-induced disruption of a vascular bed in a microphysiological system caused by type-I interferon from bronchial organoids. LAB ON A CHIP 2024; 24:3863-3879. [PMID: 38252025 DOI: 10.1039/d3lc00768e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Blood vessels show various COVID-19-related conditions including thrombosis and cytokine propagation. Existing in vitro blood vessel models cannot represent the consequent changes in the vascular structure or determine the initial infection site, making it difficult to evaluate how epithelial and endothelial tissues are damaged. Here, we developed a microphysiological system (MPS) that co-culture the bronchial organoids and the vascular bed to analyze infection site and interactions. In this system, virus-infected organoids caused damage in vascular structure. However, vasculature was not damaged or infected when the virus was directly introduced to vascular bed. The knockout of interferon-related genes and inhibition of the JAK/STAT pathway reduced the vascular damage, indicating the protective effect of interferon response suppression. The results demonstrate selective infection of bronchial epithelial cells and vascular damage by cytokines and also indicate the applicability of MPS to investigate how the infection influences vascular structure and functions.
Collapse
Affiliation(s)
- Kazuya Fujimoto
- Department of Micro Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Yoshikazu Kameda
- Department of Micro Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Yuta Nagano
- Department of Micro Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Sayaka Deguchi
- Center for iPS cell Research and Application (CiRA), Kyoto University, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Takuya Yamamoto
- Center for iPS cell Research and Application (CiRA), Kyoto University, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan.
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Rafal P Krol
- Research and Development Center, CiRA Foundation, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8397, Japan
| | - Peter Gee
- MaxCyte Inc., Gaithersburg, MD 20878, USA
| | - Yasufumi Matsumura
- Department of Clinical Laboratory medicine, Kyoto University Graduate School of Medicine, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Toru Okamoto
- Department of Microbiology, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Miki Nagao
- Department of Clinical Laboratory medicine, Kyoto University Graduate School of Medicine, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kazuo Takayama
- Center for iPS cell Research and Application (CiRA), Kyoto University, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| |
Collapse
|
9
|
Pramotton FM, Spitz S, Kamm RD. Challenges and Future Perspectives in Modeling Neurodegenerative Diseases Using Organ-on-a-Chip Technology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403892. [PMID: 38922799 PMCID: PMC11348103 DOI: 10.1002/advs.202403892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/01/2024] [Indexed: 06/28/2024]
Abstract
Neurodegenerative diseases (NDDs) affect more than 50 million people worldwide, posing a significant global health challenge as well as a high socioeconomic burden. With aging constituting one of the main risk factors for some NDDs such as Alzheimer's disease (AD) and Parkinson's disease (PD), this societal toll is expected to rise considering the predicted increase in the aging population as well as the limited progress in the development of effective therapeutics. To address the high failure rates in clinical trials, legislative changes permitting the use of alternatives to traditional pre-clinical in vivo models are implemented. In this regard, microphysiological systems (MPS) such as organ-on-a-chip (OoC) platforms constitute a promising tool, due to their ability to mimic complex and human-specific tissue niches in vitro. This review summarizes the current progress in modeling NDDs using OoC technology and discusses five critical aspects still insufficiently addressed in OoC models to date. Taking these aspects into consideration in the future MPS will advance the modeling of NDDs in vitro and increase their translational value in the clinical setting.
Collapse
Affiliation(s)
- Francesca Michela Pramotton
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sarah Spitz
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Roger D. Kamm
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
10
|
Orge I, Nogueira Pinto H, Silva M, Bidarra S, Ferreira S, Calejo I, Masereeuw R, Mihăilă S, Barrias C. Vascular units as advanced living materials for bottom-up engineering of perfusable 3D microvascular networks. Bioact Mater 2024; 38:499-511. [PMID: 38798890 PMCID: PMC11126780 DOI: 10.1016/j.bioactmat.2024.05.021] [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: 05/01/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024] Open
Abstract
The timely establishment of functional neo-vasculature is pivotal for successful tissue development and regeneration, remaining a central challenge in tissue engineering. In this study, we present a novel (micro)vascularization strategy that explores the use of specialized "vascular units" (VUs) as building blocks to initiate blood vessel formation and create perfusable, stroma-embedded 3D microvascular networks from the bottom-up. We demonstrate that VUs composed of endothelial progenitor cells and organ-specific fibroblasts exhibit high angiogenic potential when embedded in fibrin hydrogels. This leads to the formation of VUs-derived capillaries, which fuse with adjacent capillaries to form stable microvascular beds within a supportive, extracellular matrix-rich fibroblastic microenvironment. Using a custom-designed biomimetic fibrin-based vessel-on-chip (VoC), we show that VUs-derived capillaries can inosculate with endothelialized microfluidic channels in the VoC and become perfused. Moreover, VUs can establish capillary bridges between channels, extending the microvascular network throughout the entire device. When VUs and intestinal organoids (IOs) are combined within the VoC, the VUs-derived capillaries and the intestinal fibroblasts progressively reach and envelop the IOs. This promotes the formation of a supportive vascularized stroma around multiple IOs in a single device. These findings underscore the remarkable potential of VUs as building blocks for engineering microvascular networks, with versatile applications spanning from regenerative medicine to advanced in vitro models.
Collapse
Affiliation(s)
- I.D. Orge
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - H. Nogueira Pinto
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - M.A. Silva
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - S.J. Bidarra
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - S.A. Ferreira
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - I. Calejo
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - R. Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - S.M. Mihăilă
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - C.C. Barrias
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
11
|
Joy JD, Malacrida B, Laforêts F, Kotantaki P, Maniati E, Manchanda R, Annibaldi A, Hopkins S, Garrobo-Calleja I, Gautrot J, Balkwill FR. Human 3D Ovarian Cancer Models Reveal Malignant Cell-Intrinsic and -Extrinsic Factors That Influence CAR T-cell Activity. Cancer Res 2024; 84:2432-2449. [PMID: 38819641 PMCID: PMC11292204 DOI: 10.1158/0008-5472.can-23-3007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/29/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
In vitro preclinical testing of chimeric antigen receptor (CAR) T cells is mostly carried out in monolayer cell cultures. However, alternative strategies are needed to take into account the complexity and the effects of the tumor microenvironment. Here, we describe the modulation of CAR T-cell activity by malignant cells and fibroblasts in human three-dimensional (3D) in vitro cell models of increasing complexity. In models combining mucin-1 (MUC1) and TnMUC1 CAR T cells with human high-grade serous ovarian cancer cell spheroids, malignant cell-intrinsic resistance to CAR T-cell killing was due to defective death receptor signaling involving TNFα. Adding primary human fibroblasts to spheroids unexpectedly increased the ability of CAR T cells to kill resistant malignant cells as CCL2 produced by fibroblasts activated CCR2/4+ CAR T cells. However, culturing malignant cells and fibroblasts in collagen gels engendered production of a dense extracellular matrix that impeded CAR T-cell activity in a TGFβ-dependent manner. A vascularized microfluidic device was developed that allowed CAR T cells to flow through the vessels and penetrate the gels in a more physiological way, killing malignant cells in a TNFα-dependent manner. Complex 3D human cell models may provide an efficient way of screening multiple cytotoxic human immune cell constructs while also enabling evaluation of mechanisms of resistance involving cell-cell and cell-matrix interactions, thus accelerating preclinical research on cytotoxic immune cell therapies in solid tumors. Significance: Three-dimensional in vitro models of increasing complexity uncover mechanisms of resistance to CAR T cells in solid tumors, which could help accelerate development of improved CAR T-cell constructs.
Collapse
Affiliation(s)
- Joash D. Joy
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| | - Beatrice Malacrida
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| | - Florian Laforêts
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| | - Panoraia Kotantaki
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| | - Eleni Maniati
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| | - Ranjit Manchanda
- Wolfson Institute of Population Health, Cancer Research UK, Barts Centre, Queen Mary University of London, London, United Kingdom.
- Department of Gynaecological Oncology, Royal London Hospital, Barts Health NHS Trust, London, United Kingdom.
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| | | | - Sarah Hopkins
- GlaxoSmithKline Medicines Research Centre, Stevenage, United Kingdom.
| | | | - Julien Gautrot
- School of Engineering and Material Science, Centre for Bioengineering, Queen Mary University of London, London, United Kingdom.
| | - Frances R. Balkwill
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
12
|
Xu C, Alameri A, Leong W, Johnson E, Chen Z, Xu B, Leong KW. Multiscale engineering of brain organoids for disease modeling. Adv Drug Deliv Rev 2024; 210:115344. [PMID: 38810702 PMCID: PMC11265575 DOI: 10.1016/j.addr.2024.115344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/23/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Brain organoids hold great potential for modeling human brain development and pathogenesis. They recapitulate certain aspects of the transcriptional trajectory, cellular diversity, tissue architecture and functions of the developing brain. In this review, we explore the engineering strategies to control the molecular-, cellular- and tissue-level inputs to achieve high-fidelity brain organoids. We review the application of brain organoids in neural disorder modeling and emerging bioengineering methods to improve data collection and feature extraction at multiscale. The integration of multiscale engineering strategies and analytical methods has significant potential to advance insight into neurological disorders and accelerate drug development.
Collapse
Affiliation(s)
- Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Alia Alameri
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Wei Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Emily Johnson
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Zaozao Chen
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Bin Xu
- Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
13
|
Chiang H, Chung CA. Simulation of Soluble and Bound VEGF-stimulated in vitro Capillary-like Network Formation on Deformed Substrate. PLoS Comput Biol 2024; 20:e1012281. [PMID: 39038038 PMCID: PMC11262697 DOI: 10.1371/journal.pcbi.1012281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 06/26/2024] [Indexed: 07/24/2024] Open
Abstract
Capillary plexus cultivation is crucial in tissue engineering and regenerative medicine. Theoretical simulations have been conducted to supplement the expensive experimental works. However, the mechanisms connecting mechanical and chemical stimuli remained undefined, and the functions of the different VEGF forms in the culture environment were still unclear. In this paper, we developed a hybrid model for simulating short-term in vitro capillary incubations. We used the Cellular Potts model to predict individual cell migration, morphology change, and continuum mechanics to quantify biogel deformation and VEGF transport dynamics. By bridging the mechanical regulation and chemical stimulation in the model, the results showed good agreement between the predicted network topology and experiments, in which elongated cells connected, forming the network cords and round cells gathered, creating cobblestone-like aggregates. The results revealed that the capillary-like networks could develop in high integrity only when the mechanical and chemical couplings worked adequately, with the cell morphology and haptotaxis driven by the soluble and bound forms of VEGF, respectively, functioning simultaneously.
Collapse
Affiliation(s)
- Hsun Chiang
- Department of Mechanical Engineering, National Central University, Taoyuan, Taiwan
| | - Chih-Ang Chung
- Department of Mechanical Engineering, National Central University, Taoyuan, Taiwan
| |
Collapse
|
14
|
Bosmans C, Ginés Rodriguez N, Karperien M, Malda J, Moreira Teixeira L, Levato R, Leijten J. Towards single-cell bioprinting: micropatterning tools for organ-on-chip development. Trends Biotechnol 2024; 42:739-759. [PMID: 38310021 DOI: 10.1016/j.tibtech.2023.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 02/05/2024]
Abstract
Organs-on-chips (OoCs) hold promise to engineer progressively more human-relevant in vitro models for pharmaceutical purposes. Recent developments have delivered increasingly sophisticated designs, yet OoCs still lack in reproducing the inner tissue physiology required to fully resemble the native human body. This review emphasizes the need to include microarchitectural and microstructural features, and discusses promising avenues to incorporate well-defined microarchitectures down to the single-cell level. We highlight how their integration will significantly contribute to the advancement of the field towards highly organized structural and hierarchical tissues-on-chip. We discuss the combination of state-of-the-art micropatterning technologies to achieve OoCs resembling human-intrinsic complexity. It is anticipated that these innovations will yield significant advances in realization of the next generation of OoC models.
Collapse
Affiliation(s)
- Cécile Bosmans
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands
| | - Núria Ginés Rodriguez
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Liliana Moreira Teixeira
- Department of Advanced Organ bioengineering and Therapeutics, University of Twente, Enschede, The Netherlands.
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Jeroen Leijten
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
15
|
Kaminaga M, Otomo S, Tsunozaki S, Kadonosono T, Omata T. Fabrication of a Cancer Cell Aggregate Culture Device That Facilitates Observations of Nutrient and Oxygen Gradients. MICROMACHINES 2024; 15:689. [PMID: 38930659 PMCID: PMC11205477 DOI: 10.3390/mi15060689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
Three-dimensional cell culture spheroids are commonly used for drug evaluation studies because they can produce large quantities of homogeneous cell aggregates. As the spheroids grow, nutrients supplied from outer spheroid regions render the inner spheroid areas hypoxic and hyponutrient, which makes them unobservable through confocal microscopy. In this study, we fabricated a cancer cell aggregate culture device that facilitates the observation of nutrient and oxygen gradients. An alginate gel fiber was created in the cell culture chamber to ensure a flow path for supplying the culture medium. A gradient of nutrients and oxygen was generated by positioning the flow channel close to the edge of the chamber. We devised a fabrication method that uses calcium carbonate as a source of Ca2+ for the gelation of sodium alginate, which has a slow reaction rate. We then cultured a spheroid of HCT116 cells, which were derived from human colorectal carcinoma using a fluorescent ubiquitination-based cell cycle indicator. Fluorescence observation suggested the formation of a hypoxic and hyponutrient region within an area approximately 500 µm away from the alginate gel fiber. This indicates the development of a cancer cell aggregate culture device that enables the observation of different nutrition and oxygen states.
Collapse
Affiliation(s)
- Maho Kaminaga
- Department of Mechanical Engineering, National Institute of Technology, Toyota Campus, 2-1 Eisei-cho, Toyota 471-0067, Aichi, Japan
| | - Shuta Otomo
- Department of Mechanical Engineering, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori ku, Yokohama 226-0026, Kanagawa, Japan (T.O.)
| | - Seisyu Tsunozaki
- Department of Mechanical Engineering, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori ku, Yokohama 226-0026, Kanagawa, Japan (T.O.)
| | - Tetuya Kadonosono
- Department of Life Science & Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori ku, Yokohama 226-0026, Kanagawa, Japan;
| | - Toru Omata
- Department of Mechanical Engineering, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori ku, Yokohama 226-0026, Kanagawa, Japan (T.O.)
| |
Collapse
|
16
|
Hsu YH, Yang WC, Chen YT, Lin CY, Yang CF, Liu WW, Shivani S, Li PC. Spatially controlled diffusion range of tumor-associated angiogenic factors to develop a tumor model using a microfluidic resistive circuit. LAB ON A CHIP 2024; 24:2644-2657. [PMID: 38576341 DOI: 10.1039/d3lc00891f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Developing a tumor model with vessels has been a challenge in microfluidics. This difficulty is because cancer cells can overgrow in a co-culture system. The up-regulation of anti-angiogenic factors during the initial tumor development can hinder neovascularization. The standard method is to develop a quiescent vessel network before loading a tumor construct in an adjacent chamber, which simulates the interaction between a tumor and its surrounding vessels. Here, we present a new method that allows a vessel network and a tumor to develop simultaneously in two linked chambers. The physiological environment of these two chambers is controlled by a microfluidic resistive circuit using two symmetric long microchannels. Applying the resistive circuit, a diffusion-dominated environment with a small 2-D pressure gradient is created across the two chambers with velocity <10.9 nm s-1 and Péclet number <6.3 × 10-5. This 2-D pressure gradient creates a V-shaped velocity clamp to confine the tumor-associated angiogenic factors at pores between the two chambers, and it has two functions. At the early stage, vasculogenesis is stimulated to grow a vessel network in the vessel chamber with minimal influence from the tumor that is still developed in the adjacent chamber. At the post-tumor-development stage, the induced steep concentration gradient at pores mimics vessel-tumor interactions to stimulate angiogenesis to grow vessels toward the tumor. Applying this method, we demonstrate that vasculogenic vessels can grow first, followed by stimulating angiogenesis. Angiogenic vessels can grow into stroma tissue up to 1.3 mm long, and vessels can also grow into or wrap around a 625 μm tumor spheroid or a tumor tissue developed from a cell suspension. In summary, our study suggests that the interactions between a developing vasculature and a growing tumor must be controlled differently throughout the tissue development process, including at the early stage when vessels are still forming and at the later stage when the tumor needs to interact with the vessels.
Collapse
Affiliation(s)
- Yu-Hsiang Hsu
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
- Graduate School of Advanced Technology, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C
| | - Wen-Chih Yang
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Yi-Ting Chen
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Che-Yu Lin
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Chiou-Fong Yang
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Wei-Wen Liu
- Graduate Institute of Oral Biology, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C
| | - Subhashree Shivani
- Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, R.O.C
| | - Pai-Chi Li
- Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, R.O.C
| |
Collapse
|
17
|
Llorente C. The Imperative for Innovative Enteric Nervous System-Intestinal Organoid Co-Culture Models: Transforming GI Disease Modeling and Treatment. Cells 2024; 13:820. [PMID: 38786042 PMCID: PMC11119846 DOI: 10.3390/cells13100820] [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: 03/15/2024] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
This review addresses the need for innovative co-culture systems integrating the enteric nervous system (ENS) with intestinal organoids. The breakthroughs achieved through these techniques will pave the way for a transformative era in gastrointestinal (GI) disease modeling and treatment strategies. This review serves as an introduction to the companion protocol paper featured in this journal. The protocol outlines the isolation and co-culture of myenteric and submucosal neurons with small intestinal organoids. This review provides an overview of the intestinal organoid culture field to establish a solid foundation for effective protocol application. Remarkably, the ENS surpasses the number of neurons in the spinal cord. Referred to as the "second brain", the ENS orchestrates pivotal roles in GI functions, including motility, blood flow, and secretion. The ENS is organized into myenteric and submucosal plexuses. These plexuses house diverse subtypes of neurons. Due to its proximity to the gut musculature and its cell type complexity, there are methodological intricacies in studying the ENS. Diverse approaches such as primary cell cultures, three-dimensional (3D) neurospheres, and induced ENS cells offer diverse insights into the multifaceted functionality of the ENS. The ENS exhibits dynamic interactions with the intestinal epithelium, the muscle layer, and the immune system, influencing epithelial physiology, motility, immune responses, and the microbiome. Neurotransmitters, including acetylcholine (ACh), serotonin (5-HT), and vasoactive intestinal peptide (VIP), play pivotal roles in these intricate interactions. Understanding these dynamics is imperative, as the ENS is implicated in various diseases, ranging from neuropathies to GI disorders and neurodegenerative diseases. The emergence of organoid technology presents an unprecedented opportunity to study ENS interactions within the complex milieu of the small and large intestines. This manuscript underscores the urgent need for standardized protocols and advanced techniques to unravel the complexities of the ENS and its dynamic relationship with the gut ecosystem. The insights gleaned from such endeavors hold the potential to revolutionize GI disease modeling and treatment paradigms.
Collapse
Affiliation(s)
- Cristina Llorente
- Department of Medicine, University of California San Diego, MC0063, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
18
|
Yu T, Yang Q, Peng B, Gu Z, Zhu D. Vascularized organoid-on-a-chip: design, imaging, and analysis. Angiogenesis 2024; 27:147-172. [PMID: 38409567 DOI: 10.1007/s10456-024-09905-z] [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: 09/22/2023] [Accepted: 01/11/2024] [Indexed: 02/28/2024]
Abstract
Vascularized organoid-on-a-chip (VOoC) models achieve substance exchange in deep layers of organoids and provide a more physiologically relevant system in vitro. Common designs for VOoC primarily involve two categories: self-assembly of endothelial cells (ECs) to form microvessels and pre-patterned vessel lumens, both of which include the hydrogel region for EC growth and allow for controlled fluid perfusion on the chip. Characterizing the vasculature of VOoC often relies on high-resolution microscopic imaging. However, the high scattering of turbid tissues can limit optical imaging depth. To overcome this limitation, tissue optical clearing (TOC) techniques have emerged, allowing for 3D visualization of VOoC in conjunction with optical imaging techniques. The acquisition of large-scale imaging data, coupled with high-resolution imaging in whole-mount preparations, necessitates the development of highly efficient analysis methods. In this review, we provide an overview of the chip designs and culturing strategies employed for VOoC, as well as the applicable optical imaging and TOC methods. Furthermore, we summarize the vascular analysis techniques employed in VOoC, including deep learning. Finally, we discuss the existing challenges in VOoC and vascular analysis methods and provide an outlook for future development.
Collapse
Affiliation(s)
- Tingting Yu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
- Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Qihang Yang
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
- Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, China
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu, 215163, China
| | - Dan Zhu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China.
- Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China.
| |
Collapse
|
19
|
Jouybar M, de Winde CM, Wolf K, Friedl P, Mebius RE, den Toonder JMJ. Cancer-on-chip models for metastasis: importance of the tumor microenvironment. Trends Biotechnol 2024; 42:431-448. [PMID: 37914546 DOI: 10.1016/j.tibtech.2023.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/03/2023]
Abstract
Cancer-on-chip (CoC) models, based on microfluidic chips harboring chambers for 3D tumor-cell culture, enable us to create a controlled tumor microenvironment (TME). CoC models are therefore increasingly used to systematically study effects of the TME on the various steps in cancer metastasis. Moreover, CoC models have great potential for developing novel cancer therapies and for predicting patient-specific response to cancer treatments. We review recent developments in CoC models, focusing on three main TME components: (i) the anisotropic extracellular matrix (ECM) architectures, (ii) the vasculature, and (iii) the immune system. We aim to provide guidance to biologists to choose the best CoC approach for addressing questions about the role of the TME in metastasis, and to inspire engineers to develop novel CoC technologies.
Collapse
Affiliation(s)
- Mohammad Jouybar
- Microsystems, Eindhoven University of Technology, Eindhoven, The Netherlands; Institute for Complex Molecular Systems, Eindhoven, The Netherlands
| | - Charlotte M de Winde
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology & Immunology, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands; Cancer Center Amsterdam, Cancer Biology & Immunology, Amsterdam, The Netherlands
| | - Katarina Wolf
- Department of Medical BioSciences, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter Friedl
- Department of Medical BioSciences, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Cancer Genomics Center, Utrecht, The Netherlands
| | - Reina E Mebius
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology & Immunology, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands; Cancer Center Amsterdam, Cancer Biology & Immunology, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Inflammatory diseases, Amsterdam, The Netherlands
| | - Jaap M J den Toonder
- Microsystems, Eindhoven University of Technology, Eindhoven, The Netherlands; Institute for Complex Molecular Systems, Eindhoven, The Netherlands.
| |
Collapse
|
20
|
Khanna A, Oropeza BP, Huang NF. Cardiovascular human organ-on-a-chip platform for disease modeling, drug development, and personalized therapy. J Biomed Mater Res A 2024; 112:512-523. [PMID: 37668192 PMCID: PMC11089005 DOI: 10.1002/jbm.a.37602] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/16/2023] [Accepted: 08/17/2023] [Indexed: 09/06/2023]
Abstract
Cardiovascular organ-on-a-chip (OoC) devices are composed of engineered or native functional tissues that are cultured under controlled microenvironments inside microchips. These systems employ microfabrication and tissue engineering techniques to recapitulate human physiology. This review focuses on human OoC systems to model cardiovascular diseases, to perform drug screening, and to advance personalized medicine. We also address the challenges in the generation of organ chips that can revolutionize the large-scale application of these systems for drug development and personalized therapy.
Collapse
Affiliation(s)
| | - Beu P. Oropeza
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, California, USA
- Center for Tissue Regeneration, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Ngan F. Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, California, USA
- Center for Tissue Regeneration, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
| |
Collapse
|
21
|
Sanaki-Matsumiya M, Villava C, Rappez L, Haase K, Wu J, Ebisuya M. Self-organization of vascularized skeletal muscle from bovine embryonic stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586252. [PMID: 38585777 PMCID: PMC10996461 DOI: 10.1101/2024.03.22.586252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Cultured beef holds promising potential as an alternative to traditional meat options. While adult stem cells are commonly used as the cell source for cultured beef, their proliferation and differentiation capacities are limited. To produce cultured beef steaks, current manufacturing plans often require the separate preparation of multiple cell types and intricate engineering for assembling them into structured tissues. In this study, we propose and report the co-induction of skeletal muscle, neuronal, and endothelial cells from bovine embryonic stem cells (ESCs) and the self-organization of tissue structures in 2- and 3-dimensional cultures. Bovine myocytes were induced in a stepwise manner through the induction of presomitic mesoderm (PSM) from bovine ESCs. Muscle fibers with sarcomeres appeared within 15 days, displaying calcium oscillations responsive to inputs from co-induced bovine spinal neurons. Bovine endothelial cells were also co-induced via PSM, forming uniform vessel networks inside tissues. Our serum-free, rapid co-induction protocols represent a milestone toward self-organizing beef steaks with integrated vasculature and innervation.
Collapse
Affiliation(s)
- Marina Sanaki-Matsumiya
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Casandra Villava
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Luca Rappez
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Kristina Haase
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Miki Ebisuya
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany
| |
Collapse
|
22
|
Amartumur S, Nguyen H, Huynh T, Kim TS, Woo RS, Oh E, Kim KK, Lee LP, Heo C. Neuropathogenesis-on-chips for neurodegenerative diseases. Nat Commun 2024; 15:2219. [PMID: 38472255 PMCID: PMC10933492 DOI: 10.1038/s41467-024-46554-8] [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: 10/04/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.
Collapse
Affiliation(s)
- Sarnai Amartumur
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Huong Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Thuy Huynh
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Testaverde S Kim
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, 34824, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Anti-microbial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
| | - Luke P Lee
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA.
| | - Chaejeong Heo
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|
23
|
Ma X, Wang Q, Li G, Li H, Xu S, Pang D. Cancer organoids: A platform in basic and translational research. Genes Dis 2024; 11:614-632. [PMID: 37692477 PMCID: PMC10491878 DOI: 10.1016/j.gendis.2023.02.052] [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: 05/14/2022] [Accepted: 02/16/2023] [Indexed: 09/12/2023] Open
Abstract
An accumulation of previous work has established organoids as good preclinical models of human tumors, facilitating translation from basic research to clinical practice. They are changing the paradigm of preclinical cancer research because they can recapitulate the heterogeneity and pathophysiology of human cancers and more closely approximate the complex tissue environment and structure found in clinical tumors than in vitro cell lines and animal models. However, the potential applications of cancer organoids remain to be comprehensively summarized. In the review, we firstly describe what is currently known about cancer organoid culture and then discuss in depth the basic mechanisms, including tumorigenesis and tumor metastasis, and describe recent advances in patient-derived tumor organoids (PDOs) for drug screening and immunological studies. Finally, the present challenges faced by organoid technology in clinical practice and its prospects are discussed. This review highlights that organoids may offer a novel therapeutic strategy for cancer research.
Collapse
Affiliation(s)
- Xin Ma
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
| | - Qin Wang
- Sino-Russian Medical Research Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
- Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang 150086, China
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Guozheng Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
| | - Hui Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
| | - Shouping Xu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
- Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang 150086, China
| | - Da Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
- Sino-Russian Medical Research Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
- Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang 150086, China
| |
Collapse
|
24
|
Nashimoto Y, Konno A, Imaizumi T, Nishikawa K, Ino K, Hori T, Kaji H, Shintaku H, Goto M, Shiku H. Microfluidic vascular formation model for assessing angiogenic capacities of single islets. Biotechnol Bioeng 2024; 121:1050-1059. [PMID: 38131167 DOI: 10.1002/bit.28631] [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: 05/26/2023] [Revised: 09/12/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023]
Abstract
Pancreatic islet transplantation presents a promising therapy for individuals suffering from type 1 diabetes. To maintain the function of transplanted islets in vivo, it is imperative to induce angiogenesis. However, the mechanisms underlying angiogenesis triggered by islets remain unclear. In this study, we introduced a microphysiological system to study the angiogenic capacity and dynamics of individual islets. The system, which features an open-top structure, uniquely facilitates the inoculation of islets and the longitudinal observation of vascular formation in in vivo like microenvironment with islet-endothelial cell communication. By leveraging our system, we discovered notable islet-islet heterogeneity in the angiogenic capacity. Transcriptomic analysis of the vascularized islets revealed that islets with high angiogenic capacity exhibited upregulation of genes related to insulin secretion and downregulation of genes related to angiogenesis and fibroblasts. In conclusion, our microfluidic approach is effective in characterizing the vascular formation of individual islets and holds great promise for elucidating the angiogenic mechanisms that enhance islet transplantation therapy.
Collapse
Affiliation(s)
- Yuji Nashimoto
- Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Miyagi, Japan
- Graduate School of Engineering, Tohoku University, Miyagi, Japan
- Graduate School of Environmental Studies, Tohoku University, Miyagi, Japan
- Cluster for Pioneering Research, RIKEN, Saitama, Japan
| | - An Konno
- Graduate School of Environmental Studies, Tohoku University, Miyagi, Japan
| | - Takuto Imaizumi
- Graduate School of Environmental Studies, Tohoku University, Miyagi, Japan
| | | | - Kosuke Ino
- Graduate School of Engineering, Tohoku University, Miyagi, Japan
| | - Takeshi Hori
- Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hirokazu Kaji
- Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hirofumi Shintaku
- Cluster for Pioneering Research, RIKEN, Saitama, Japan
- Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Masafumi Goto
- Division of Transplantation and Regenerative Medicine, Graduate School of Medicine, Tohoku University, Miyagi, Japan
| | - Hitoshi Shiku
- Graduate School of Engineering, Tohoku University, Miyagi, Japan
- Graduate School of Environmental Studies, Tohoku University, Miyagi, Japan
| |
Collapse
|
25
|
Shaji M, Tamada A, Fujimoto K, Muguruma K, Karsten SL, Yokokawa R. Deciphering potential vascularization factors of on-chip co-cultured hiPSC-derived cerebral organoids. LAB ON A CHIP 2024; 24:680-696. [PMID: 38284292 DOI: 10.1039/d3lc00930k] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
The lack of functional vascular system in stem cell-derived cerebral organoids (COs) limits their utility in modeling developmental processes and disease pathologies. Unlike other organs, brain vascularization is poorly understood, which makes it particularly difficult to mimic in vitro. Although several attempts have been made to vascularize COs, complete vascularization leading to functional capillary network development has only been achieved via transplantation into a mouse brain. Understanding the cues governing neurovascular communication is therefore imperative for establishing an efficient in vitro system for vascularized cerebral organoids that can emulate human brain development. Here, we used a multidisciplinary approach combining microfluidics, organoids, and transcriptomics to identify molecular changes in angiogenic programs that impede the successful in vitro vascularization of human induced pluripotent stem cell (iPSC)-derived COs. First, we established a microfluidic cerebral organoid (CO)-vascular bed (VB) co-culture system and conducted transcriptome analysis on the outermost cell layer of COs cultured on the preformed VB. Results revealed coordinated regulation of multiple pro-angiogenic factors and their downstream targets. The VEGF-HIF1A-AKT network was identified as a central pathway involved in the angiogenic response of cerebral organoids to the preformed VB. Among the 324 regulated genes associated with angiogenesis, six transcripts represented significantly regulated growth factors with the capacity to influence angiogenic activity during co-culture. Subsequent on-chip experiments demonstrated the angiogenic and vasculogenic potential of cysteine-rich angiogenic inducer 61 (CYR61) and hepatoma-derived growth factor (HDGF) as potential enhancers of organoid vascularization. Our study provides the first global analysis of cerebral organoid response to three-dimensional microvasculature for in vitro vascularization.
Collapse
Affiliation(s)
- Maneesha Shaji
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto - 615-8540, Japan.
| | - Atsushi Tamada
- Department of iPS Cell Applied Medicine, Kansai Medical University, 2-5-1 Shin-machi, Hirakata City, Osaka - 573-1010, Japan.
| | - Kazuya Fujimoto
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto - 615-8540, Japan.
| | - Keiko Muguruma
- Department of iPS Cell Applied Medicine, Kansai Medical University, 2-5-1 Shin-machi, Hirakata City, Osaka - 573-1010, Japan.
| | - Stanislav L Karsten
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto - 615-8540, Japan.
| | - Ryuji Yokokawa
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto - 615-8540, Japan.
| |
Collapse
|
26
|
Di Cio S, Marhuenda E, Haddrick M, Gautrot JE. Vascularised cardiac spheroids-on-a-chip for testing the toxicity of therapeutics. Sci Rep 2024; 14:3370. [PMID: 38336810 PMCID: PMC10858047 DOI: 10.1038/s41598-024-53678-w] [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: 09/13/2023] [Accepted: 02/03/2024] [Indexed: 02/12/2024] Open
Abstract
Microfabricated organ-on-a-chips are rapidly becoming the gold standard for the testing of safety and efficacy of therapeutics. A broad range of designs has emerged, but recreating microvascularised tissue models remains difficult in many cases. This is particularly relevant to mimic the systemic delivery of therapeutics, to capture the complex multi-step processes associated with trans-endothelial transport or diffusion, uptake by targeted tissues and associated metabolic response. In this report, we describe the formation of microvascularised cardiac spheroids embedded in microfluidic chips. Different protocols used for embedding spheroids within vascularised multi-compartment microfluidic chips were investigated first to identify the importance of the spheroid processing, and co-culture with pericytes on the integration of the spheroid within the microvascular networks formed. The architecture of the resulting models, the expression of cardiac and endothelial markers and the perfusion of the system was then investigated. This confirmed the excellent stability of the vascular networks formed, as well as the persistent expression of cardiomyocyte markers such as cTNT and the assembly of striated F-actin, myosin and α-actinin cytoskeletal networks typically associated with contractility and beating. The ability to retain beating over prolonged periods of time was quantified, over 25 days, demonstrating not only perfusability but also functional performance of the tissue model. Finally, as a proof-of-concept of therapeutic testing, the toxicity of one therapeutic associated with cardiac disfunction was evaluated, identifying differences between direct in vitro testing on suspended spheroids and vascularised models.
Collapse
Affiliation(s)
- Stefania Di Cio
- Institute of Bioengineering, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
- School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| | - Emilie Marhuenda
- Institute of Bioengineering, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
- School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| | - Malcolm Haddrick
- Medicines Discovery Catapult, Alderley Park, Cheshire, SK10 4TG, UK
| | - Julien E Gautrot
- Institute of Bioengineering, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK.
- School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK.
| |
Collapse
|
27
|
De Lorenzi F, Hansen N, Theek B, Daware R, Motta A, Breuel S, Nasehi R, Baumeister J, Schöneberg J, Stojanović N, von Stillfried S, Vogt M, Müller-Newen G, Maurer J, Sofias AM, Lammers T, Fischer H, Kiessling F. Engineering Mesoscopic 3D Tumor Models with a Self-Organizing Vascularized Matrix. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2303196. [PMID: 37865947 DOI: 10.1002/adma.202303196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/05/2023] [Indexed: 10/24/2023]
Abstract
Advanced in vitro systems such as multicellular spheroids and lab-on-a-chip devices have been developed, but often fall short in reproducing the tissue scale and self-organization of human diseases. A bioprinted artificial tumor model is introduced with endothelial and stromal cells self-organizing into perfusable and functional vascular structures. This model uses 3D hydrogel matrices to embed multicellular tumor spheroids, allowing them to grow to mesoscopic scales and to interact with endothelial cells. It is shown that angiogenic multicellular tumor spheroids promote the growth of a vascular network, which in turn further enhances the growth of cocultivated tumor spheroids. The self-developed vascular structure infiltrates the tumor spheroids, forms functional connections with the bioprinted endothelium, and can be perfused by erythrocytes and polystyrene microspheres. Moreover, cancer cells migrate spontaneously from the tumor spheroid through the self-assembled vascular network into the fluid flow. Additionally, tumor type specific characteristics of desmoplasia, angiogenesis, and metastatic propensity are preserved between patient-derived samples and tumors derived from this same material growing in the bioreactors. Overall, this modular approach opens up new avenues for studying tumor pathophysiology and cellular interactions in vitro, providing a platform for advanced drug testing while reducing the need for in vivo experimentation.
Collapse
Affiliation(s)
- Federica De Lorenzi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, 52074, Aachen, Germany
- Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIOABCD), RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Nadja Hansen
- Department of Dental Materials and Biomaterials Research (ZWBF), RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Benjamin Theek
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Rasika Daware
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Alessandro Motta
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Saskia Breuel
- Department of Gynecology and Obstetrics, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Ramin Nasehi
- Department of Dental Materials and Biomaterials Research (ZWBF), RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Julian Baumeister
- Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIOABCD), RWTH Aachen University Hospital, 52074, Aachen, Germany
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Jan Schöneberg
- Department of Dental Materials and Biomaterials Research (ZWBF), RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Natalija Stojanović
- Department of Dental Materials and Biomaterials Research (ZWBF), RWTH Aachen University Hospital, 52074, Aachen, Germany
| | | | - Michael Vogt
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Gerhard Müller-Newen
- Institute of Biochemistry and Molecular Biology, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Jochen Maurer
- Department of Gynecology and Obstetrics, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Alexandros Marios Sofias
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, 52074, Aachen, Germany
- Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIOABCD), RWTH Aachen University Hospital, 52074, Aachen, Germany
- Norwegian University of Science and Technology (NTNU), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Trondheim, 7491, Norway
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, 52074, Aachen, Germany
- Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIOABCD), RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research (ZWBF), RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, 52074, Aachen, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, 28359, Bremen, Germany
| |
Collapse
|
28
|
Hirth E, Cao W, Peltonen M, Kapetanovic E, Dietsche C, Svanberg S, Filippova M, Reddy S, Dittrich PS. Self-assembled and perfusable microvasculature-on-chip for modeling leukocyte trafficking. LAB ON A CHIP 2024; 24:292-304. [PMID: 38086670 PMCID: PMC10793075 DOI: 10.1039/d3lc00719g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/20/2023] [Indexed: 01/18/2024]
Abstract
Leukocyte recruitment from blood to tissue is a process that occurs at the level of capillary vessels during both physiological and pathological conditions. This process is also relevant for evaluating novel adoptive cell therapies, in which the trafficking of therapeutic cells such as chimeric antigen receptor (CAR)-T cells throughout the capillaries of solid tumors is important. Local variations in blood flow, mural cell concentration, and tissue stiffness contribute to the regulation of capillary vascular permeability and leukocyte trafficking throughout the capillary microvasculature. We developed a platform to mimic a biologically functional human arteriole-venule microcirculation system consisting of pericytes (PCs) and arterial and venous primary endothelial cells (ECs) embedded within a hydrogel, which self-assembles into a perfusable, heterogeneous microvasculature. Our device shows a preferential association of PCs with arterial ECs that drives the flow-dependent formation of microvasculature networks. We show that PCs stimulate basement membrane matrix synthesis, which affects both vessel diameter and permeability in a manner correlating with the ratio of ECs to PCs. Moreover, we demonstrate that hydrogel concentration can affect capillary morphology but has no observed effect on vascular permeability. The biological function of our capillary network was demonstrated using an inflammation model, where significantly higher expression of cytokines, chemokines, and adhesion molecules was observed after tumor necrosis factor-alpha (TNF-α) treatment. Accordingly, T cell adherence and transendothelial migration were significantly increased in the immune-activated state. Taken together, our platform allows the generation of a perfusable microvasculature that recapitulates the structure and function of an in vivo capillary bed that can be used as a model for developing potential immunotherapies.
Collapse
Affiliation(s)
- Elisabeth Hirth
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Wuji Cao
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Marina Peltonen
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Edo Kapetanovic
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Claudius Dietsche
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Sara Svanberg
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Maria Filippova
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Sai Reddy
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Petra S Dittrich
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| |
Collapse
|
29
|
Tian M, Ma Z, Yang GZ. Micro/nanosystems for controllable drug delivery to the brain. Innovation (N Y) 2024; 5:100548. [PMID: 38161522 PMCID: PMC10757293 DOI: 10.1016/j.xinn.2023.100548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/26/2023] [Indexed: 01/03/2024] Open
Abstract
Drug delivery to the brain is crucial in the treatment for central nervous system disorders. While significant progress has been made in recent years, there are still major challenges in achieving controllable drug delivery to the brain. Unmet clinical needs arise from various factors, including controlled drug transport, handling large drug doses, methods for crossing biological barriers, the use of imaging guidance, and effective models for analyzing drug delivery. Recent advances in micro/nanosystems have shown promise in addressing some of these challenges. These include the utilization of microfluidic platforms to test and validate the drug delivery process in a controlled and biomimetic setting, the development of novel micro/nanocarriers for large drug loads across the blood-brain barrier, and the implementation of micro-intervention systems for delivering drugs through intraparenchymal or peripheral routes. In this article, we present a review of the latest developments in micro/nanosystems for controllable drug delivery to the brain. We also delve into the relevant diseases, biological barriers, and conventional methods. In addition, we discuss future prospects and the development of emerging robotic micro/nanosystems equipped with directed transportation, real-time image guidance, and closed-loop control.
Collapse
Affiliation(s)
- Mingzhen Tian
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhichao Ma
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guang-Zhong Yang
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
30
|
Bhattacharya A, Alam K, Roy NS, Kaur K, Kaity S, Ravichandiran V, Roy S. Exploring the interaction between extracellular matrix components in a 3D organoid disease model to replicate the pathophysiology of breast cancer. J Exp Clin Cancer Res 2023; 42:343. [PMID: 38102637 PMCID: PMC10724947 DOI: 10.1186/s13046-023-02926-4] [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: 10/20/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
In vitro models are necessary to study the pathophysiology of the disease and the development of effective, tailored treatment methods owing to the complexity and heterogeneity of breast cancer and the large population affected by it. The cellular connections and tumor microenvironments observed in vivo are often not recapitulated in conventional two-dimensional (2D) cell cultures. Therefore, developing 3D in vitro models that mimic the complex architecture and physiological circumstances of breast tumors is crucial for advancing our understanding of the illness. A 3D scaffold-free in vitro disease model mimics breast cancer pathophysiology by allowing cells to self-assemble/pattern into 3D structures, in contrast with other 3D models that rely on artificial scaffolds. It is possible that this model, whether applied to breast tumors using patient-derived primary cells (fibroblasts, endothelial cells, and cancer cells), can accurately replicate the observed heterogeneity. The complicated interactions between different cell types are modelled by integrating critical components of the tumor microenvironment, such as the extracellular matrix, vascular endothelial cells, and tumor growth factors. Tissue interactions, immune cell infiltration, and the effects of the milieu on drug resistance can be studied using this scaffold-free 3D model. The scaffold-free 3D in vitro disease model for mimicking tumor pathophysiology in breast cancer is a useful tool for studying the molecular basis of the disease, identifying new therapeutic targets, and evaluating treatment modalities. It provides a more physiologically appropriate high-throughput platform for screening large compound library in a 96-384 well format. We critically discussed the rapid development of personalized treatment strategies and accelerated drug screening platforms to close the gap between traditional 2D cell culture and in vivo investigations.
Collapse
Affiliation(s)
- Anamitra Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kamare Alam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Nakka Sharmila Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Santanu Kaity
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
31
|
Liang J, Zhao J, Chen Y, Li B, Li Y, Lu F, Dong Z. New Insights and Advanced Strategies for In Vitro Construction of Vascularized Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:692-709. [PMID: 37409413 DOI: 10.1089/ten.teb.2023.0044] [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: 07/07/2023]
Abstract
Inadequate vascularization is a significant barrier to clinical application of large-volume tissue engineered grafts. In contrast to in vivo vascularization, in vitro prevascularization shortens the time required for host vessels to grow into the graft core and minimizes necrosis in the core region of the graft. However, the challenge of prevascularization is to construct hierarchical perfusable vascular networks, increase graft volume, and form a vascular tip that can anastomose with host vessels. Understanding advances in in vitro prevascularization techniques and new insights into angiogenesis could overcome these obstacles. In the present review, we discuss new perspectives on angiogenesis, the differences between in vivo and in vitro tissue vascularization, the four elements of prevascularized constructs, recent advances in perfusion-based in vitro prevascularized tissue fabrication, and prospects for large-volume prevascularized tissue engineering.
Collapse
Affiliation(s)
- Jiancong Liang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jing Zhao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yunzi Chen
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Bin Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ye Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ziqing Dong
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
32
|
Wang YT, Meng XT. A review of the evidence to support electrical stimulation -induced vascularization in engineered tissue. Regen Ther 2023; 24:237-244. [PMID: 37534238 PMCID: PMC10393514 DOI: 10.1016/j.reth.2023.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/25/2023] [Accepted: 07/10/2023] [Indexed: 08/04/2023] Open
Abstract
Tissue engineering presents a promising solution for regenerative medicine and the success depends on the supply of oxygen/nutrients to the cells by rapid vascularization. More and more technologies are being developed to facilitate vascularization of engineered tissues. In this review, we indicated that a regulatory system which influences all angiogenesis associated cells to achieve their desired functional state is ideal for the construction of vascularized engineered tissues in vitro. We presented the evidence that electrical stimulation (ES) enhances the synergistic promotion of co-cultured angiogenesis associated cells and its potential regulatory mechanisms, highlighted the potential advantages of a combination of mesenchymal stem cells (MSCs), endothelial cells (ECs) and ES to achieve tissue vascularization, with particular emphasis on the different biological pathways of ES-regulated ECs. Finally, we proposed the future direction of using ES to reconstruct engineered tissue blood vessels, pointed out the potential advantages and disadvantages of ES application on tissue vascularization.
Collapse
Affiliation(s)
- Ying-tong Wang
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, PR China
- The Undergraduate Center of Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Xiao-ting Meng
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, PR China
| |
Collapse
|
33
|
Murphy AR, Allenby MC. In vitro microvascular engineering approaches and strategies for interstitial tissue integration. Acta Biomater 2023; 171:114-130. [PMID: 37717711 DOI: 10.1016/j.actbio.2023.09.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023]
Abstract
The increasing gap between clinical demand for tissue or organ transplants and the availability of donated tissue highlights the emerging opportunities for lab-grown or synthetically engineered tissue. While the field of tissue engineering has existed for nearly half a century, its clinical translation remains unrealised, in part, due to a limited ability to engineer sufficient vascular supply into fabricated tissue, which is necessary to enable nutrient and waste exchange, prevent cellular necrosis, and support tissue proliferation. Techniques to develop anatomically relevant, functional vascular networks in vitro have made significant progress in the last decade, however, the challenge now remains as to how best incorporate these throughout dense parenchymal tissue-like structures to address diffusion-limited development and allow for the fabrication of large-scale vascularised tissue. This review explores advances made in the laboratory engineering of vasculature structures and summarises recent attempts to integrate vascular networks together with sophisticated in vitro avascular tissue and organ-like structures. STATEMENT OF SIGNIFICANCE: The ability to grow full scale, functional tissue and organs in vitro is primarily limited by an inability to adequately diffuse oxygen and nutrients throughout developing cellularised structures, which generally results from the absence of perfusable vessel networks. Techniques to engineering both perfusable vascular networks and avascular miniaturised organ-like structures have recently increased in complexity, sophistication, and physiological relevance. However, integrating these two essential elements into a single functioning vascularised tissue structure represents a significant spatial and temporal engineering challenge which is yet to be surmounted. Here, we explore a range of vessel morphogenic phenomena essential for tissue-vascular co-development, as well as evaluate a range of recent noteworthy approaches for generating vascularised tissue products in vitro.
Collapse
Affiliation(s)
- A R Murphy
- School of Chemical Engineering, Faculty of Engineering, Architecture and Information Technology, The University of Queensland, St Lucia, QLD 4100, Australia
| | - M C Allenby
- School of Chemical Engineering, Faculty of Engineering, Architecture and Information Technology, The University of Queensland, St Lucia, QLD 4100, Australia; Centre for Biomedical Technologies, School of Medical, Mechanical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia.
| |
Collapse
|
34
|
Liu Y, Yao X, Fan C, Zhang G, Luo X, Qian Y. Microfabrication and lab-on-a-chip devices promote in vitromodeling of neural interfaces for neuroscience researches and preclinical applications. Biofabrication 2023; 16:012002. [PMID: 37832555 DOI: 10.1088/1758-5090/ad032a] [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: 10/13/2023] [Indexed: 10/15/2023]
Abstract
Neural tissues react to injuries through the orchestration of cellular reprogramming, generating specialized cells and activating gene expression that helps with tissue remodeling and homeostasis. Simplified biomimetic models are encouraged to amplify the physiological and morphological changes during neural regeneration at cellular and molecular levels. Recent years have witnessed growing interest in lab-on-a-chip technologies for the fabrication of neural interfaces. Neural system-on-a-chip devices are promisingin vitromicrophysiological platforms that replicate the key structural and functional characteristics of neural tissues. Microfluidics and microelectrode arrays are two fundamental techniques that are leveraged to address the need for microfabricated neural devices. In this review, we explore the innovative fabrication, mechano-physiological parameters, spatiotemporal control of neural cell cultures and chip-based neurogenesis. Although the high variability in different constructs, and the restriction in experimental and analytical access limit the real-life applications of microphysiological models, neural system-on-a-chip devices have gained considerable translatability for modeling neuropathies, drug screening and personalized therapy.
Collapse
Affiliation(s)
- Yang Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Xiangyun Yao
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
| | - Guifeng Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Xi Luo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Yun Qian
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
| |
Collapse
|
35
|
Mieville V, Griffioen AW, Benamran D, Nowak-Sliwinska P. Advanced in vitro models for renal cell carcinoma therapy design. Biochim Biophys Acta Rev Cancer 2023; 1878:188942. [PMID: 37343729 DOI: 10.1016/j.bbcan.2023.188942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
Renal cell carcinoma (RCC) and its principal subtype, clear cell RCC, are the most diagnosed kidney cancer. Despite substantial improvement over the last decades, current pharmacological intervention still fails to achieve long-term therapeutic success. RCC is characterized by a high intra- and inter-tumoral heterogeneity and is heavily influenced by the crosstalk of the cells composing the tumor microenvironment, such as cancer-associated fibroblasts, endothelial cells and immune cells. Moreover, multiple physicochemical properties such as pH, interstitial pressure or oxygenation may also play an important role. These elements are often poorly recapitulated in in vitro models used for drug development. This inadequate recapitulation of the tumor is partially responsible for the current lack of an effective and curative treatment. Therefore, there are needs for more complex in vitro or ex vivo drug screening models. In this review, we discuss the current state-of-the-art of RCC models and suggest strategies for their further development.
Collapse
Affiliation(s)
- Valentin Mieville
- School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland; Translational Research Center in Oncohaematology, Geneva, Switzerland
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Daniel Benamran
- Division of Urology, Geneva University Hospitals, Geneva, Switzerland
| | - Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland; Translational Research Center in Oncohaematology, Geneva, Switzerland.
| |
Collapse
|
36
|
Rifai A, Weerasinghe DK, Tilaye GA, Nisbet D, Hodge JM, Pasco JA, Williams LJ, Samarasinghe RM, Williams RJ. Biofabrication of functional bone tissue: defining tissue-engineered scaffolds from nature. Front Bioeng Biotechnol 2023; 11:1185841. [PMID: 37614632 PMCID: PMC10444209 DOI: 10.3389/fbioe.2023.1185841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/24/2023] [Indexed: 08/25/2023] Open
Abstract
Damage to bone leads to pain and loss of movement in the musculoskeletal system. Although bone can regenerate, sometimes it is damaged beyond its innate capacity. Research interest is increasingly turning to tissue engineering (TE) processes to provide a clinical solution for bone defects. Despite the increasing biomimicry of tissue-engineered scaffolds, significant gaps remain in creating the complex bone substitutes, which include the biochemical and physical conditions required to recapitulate bone cells' natural growth, differentiation and maturation. Combining advanced biomaterials with new additive manufacturing technologies allows the development of 3D tissue, capable of forming cell aggregates and organoids based on natural and stimulated cues. Here, we provide an overview of the structure and mechanical properties of natural bone, the role of bone cells, the remodelling process, cytokines and signalling pathways, causes of bone defects and typical treatments and new TE strategies. We highlight processes of selecting biomaterials, cells and growth factors. Finally, we discuss innovative tissue-engineered models that have physiological and anatomical relevance for cancer treatments, injectable stimuli gels, and other therapeutic drug delivery systems. We also review current challenges and prospects of bone TE. Overall, this review serves as guide to understand and develop better tissue-engineered bone designs.
Collapse
Affiliation(s)
- Aaqil Rifai
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - D. Kavindi Weerasinghe
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Gebreselassie Addisu Tilaye
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - David Nisbet
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, VIC, Australia
- Laboratory of Advanced Biomaterials, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Aikenhead Centre for Medical Discovery, St. Vincent’s Hospital, Melbourne, VIC, Australia
| | - Jason M. Hodge
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| | - Julie A. Pasco
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Department of Medicine-Western Health, The University of Melbourne, St Albans, VIC, Australia
| | - Lana J. Williams
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| | - Rasika M. Samarasinghe
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Richard J. Williams
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Aikenhead Centre for Medical Discovery, St. Vincent’s Hospital, Melbourne, VIC, Australia
| |
Collapse
|
37
|
Gerardo‐Nava JL, Jansen J, Günther D, Klasen L, Thiebes AL, Niessing B, Bergerbit C, Meyer AA, Linkhorst J, Barth M, Akhyari P, Stingl J, Nagel S, Stiehl T, Lampert A, Leube R, Wessling M, Santoro F, Ingebrandt S, Jockenhoevel S, Herrmann A, Fischer H, Wagner W, Schmitt RH, Kiessling F, Kramann R, De Laporte L. Transformative Materials to Create 3D Functional Human Tissue Models In Vitro in a Reproducible Manner. Adv Healthc Mater 2023; 12:e2301030. [PMID: 37311209 PMCID: PMC11468549 DOI: 10.1002/adhm.202301030] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/21/2023] [Indexed: 06/15/2023]
Abstract
Recreating human tissues and organs in the petri dish to establish models as tools in biomedical sciences has gained momentum. These models can provide insight into mechanisms of human physiology, disease onset, and progression, and improve drug target validation, as well as the development of new medical therapeutics. Transformative materials play an important role in this evolution, as they can be programmed to direct cell behavior and fate by controlling the activity of bioactive molecules and material properties. Using nature as an inspiration, scientists are creating materials that incorporate specific biological processes observed during human organogenesis and tissue regeneration. This article presents the reader with state-of-the-art developments in the field of in vitro tissue engineering and the challenges related to the design, production, and translation of these transformative materials. Advances regarding (stem) cell sources, expansion, and differentiation, and how novel responsive materials, automated and large-scale fabrication processes, culture conditions, in situ monitoring systems, and computer simulations are required to create functional human tissue models that are relevant and efficient for drug discovery, are described. This paper illustrates how these different technologies need to converge to generate in vitro life-like human tissue models that provide a platform to answer health-based scientific questions.
Collapse
|
38
|
Sharma A, Fernandes DC, Reis RL, Gołubczyk D, Neumann S, Lukomska B, Janowski M, Kortylewski M, Walczak P, Oliveira JM, Maciaczyk J. Cutting-edge advances in modeling the blood-brain barrier and tools for its reversible permeabilization for enhanced drug delivery into the brain. Cell Biosci 2023; 13:137. [PMID: 37501215 PMCID: PMC10373415 DOI: 10.1186/s13578-023-01079-3] [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: 04/21/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023] Open
Abstract
The blood-brain barrier (BBB) is a sophisticated structure whose full functionality is required for maintaining the executive functions of the central nervous system (CNS). Tight control of transport across the barrier means that most drugs, particularly large size, which includes powerful biologicals, cannot reach their targets in the brain. Notwithstanding the remarkable advances in characterizing the cellular nature of the BBB and consequences of BBB dysfunction in pathology (brain metastasis, neurological diseases), it remains challenging to deliver drugs to the CNS. Herein, we outline the basic architecture and key molecular constituents of the BBB. In addition, we review the current status of approaches that are being explored to temporarily open the BBB in order to allow accumulation of therapeutics in the CNS. Undoubtedly, the major concern in field is whether it is possible to open the BBB in a meaningful way without causing negative consequences. In this context, we have also listed few other important key considerations that can improve our understanding about the dynamics of the BBB.
Collapse
Affiliation(s)
- Amit Sharma
- Department of Stereotacitc and Functional Neurosurgery, University Hospital Bonn, 53127, Bonn, Germany
| | - Diogo C Fernandes
- 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, 4710-057, Braga, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga, Portugal
| | - Dominika Gołubczyk
- Ti-Com, Polish Limited Liability Company, 10-683, Olsztyn, Poland
- Center for Translational Medicine, Warsaw University of Life Sciences, 02-797, Warsaw, Poland
| | - Silke Neumann
- Department of Pathology, University of Otago, Dunedin, 9054, New Zealand
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marcin Kortylewski
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - J Miguel Oliveira
- 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, 4710-057, Braga, Portugal.
| | - Jarek Maciaczyk
- Department of Stereotacitc and Functional Neurosurgery, University Hospital Bonn, 53127, Bonn, Germany.
- Department of Surgical Sciences, University of Otago, Dunedin, 9054, New Zealand.
| |
Collapse
|
39
|
Zeng X, Ma Q, Li XK, You LT, Li J, Fu X, You FM, Ren YF. Patient-derived organoids of lung cancer based on organoids-on-a-chip: enhancing clinical and translational applications. Front Bioeng Biotechnol 2023; 11:1205157. [PMID: 37304140 PMCID: PMC10250649 DOI: 10.3389/fbioe.2023.1205157] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
Lung cancer is one of the most common malignant tumors worldwide, with high morbidity and mortality due to significant individual characteristics and genetic heterogeneity. Personalized treatment is necessary to improve the overall survival rate of the patients. In recent years, the development of patient-derived organoids (PDOs) enables lung cancer diseases to be simulated in the real world, and closely reflects the pathophysiological characteristics of natural tumor occurrence and metastasis, highlighting their great potential in biomedical applications, translational medicine, and personalized treatment. However, the inherent defects of traditional organoids, such as poor stability, the tumor microenvironment with simple components and low throughput, limit their further clinical transformation and applications. In this review, we summarized the developments and applications of lung cancer PDOs and discussed the limitations of traditional PDOs in clinical transformation. Herein, we looked into the future and proposed that organoids-on-a-chip based on microfluidic technology are advantageous for personalized drug screening. In addition, combined with recent advances in lung cancer research, we explored the translational value and future development direction of organoids-on-a-chip in the precision treatment of lung cancer.
Collapse
Affiliation(s)
- Xiao Zeng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiong Ma
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xue-Ke Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Cancer Institute, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Li-Ting You
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jia Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xi Fu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Feng-Ming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Cancer Institute, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yi-Feng Ren
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Cancer Institute, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
40
|
Yu J, Yin Y, Leng Y, Zhang J, Wang C, Chen Y, Li X, Wang X, Liu H, Liao Y, Jin Y, Zhang Y, Lu K, Wang K, Wang X, Wang L, Zheng F, Gu Z, Li Y, Fan Y. Emerging strategies of engineering retinal organoids and organoid-on-a-chip in modeling intraocular drug delivery: current progress and future perspectives. Adv Drug Deliv Rev 2023; 197:114842. [PMID: 37105398 DOI: 10.1016/j.addr.2023.114842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Retinal diseases are a rising concern as major causes of blindness in an aging society; therapeutic options are limited, and the precise pathogenesis of these diseases remains largely unknown. Intraocular drug delivery and nanomedicines offering targeted, sustained, and controllable delivery are the most challenging and popular topics in ocular drug development and toxicological evaluation. Retinal organoids (ROs) and organoid-on-a-chip (ROoC) are both emerging as promising in-vitro models to faithfully recapitulate human eyes for retinal research in the replacement of experimental animals and primary cells. In this study, we review the generation and application of ROs resembling the human retina in cell subtypes and laminated structures and introduce the emerging engineered ROoC as a technological opportunity to address critical issues. On-chip vascularization, perfusion, and close inter-tissue interactions recreate physiological environments in vitro, whilst integrating with biosensors facilitates real-time analysis and monitoring during organogenesis of the retina representing engineering efforts in ROoC models. We also emphasize that ROs and ROoCs hold the potential for applications in modeling intraocular drug delivery in vitro and developing next-generation retinal drug delivery strategies.
Collapse
Affiliation(s)
- Jiaheng Yu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yuqi Yin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yubing Leng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Jingcheng Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Chunyan Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yanyun Chen
- Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xiaorui Li
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Xudong Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Hui Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yulong Liao
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yishan Jin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yihan Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Keyu Lu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Kehao Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Xiaofei Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Lizhen Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Fuyin Zheng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China.
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Yubo Fan
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China.
| |
Collapse
|
41
|
Dibble M, Di Cio' S, Luo P, Balkwill F, Gautrot JE. The impact of pericytes on the stability of microvascular networks in response to nanoparticles. Sci Rep 2023; 13:5729. [PMID: 37029151 PMCID: PMC10082022 DOI: 10.1038/s41598-023-31352-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 03/10/2023] [Indexed: 04/09/2023] Open
Abstract
Recapitulating the normal physiology of the microvasculature is pivotal in the development of more complex in-vitro models and organ-on-chip designs. Pericytes are an important component of the vasculature, promoting vessel stability, inhibiting vascular permeability and maintaining the vascular hierarchical architecture. The use of such co-culture for the testing of therapeutics and nanoparticle safety is increasingly considered for the validation of therapeutic strategies. This report presents the use of a microfluidic model for such applications. Interactions between endothelial cells and pericytes are first explored. We identify basal conditions required to form stable and reproducible endothelial networks. We then investigate interactions between endothelial cells and pericytes via direct co-culture. In our system, pericytes prevented vessel hyperplasia and maintained vessel length in prolonged culture (> 10 days). In addition, these vessels displayed barrier function and expression of junction markers associated with vessel maturation, including VE-cadherin, β-catenin and ZO-1. Furthermore, pericytes maintained vessel integrity following stress (nutrient starvation) and prevented vessel regression, in contrast to the striking dissociation of networks in endothelial monocultures. This response was also observed when endothelial/pericyte co-cultures were exposed to high concentrations of moderately toxic cationic nanoparticles used for gene delivery. This study highlights the importance of pericytes in protecting vascular networks from stress and external agents and their importance to the design of advanced in-vitro models, including for the testing of nanotoxicity, to better recapitulate physiological response and avoid false positives.
Collapse
Affiliation(s)
- Matthew Dibble
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
- School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| | - Stefania Di Cio'
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
- School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| | - Piaopiao Luo
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
- School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| | - Frances Balkwill
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
- Barts Cancer Institute, Queen Mary, University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Julien E Gautrot
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK.
- School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK.
| |
Collapse
|
42
|
Voges HK, Foster SR, Reynolds L, Parker BL, Devilée L, Quaife-Ryan GA, Fortuna PRJ, Mathieson E, Fitzsimmons R, Lor M, Batho C, Reid J, Pocock M, Friedman CE, Mizikovsky D, Francois M, Palpant NJ, Needham EJ, Peralta M, Monte-Nieto GD, Jones LK, Smyth IM, Mehdiabadi NR, Bolk F, Janbandhu V, Yao E, Harvey RP, Chong JJH, Elliott DA, Stanley EG, Wiszniak S, Schwarz Q, James DE, Mills RJ, Porrello ER, Hudson JE. Vascular cells improve functionality of human cardiac organoids. Cell Rep 2023:112322. [PMID: 37105170 DOI: 10.1016/j.celrep.2023.112322] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/13/2023] [Accepted: 03/15/2023] [Indexed: 04/29/2023] Open
Abstract
Crosstalk between cardiac cells is critical for heart performance. Here we show that vascular cells within human cardiac organoids (hCOs) enhance their maturation, force of contraction, and utility in disease modeling. Herein we optimize our protocol to generate vascular populations in addition to epicardial, fibroblast, and cardiomyocyte cells that self-organize into in-vivo-like structures in hCOs. We identify mechanisms of communication between endothelial cells, pericytes, fibroblasts, and cardiomyocytes that ultimately contribute to cardiac organoid maturation. In particular, (1) endothelial-derived LAMA5 regulates expression of mature sarcomeric proteins and contractility, and (2) paracrine platelet-derived growth factor receptor β (PDGFRβ) signaling from vascular cells upregulates matrix deposition to augment hCO contractile force. Finally, we demonstrate that vascular cells determine the magnitude of diastolic dysfunction caused by inflammatory factors and identify a paracrine role of endothelin driving dysfunction. Together this study highlights the importance and role of vascular cells in organoid models.
Collapse
Affiliation(s)
- Holly K Voges
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia; Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Department of Paediatrics, School of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - Simon R Foster
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Liam Reynolds
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Benjamin L Parker
- Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney, NSW 2006, Australia; Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lynn Devilée
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Gregory A Quaife-Ryan
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | | | - Ellen Mathieson
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | | | - Mary Lor
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Christopher Batho
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Janice Reid
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Mark Pocock
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Clayton E Friedman
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, QLD, Australia
| | - Dalia Mizikovsky
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, QLD, Australia
| | - Mathias Francois
- The Centenary Institute, David Richmond Program for Cardiovascular Research: Gene Regulation and Editing, Sydney Medical School, University of Sydney, Sydney, NSW 2050, Australia
| | - Nathan J Palpant
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, QLD, Australia
| | - Elise J Needham
- Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Marina Peralta
- Australian Regenerative Medicine Institute. Monash University, Clayton, VIC 3800, Australia
| | | | - Lynelle K Jones
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedical Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Ian M Smyth
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedical Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Neda R Mehdiabadi
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - Francesca Bolk
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - Vaibhao Janbandhu
- Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Ernestene Yao
- Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia; School of Biotechnology and Biomolecular Science, UNSW Sydney, Sydney, NSW 2052, Australia
| | - James J H Chong
- Centre for Heart Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW 2145, Australia; Department of Cardiology, Westmead Hospital, Westmead, NSW 2145, Australia
| | - David A Elliott
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Department of Paediatrics, School of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - Edouard G Stanley
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Department of Paediatrics, School of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - Sophie Wiszniak
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5001, Australia
| | - Quenten Schwarz
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5001, Australia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Science, The University of Sydney, Sydney, NSW 2006, Australia; Sydney Medical School, The University of Sydney, Sydney, 2010 NSW, Australia
| | - Richard J Mills
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia; Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Department of Paediatrics, School of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - Enzo R Porrello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia; Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne, VIC 3052, Australia; Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, The Royal Children's Hospital, Melbourne, VIC 3052, Australia.
| | - James E Hudson
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
43
|
Zhou L, Liu L, Chang MA, Ma C, Chen W, Chen P. Spatiotemporal dissection of tumor microenvironment via in situ sensing and monitoring in tumor-on-a-chip. Biosens Bioelectron 2023; 225:115064. [PMID: 36680970 PMCID: PMC9918721 DOI: 10.1016/j.bios.2023.115064] [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: 10/23/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Real-time monitoring in the tumor microenvironment provides critical insights of cancer progression and mechanistic understanding of responses to cancer treatments. However, clinical challenges and significant questions remain regarding assessment of limited clinical tissue samples, establishment of validated, controllable pre-clinical cancer models, monitoring of static versus dynamic markers, and the translation of insights gained from in vitro tumor microenvironments to systematic investigation and understanding in clinical practice. State-of-art tumor-on-a-chip strategies will be reviewed herein, and emerging real-time sensing and monitoring platforms for on-chip analysis of tumor microenvironment will also be examined. The integration of the sensors with tumor-on-a-chip platforms to provide spatiotemporal information of the tumor microenvironment and the associated challenges will be further evaluated. Though optimal integrated systems for in situ monitoring are still in evolution, great promises lie ahead that will open new paradigm for rapid, comprehensive analysis of cancer development and assist clinicians with powerful tools to guide the diagnosis, prognosis and treatment course in cancer.
Collapse
Affiliation(s)
- Lang Zhou
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Lunan Liu
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Muammar Ali Chang
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Pengyu Chen
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, 36849, USA.
| |
Collapse
|
44
|
Gkini V, Namba T. Glutaminolysis and the Control of Neural Progenitors in Neocortical Development and Evolution. Neuroscientist 2023; 29:177-189. [PMID: 35057642 PMCID: PMC10018057 DOI: 10.1177/10738584211069060] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Multiple types of neural progenitor cells (NPCs) contribute to the development of the neocortex, a brain region responsible for our higher cognitive abilities. Proliferative capacity of NPCs varies among NPC types, developmental stages, and species. The higher proliferative capacity of NPCs in the developing human neocortex is thought to be a major contributing factor why humans have the most expanded neocortex within primates. Recent studies have shed light on the importance of cell metabolism in the neocortical NPC proliferative capacity. Specifically, glutaminolysis, a metabolic pathway that converts glutamine to glutamate and then to α-ketoglutarate, has been shown to play a critical role in human NPCs, both in apical and basal progenitors. In this review, we summarize our current knowledge of NPC metabolism, focusing especially on glutaminolysis, and discuss the role of NPC metabolism in neocortical development, evolution, and neurodevelopmental disorders, providing a broader perspective on a newly emerging research field.
Collapse
Affiliation(s)
- Vasiliki Gkini
- Neuroscience Center, HiLIFE—Helsinki
Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Takashi Namba
- Neuroscience Center, HiLIFE—Helsinki
Institute of Life Science, University of Helsinki, Helsinki, Finland
- Takashi Namba, Neuroscience Center, HiLIFE
— Helsinki Institute of Life Science, University of Helsinki, PO 63,
Haartmaninkatu 8, Helsinki 00014, Finland.
| |
Collapse
|
45
|
El Harane S, Zidi B, El Harane N, Krause KH, Matthes T, Preynat-Seauve O. Cancer Spheroids and Organoids as Novel Tools for Research and Therapy: State of the Art and Challenges to Guide Precision Medicine. Cells 2023; 12:cells12071001. [PMID: 37048073 PMCID: PMC10093533 DOI: 10.3390/cells12071001] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 04/14/2023] Open
Abstract
Spheroids and organoids are important novel players in medical and life science research. They are gradually replacing two-dimensional (2D) cell cultures. Indeed, three-dimensional (3D) cultures are closer to the in vivo reality and open promising perspectives for academic research, drug screening, and personalized medicine. A large variety of cells and tissues, including tumor cells, can be the starting material for the generation of 3D cultures, including primary tissues, stem cells, or cell lines. A panoply of methods has been developed to generate 3D structures, including spontaneous or forced cell aggregation, air-liquid interface conditions, low cell attachment supports, magnetic levitation, and scaffold-based technologies. The choice of the most appropriate method depends on (i) the origin of the tissue, (ii) the presence or absence of a disease, and (iii) the intended application. This review summarizes methods and approaches for the generation of cancer spheroids and organoids, including their advantages and limitations. We also highlight some of the challenges and unresolved issues in the field of cancer spheroids and organoids, and discuss possible therapeutic applications.
Collapse
Affiliation(s)
- Sanae El Harane
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Bochra Zidi
- Department of Medicine, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Nadia El Harane
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Thomas Matthes
- Department of Medicine, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Olivier Preynat-Seauve
- Department of Medicine, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
- Laboratory of Experimental Cell Therapy, Department of Diagnostics, Geneva University Hospitals, 1206 Geneva, Switzerland
| |
Collapse
|
46
|
Monteduro AG, Rizzato S, Caragnano G, Trapani A, Giannelli G, Maruccio G. Organs-on-chips technologies – A guide from disease models to opportunities for drug development. Biosens Bioelectron 2023; 231:115271. [PMID: 37060819 DOI: 10.1016/j.bios.2023.115271] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 11/24/2022] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
Current in-vitro 2D cultures and animal models present severe limitations in recapitulating human physiopathology with striking discrepancies in estimating drug efficacy and side effects when compared to human trials. For these reasons, microphysiological systems, organ-on-chip and multiorgans microdevices attracted considerable attention as novel tools for high-throughput and high-content research to achieve an improved understanding of diseases and to accelerate the drug development process towards more precise and eventually personalized standards. This review takes the form of a guide on this fast-growing field, providing useful introduction to major themes and indications for further readings. We start analyzing Organs-on-chips (OOC) technologies for testing the major drug administration routes: (1) oral/rectal route by intestine-on-a-chip, (2) inhalation by lung-on-a-chip, (3) transdermal by skin-on-a-chip and (4) intravenous through vascularization models, considering how drugs penetrate in the bloodstream and are conveyed to their targets. Then, we focus on OOC models for (other) specific organs and diseases: (1) neurodegenerative diseases with brain models and blood brain barriers, (2) tumor models including their vascularization, organoids/spheroids, engineering and screening of antitumor drugs, (3) liver/kidney on chips and multiorgan models for gastrointestinal diseases and metabolic assessment of drugs and (4) biomechanical systems recapitulating heart, muscles and bones structures and related diseases. Successively, we discuss technologies and materials for organ on chips, analyzing (1) microfluidic tools for organs-on-chips, (2) sensor integration for real-time monitoring, (3) materials and (4) cell lines for organs on chips. (Nano)delivery approaches for therapeutics and their on chip assessment are also described. Finally, we conclude with a critical discussion on current significance/relevance, trends, limitations, challenges and future prospects in terms of revolutionary impact on biomedical research, preclinical models and drug development.
Collapse
Affiliation(s)
- Anna Grazia Monteduro
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Silvia Rizzato
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Giusi Caragnano
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Adriana Trapani
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Gianluigi Giannelli
- National Institute of Gastroenterology IRCCS "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy
| | - Giuseppe Maruccio
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy.
| |
Collapse
|
47
|
Cao UMN, Zhang Y, Chen J, Sayson D, Pillai S, Tran SD. Microfluidic Organ-on-A-chip: A Guide to Biomaterial Choice and Fabrication. Int J Mol Sci 2023; 24:3232. [PMID: 36834645 PMCID: PMC9966054 DOI: 10.3390/ijms24043232] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Organ-on-A-chip (OoAC) devices are miniaturized, functional, in vitro constructs that aim to recapitulate the in vivo physiology of an organ using different cell types and extracellular matrix, while maintaining the chemical and mechanical properties of the surrounding microenvironments. From an end-point perspective, the success of a microfluidic OoAC relies mainly on the type of biomaterial and the fabrication strategy employed. Certain biomaterials, such as PDMS (polydimethylsiloxane), are preferred over others due to their ease of fabrication and proven success in modelling complex organ systems. However, the inherent nature of human microtissues to respond differently to surrounding stimulations has led to the combination of biomaterials ranging from simple PDMS chips to 3D-printed polymers coated with natural and synthetic materials, including hydrogels. In addition, recent advances in 3D printing and bioprinting techniques have led to the powerful combination of utilizing these materials to develop microfluidic OoAC devices. In this narrative review, we evaluate the different materials used to fabricate microfluidic OoAC devices while outlining their pros and cons in different organ systems. A note on combining the advances made in additive manufacturing (AM) techniques for the microfabrication of these complex systems is also discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Simon D. Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada
| |
Collapse
|
48
|
Goldrick C, Guri I, Herrera-Oropeza G, O’Brien-Gore C, Roy E, Wojtynska M, Spagnoli FM. 3D multicellular systems in disease modelling: From organoids to organ-on-chip. Front Cell Dev Biol 2023; 11:1083175. [PMID: 36819106 PMCID: PMC9933985 DOI: 10.3389/fcell.2023.1083175] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Cell-cell interactions underlay organ formation and function during homeostasis. Changes in communication between cells and their surrounding microenvironment are a feature of numerous human diseases, including metabolic disease and neurological disorders. In the past decade, cross-disciplinary research has been conducted to engineer novel synthetic multicellular organ systems in 3D, including organoids, assembloids, and organ-on-chip models. These model systems, composed of distinct cell types, satisfy the need for a better understanding of complex biological interactions and mechanisms underpinning diseases. In this review, we discuss the emerging field of building 3D multicellular systems and their application for modelling the cellular interactions at play in diseases. We report recent experimental and computational approaches for capturing cell-cell interactions as well as progress in bioengineering approaches for recapitulating these complexities ex vivo. Finally, we explore the value of developing such multicellular systems for modelling metabolic, intestinal, and neurological disorders as major examples of multisystemic diseases, we discuss the advantages and disadvantages of the different approaches and provide some recommendations for further advancing the field.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Francesca M. Spagnoli
- Faculty of Life Sciences, Centre for Gene Therapy and Regenerative Medicine, Guy’s Campus, King’s College London, London, United Kingdom
| |
Collapse
|
49
|
Yeap YJ, Teddy TJW, Lee MJ, Goh M, Lim KL. From 2D to 3D: Development of Monolayer Dopaminergic Neuronal and Midbrain Organoid Cultures for Parkinson's Disease Modeling and Regenerative Therapy. Int J Mol Sci 2023; 24:ijms24032523. [PMID: 36768843 PMCID: PMC9917335 DOI: 10.3390/ijms24032523] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Parkinson's Disease (PD) is a prevalent neurodegenerative disorder that is characterized pathologically by the loss of A9-specific dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) of the midbrain. Despite intensive research, the etiology of PD is currently unresolved, and the disease remains incurable. This, in part, is due to the lack of an experimental disease model that could faithfully recapitulate the features of human PD. However, the recent advent of induced pluripotent stem cell (iPSC) technology has allowed PD models to be created from patient-derived cells. Indeed, DA neurons from PD patients are now routinely established in many laboratories as monolayers as well as 3D organoid cultures that serve as useful toolboxes for understanding the mechanism underlying PD and also for drug discovery. At the same time, the iPSC technology also provides unprecedented opportunity for autologous cell-based therapy for the PD patient to be performed using the patient's own cells as starting materials. In this review, we provide an update on the molecular processes underpinning the development and differentiation of human pluripotent stem cells (PSCs) into midbrain DA neurons in both 2D and 3D cultures, as well as the latest advancements in using these cells for drug discovery and regenerative medicine. For the novice entering the field, the cornucopia of differentiation protocols reported for the generation of midbrain DA neurons may seem daunting. Here, we have distilled the essence of the different approaches and summarized the main factors driving DA neuronal differentiation, with the view to provide a useful guide to newcomers who are interested in developing iPSC-based models of PD.
Collapse
Affiliation(s)
- Yee Jie Yeap
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Tng J. W. Teddy
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Interdisciplinary Graduate Programme (IGP-Neuroscience), Nanyang Technological University, Singapore 639798, Singapore
| | - Mok Jung Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Micaela Goh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Kah Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- National Neuroscience Institute, Singapore 308433, Singapore
- Department of Brain Sciences, Imperial College London, London SW7 2AZ, UK
- Department of Anatomy, Shanxi Medical University, Taiyuan 030001, China
- Correspondence:
| |
Collapse
|
50
|
Abstract
Oral and maxillofacial organoids, as three-dimensional study models of organs, have attracted increasing attention in tissue regeneration and disease modeling. However, traditional strategies for organoid construction still fail to precisely recapitulate the key characteristics of real organs, due to the difficulty in controlling the self-organization of cells in vitro. This review aims to summarize the recent progress of novel approaches to engineering oral and maxillofacial organoids. First, we introduced the necessary components and their roles in forming oral and maxillofacial organoids. Besides, we discussed cutting-edge technology in advancing the architecture and function of organoids, especially focusing on oral and maxillofacial tissue regeneration via novel strategy with designed cell-signal scaffold compounds. Finally, current limitations and future prospects of oral and maxillofacial organoids were represented to provide guidance for further disciplinary progression and clinical application to achieve organ regeneration.
Collapse
Affiliation(s)
- Yu Wang
- Department of Implantology, School & Hospital of Stomatology, Tongji University Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200040, China
| | - Yao Sun
- Department of Implantology, School & Hospital of Stomatology, Tongji University Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200040, China
| |
Collapse
|