1
|
Honarpisheh M, Lei Y, Follenzi A, Cucci A, Olgasi C, Berishvili E, Lebreton F, Bellofatto K, Piemonti L, Citro A, Campo F, Pignatelli C, Thaunat O, Kemter E, Kraetzl M, Wolf E, Seissler J, Wolf-van Buerck L. Spheroids Composed of Reaggregated Neonatal Porcine Islets and Human Endothelial Cells Accelerate Development of Normoglycemia in Diabetic Mice. Cells 2025; 14:366. [PMID: 40072094 PMCID: PMC11898817 DOI: 10.3390/cells14050366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/13/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025] Open
Abstract
The engraftment of transplanted islets depends on the rapid establishment of a novel vascular network. The present study evaluated the effects of cord blood-derived blood outgrowth endothelial cells (BOECs) on the viability of neonatal porcine islets (NPIs) and the post-transplant outcome of grafted NPIs. Dispersed NPIs and human BOECs were reaggregated on microwell cell culture plates and tested for their anti-apoptotic and pro-angiogenic capacity by qRT-PCR and immunohistochemistry. The in vivo functionality was analyzed after transplantation into diabetic NOD-SCID IL2rγ-/- (NSG) mice. The spheroids, which contained reaggregated neonatal porcine islet cells (REPIs) and BOECs, exhibited enhanced viability and a significantly elevated gene expression of VEGFA, angiopoetin-1, heme oxygenase-1, and TNFAIP3 (A20) in vitro. The development of normoglycemia was significantly faster in animals transplanted with spheroids in comparison to the only REPI group (median 51.5 days versus 60 days) (p < 0.05). Furthermore, intragraft vascular density was substantially increased (p < 0.01). The co-transplantation of prevascularized REPI-BOEC spheroids resulted in superior angiogenesis and accelerated in vivo function. These findings may provide a novel tool to enhance the efficacy of porcine islet xenotransplantation.
Collapse
Affiliation(s)
- Mohsen Honarpisheh
- Medizinische Klinik und Poliklinik IV, Diabetes Zentrum, Klinikum der Universität München, LMU Munich, 80336 Munich, Germany; (M.H.); (Y.L.); (J.S.)
| | - Yutian Lei
- Medizinische Klinik und Poliklinik IV, Diabetes Zentrum, Klinikum der Universität München, LMU Munich, 80336 Munich, Germany; (M.H.); (Y.L.); (J.S.)
| | - Antonia Follenzi
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (A.F.); (A.C.)
| | - Alessia Cucci
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (A.F.); (A.C.)
| | - Cristina Olgasi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Ekaterine Berishvili
- Tissue Engineering and Organ Regeneration Lab, University of Geneva, Department of Surgery, CH-1211 Geneva, Switzerland; (E.B.); (F.L.); (K.B.)
| | - Fanny Lebreton
- Tissue Engineering and Organ Regeneration Lab, University of Geneva, Department of Surgery, CH-1211 Geneva, Switzerland; (E.B.); (F.L.); (K.B.)
| | - Kevin Bellofatto
- Tissue Engineering and Organ Regeneration Lab, University of Geneva, Department of Surgery, CH-1211 Geneva, Switzerland; (E.B.); (F.L.); (K.B.)
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (L.P.); (A.C.); (C.P.)
- Department of Endocrinology, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Antonio Citro
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (L.P.); (A.C.); (C.P.)
| | - Francesco Campo
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (L.P.); (A.C.); (C.P.)
- Department of Endocrinology, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Cataldo Pignatelli
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (L.P.); (A.C.); (C.P.)
| | - Olivier Thaunat
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon I, 69364 Lyon, France;
- Department of Nephrology Transplantation and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, 69003 Lyon, France
| | - Elisabeth Kemter
- Molecular Animal Breeding and Biotechnology, Gene Centre and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (E.K.); (E.W.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Martin Kraetzl
- Molecular Animal Breeding and Biotechnology, Gene Centre and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (E.K.); (E.W.)
| | - Eckhard Wolf
- Molecular Animal Breeding and Biotechnology, Gene Centre and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (E.K.); (E.W.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Jochen Seissler
- Medizinische Klinik und Poliklinik IV, Diabetes Zentrum, Klinikum der Universität München, LMU Munich, 80336 Munich, Germany; (M.H.); (Y.L.); (J.S.)
| | - Lelia Wolf-van Buerck
- Medizinische Klinik und Poliklinik IV, Diabetes Zentrum, Klinikum der Universität München, LMU Munich, 80336 Munich, Germany; (M.H.); (Y.L.); (J.S.)
| | | |
Collapse
|
2
|
Sun Y, Liu J, Zhu L, Huang F, Dong Y, Liu S, Chen S, Ji W, Lu J, Liu L, Li S. Treatment Response to Oncolytic Virus in Patient-Derived Breast Cancer and Hypopharyngeal Cancer Organoids: Evaluation via a Microfluidics Organ-on-a-Chip System. Bioengineering (Basel) 2025; 12:146. [PMID: 40001666 PMCID: PMC11851931 DOI: 10.3390/bioengineering12020146] [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: 12/17/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
In this study, we present an oncolytic virus (OV) evaluation system established using microfluidic organ-on-a-chip (OOC) systems and patient-derived organoids (PDOs), which was used in the development of a novel oncolytic virus, AD4-GHPE. An OV offers advantages such as good targeting ability and minimal side effects, and it has achieved significant breakthroughs when combined with immunotherapy in recent clinical trials. The development of OVs has become an emerging research focus. PDOs can preserve the heterogeneity of in situ tumor tissues, whereas microfluidic OOC systems can automate and standardize various experimental procedures. These systems have been applied in cutting-edge drug screening and cell therapy experiments; however, their use in functionally complex oncolytic viruses remains to be explored. In this study, we constructed a novel recombinant oncolytic adenovirus, AD4-GHPE, and evaluated OOC systems and PDOs through various functional validations in hypopharyngeal and breast cancer organoids. The results confirmed that AD4-GHPE exhibits three antitumor mechanisms, namely, tumor-specific cytotoxicity, a reduction in programmed death ligand 1 (PD-L1) expression in tumor cells to increase CD8+ T-cell activity, and granulocyte-macrophage colony-stimulating factor (GM-CSF) secretion. The evaluation system combining OOC systems and PDOs was efficient and reliable, providing personalized OV treatment recommendations for patients and offering industrialized and standardized research ideas for the development of OVs.
Collapse
Affiliation(s)
- Yu Sun
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Jiaqi Liu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Li Zhu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Fang Huang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Yanbo Dong
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
| | - Shuang Liu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Siyi Chen
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
- Medical School of Chinese PLA, Beijing 100853, China
| | - Wei Ji
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Jingjing Lu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Liangfa Liu
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
| | - Shanhu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| |
Collapse
|
3
|
Martinelli C, Bocconi A, Milone S, Baldissera T, Cherubin L, Buccioli G, Perottoni S, Conci C, Cerullo G, Osellame R, Chirico G, Jacchetti E, Raimondi MT. A 3D millifluidic model of a dermal perivascular microenvironment on a chip. LAB ON A CHIP 2025; 25:423-439. [PMID: 39757968 PMCID: PMC11701800 DOI: 10.1039/d4lc00898g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025]
Abstract
The process of angiogenesis plays a pivotal role in skin regeneration, ensuring the provision of nutrients and oxygen to the nascent tissue, thanks to the formation of novel microvascular networks supporting functional tissue regeneration. Unfortunately, most of the current therapeutic approaches for skin regeneration lack vascularization, required to promote effective angiogenesis. Thus, in vitro tridimensional models, complemented with specific biochemical signals, can be a valuable tool to unravel the neovascularization mechanisms and develop novel clinical strategies. In this work, we designed and validated a tridimensional microstructured dynamic model of the dermal perivascular microenvironment on a chip. We carried out the fabrication of an array of microstructures by two-photon laser polymerization, then used as a 3D substrate for co-culture of human dermal fibroblasts and endothelial cells. We included the substrate in a miniaturized optically accessible bioreactor (MOAB) which provides the physiological interstitial flow, upon perfusion in the presence or absence of the pro-angiogenic stimuli VEGF and TGF-β1. We determined the parameters to be applied under dynamic conditions by an in silico model simulating individual 3D microenvironments within the bioreactor's chambers. We computed the fluid velocity and wall shear stress acting on endothelial cells along with the oxygen concentration profile, and we chose the most suitable flow rate for maintaining dermal physiological conditions. Experimental results showed the effectiveness of the developed platform as a 3D dynamic model of angiogenesis. This is the first combined experimental and computational study involving chemically stimulated 3D co-cultures for successfully simulating the physiological dermal perivascular microenvironment.
Collapse
Affiliation(s)
- Chiara Martinelli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Alberto Bocconi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Sofia Milone
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Teresa Baldissera
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Leonardo Cherubin
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Giovanni Buccioli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Simone Perottoni
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Claudio Conci
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Giulio Cerullo
- Institute for Photonics and Nanotechnologies (IFN), CNR and Department of Physics, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy.
| | - Roberto Osellame
- Institute for Photonics and Nanotechnologies (IFN), CNR and Department of Physics, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy.
| | - Giuseppe Chirico
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza, 3, 20126 Milan, Italy.
| | - Emanuela Jacchetti
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Piazza L. da Vinci, 32, 20133 Milan, Italy.
| |
Collapse
|
4
|
Wu Y, Li X, Liu H, Yang X, Li R, Zhao H, Shang Z. Organoids in the oral and maxillofacial region: present and future. Int J Oral Sci 2024; 16:61. [PMID: 39482304 PMCID: PMC11528035 DOI: 10.1038/s41368-024-00324-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: 12/19/2023] [Revised: 06/18/2024] [Accepted: 09/12/2024] [Indexed: 11/03/2024] Open
Abstract
The oral and maxillofacial region comprises a variety of organs made up of multiple soft and hard tissue, which are anatomically vulnerable to the pathogenic factors of trauma, inflammation, and cancer. The studies of this intricate entity have been long-termly challenged by a lack of versatile preclinical models. Recently, the advancements in the organoid industry have provided novel strategies to break through this dilemma. Here, we summarize the existing biological and engineering approaches that were employed to generate oral and maxillofacial organoids. Then, we detail the use of modified co-culture methods, such as cell cluster co-inoculation and air-liquid interface culture technology to reconstitute the vascular network and immune microenvironment in assembled organoids. We further retrospect the existing oral and maxillofacial assembled organoids and their potential to recapitulate the homeostasis in parental tissues such as tooth, salivary gland, and mucosa. Finally, we discuss how the next-generation organoids may benefit to regenerative and precision medicine for treatment of oral-maxillofacial illness.
Collapse
Affiliation(s)
- Yufei Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xiang Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hanzhe Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xiao Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Rui Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hui Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
5
|
Polak M, Karbowniczek JE, Stachewicz U. Strategies in Electrospun Polymer and Hybrid Scaffolds for Enhanced Cell Integration and Vascularization for Bone Tissue Engineering and Organoids. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2022. [PMID: 39696966 DOI: 10.1002/wnan.2022] [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: 07/13/2024] [Revised: 11/05/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024]
Abstract
Addressing the demand for bone substitutes, tissue engineering responds to the high prevalence of orthopedic surgeries worldwide and the limitations of conventional tissue reconstruction techniques. Materials, cells, and growth factors constitute the core elements in bone tissue engineering, influencing cellular behavior crucial for regenerative treatments. Scaffold design, including architectural features and porosity, significantly impacts cellular penetration, proliferation, differentiation, and vascularization. This review discusses the hierarchical structure of bone and the process of neovascularization in the context of biofabrication of scaffolds. We focus on the role of electrospinning and its modifications in scaffold fabrication to improve scaffold properties to enhance further tissue regeneration, for example, by boosting oxygen and nutrient delivery. We highlight how scaffold design impacts osteogenesis and the overall success of regenerative treatments by mimicking the extracellular matrix (ECM). Additionally, we explore the emerging field of bone organoids-self-assembled, three-dimensional (3D) structures derived from stem cells that replicate native bone tissue's architecture and functionality. While bone organoids hold immense potential for modeling bone diseases and facilitating regenerative treatments, their main limitation remains insufficient vascularization. Hence, we evaluate innovative strategies for pre-vascularization and discuss the latest techniques for assessing and improving vascularization in both scaffolds and organoids presenting the most commonly used cell lines and biological models. Moreover, we analyze cutting-edge techniques for assessing vascularization, evaluating their advantages and drawbacks to propose complex solutions. Finally, by integrating these approaches, we aim to advance the development of bioactive materials that promote successful bone regeneration.
Collapse
Affiliation(s)
- Martyna Polak
- Faculty of Metals Engineering and Industrial Computer Science, AGH University of Krakow, Kraków, Poland
| | - Joanna Ewa Karbowniczek
- Faculty of Metals Engineering and Industrial Computer Science, AGH University of Krakow, Kraków, Poland
| | - Urszula Stachewicz
- Faculty of Metals Engineering and Industrial Computer Science, AGH University of Krakow, Kraków, Poland
| |
Collapse
|
6
|
Werschler N, Quintard C, Nguyen S, Penninger J. Engineering next generation vascularized organoids. Atherosclerosis 2024; 398:118529. [PMID: 39304390 DOI: 10.1016/j.atherosclerosis.2024.118529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/31/2024] [Accepted: 06/21/2024] [Indexed: 09/22/2024]
Abstract
Organoids are self-organizing 3D cell culture models that are valuable for studying the mechanisms underlying both development and disease in multiple species, particularly, in humans. These 3D engineered tissues can mimic the structure and function of human organs in vitro. Methods to generate organoids have substantially improved to better resemble, in various ways, their in vivo counterpart. One of the major limitations in current organoid models is the lack of a functional vascular compartment. Here we discuss methodological approaches to generating perfusable blood vessel networks in organoid systems. Inclusion of perfused vascular compartments markedly enhances the physiological relevance of organoid systems and is a critical step in the establishment of next generation, higher-complexity in vitro systems for use in developmental, clinical, and drug-development settings.
Collapse
Affiliation(s)
- Nicolas Werschler
- University of British Columbia, Life Sciences Institute, Vancouver, Canada; University of British Columbia, School of Biomedical Engineering, Vancouver, Canada.
| | - Clement Quintard
- University of British Columbia, Life Sciences Institute, Vancouver, Canada; University of British Columbia, Medical Genetics, Vancouver, Canada
| | - Stephanie Nguyen
- University of British Columbia, School of Biomedical Engineering, Vancouver, Canada
| | - Josef Penninger
- University of British Columbia, Life Sciences Institute, Vancouver, Canada; University of British Columbia, School of Biomedical Engineering, Vancouver, Canada; University of British Columbia, Medical Genetics, Vancouver, Canada; Helmholtz Centre for Infection Research, Germany; Eric Kandel Institute, Department of Laboratory Medicine, Medical University of Vienna, Austria; IMBA Institute of Molecular Biotechnology, Vienna, Austria
| |
Collapse
|
7
|
Yrjänäinen A, Mesiä E, Lampela E, Kreutzer J, Vihinen J, Tornberg K, Vuorenpää H, Miettinen S, Kallio P, Mäki AJ. Barrier-free, open-top microfluidic chip for generating two distinct, interconnected 3D microvascular networks. Sci Rep 2024; 14:22916. [PMID: 39358415 PMCID: PMC11447027 DOI: 10.1038/s41598-024-74493-3] [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: 05/27/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024] Open
Abstract
Developing microphysiological cell culture platforms with a three-dimensional (3D) microenvironment has been a significant advancement from traditional monolayer cultures. Still, most of the current microphysiological platforms are limited in closed designs, i.e. are not accessible after 3D cell culture loading. Here, we report an open-top microfluidic chip which enables the generation of two sequentially loaded 3D cell cultures without physical barriers restricting the nurture, gas exchange and cellular communication. As a proof-of-concept, we demonstrated the formation of two 3D vasculatures, one in the upper and the other in the lower compartment, under three distinct flow conditions: asymmetric side-to-center, symmetric side-to-center and symmetric center-to-side. We used computational modelling to characterize initial flow pressures in cell culture compartments. We showed prominent vessel formation and branched vasculatures in upper and lower cell culture compartments with interconnecting, lumenized vessels with in vivo-relevant diameter in all flow conditions. With advanced image processing, we quantified and compared the overall vascular network volume and the total length formed in asymmetric side-to-center, symmetric side-to-center and symmetric center-to-side flow conditions. Our results indicate that the developed chip can house two distinct 3D cell cultures with merging vessels between compartments and by providing asymmetric side-to-center or symmetric center-to-side flow vascular morphogenesis is enhanced in terms of overall network length. The developed open-top microfluidic chip may find various applications in generation of tissue-specific 3D-3D co-cultures for studying cellular interactions in vascularized tissues and organs.
Collapse
Grants
- 9AB043, 9AC057 Wellbeing services county of Pirkanmaa
- 9AB043, 9AC057 Wellbeing services county of Pirkanmaa
- 9AB043, 9AC057 Wellbeing services county of Pirkanmaa
- 9AB043, 9AC057 Wellbeing services county of Pirkanmaa
- 9AB043, 9AC057 Wellbeing services county of Pirkanmaa
- 9AB043, 9AC057 Wellbeing services county of Pirkanmaa
- 9AB043, 9AC057 Wellbeing services county of Pirkanmaa
- 9AB043, 9AC057 Wellbeing services county of Pirkanmaa
- 9AB043, 9AC057 Wellbeing services county of Pirkanmaa
- 9AB043, 9AC057 Wellbeing services county of Pirkanmaa
Collapse
Affiliation(s)
- Alma Yrjänäinen
- Adult Stem Cell Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Pirkanmaa, Finland.
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Pirkanmaa, Finland.
| | - Elina Mesiä
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Pirkanmaa, Finland
| | - Ella Lampela
- Adult Stem Cell Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Pirkanmaa, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Pirkanmaa, Finland
| | - Joose Kreutzer
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Pirkanmaa, Finland
| | - Jorma Vihinen
- Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Pirkanmaa, Finland
| | - Kaisa Tornberg
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Pirkanmaa, Finland
| | - Hanna Vuorenpää
- Adult Stem Cell Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Pirkanmaa, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Pirkanmaa, Finland
| | - Susanna Miettinen
- Adult Stem Cell Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Pirkanmaa, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Pirkanmaa, Finland
| | - Pasi Kallio
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Pirkanmaa, Finland
| | - Antti-Juhana Mäki
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Pirkanmaa, Finland
| |
Collapse
|
8
|
Wang L, Wan J, Xu Y, Huang Y, Wang D, Zhu D, Chen Q, Lu Y, Guo Q. Endothelial Cells Promote Pseudo-islet Function Through BTC-EGFR-JAK/STAT Signaling Pathways. Ann Biomed Eng 2024; 52:2610-2626. [PMID: 38829457 DOI: 10.1007/s10439-024-03548-3] [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: 09/20/2023] [Accepted: 05/20/2024] [Indexed: 06/05/2024]
Abstract
Interactions between cells are of fundamental importance in affecting cell function. In vivo, endothelial cells and islet cells are close to each other, which makes endothelial cells essential for islet cell development and maintenance of islet cell function. We used endothelial cells to construct 3D pseudo-islets, which demonstrated better glucose regulation and greater insulin secretion compared to conventional pseudo-islets in both in vivo and in vitro trials. However, the underlying mechanism of how endothelial cells promote beta cell function localized within islets is still unknown. We performed transcriptomic sequencing, differential gene analysis, and enrichment analysis on two types of pseudo-islets to show that endothelial cells can promote the function of internal beta cells in pseudo-islets through the BTC-EGFR-JAK/STAT signaling pathway. Min6 cells secreted additional BTC after co-culture of endothelial cells with MIN6 cells outside the body. After BTC knockout in vitro, we found that beta cells functioned differently: insulin secretion levels decreased significantly, while the expression of key proteins in the EGFR-mediated JAK/STAT signaling pathway simultaneously decreased, further confirming our results. Through our experiments, we elucidate the molecular mechanisms by which endothelial cells maintain islet function in vitro, which provides a theoretical basis for the construction of pseudo-islets and islet cell transplants for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Lin Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Jian Wan
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Yang Xu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Dongzhi Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Donghui Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Qiyang Chen
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Yuhua Lu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Qingsong Guo
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
9
|
Fritschen A, Lindner N, Scholpp S, Richthof P, Dietz J, Linke P, Guttenberg Z, Blaeser A. High-Scale 3D-Bioprinting Platform for the Automated Production of Vascularized Organs-on-a-Chip. Adv Healthc Mater 2024; 13:e2304028. [PMID: 38511587 PMCID: PMC11469029 DOI: 10.1002/adhm.202304028] [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: 11/16/2023] [Revised: 03/18/2024] [Indexed: 03/22/2024]
Abstract
3D bioprinting possesses the potential to revolutionize contemporary methodologies for fabricating tissue models employed in pharmaceutical research and experimental investigations. This is enhanced by combining bioprinting with advanced organs-on-a-chip (OOCs), which includes a complex arrangement of multiple cell types representing organ-specific cells, connective tissue, and vasculature. However, both OOCs and bioprinting so far demand a high degree of manual intervention, thereby impeding efficiency and inhibiting scalability to meet technological requirements. Through the combination of drop-on-demand bioprinting with robotic handling of microfluidic chips, a print procedure is achieved that is proficient in managing three distinct tissue models on a chip within only a minute, as well as capable of consecutively processing numerous OOCs without manual intervention. This process rests upon the development of a post-printing sealable microfluidic chip, that is compatible with different types of 3D-bioprinters and easily connected to a perfusion system. The capabilities of the automized bioprint process are showcased through the creation of a multicellular and vascularized liver carcinoma model on the chip. The process achieves full vascularization and stable microvascular network formation over 14 days of culture time, with pronounced spheroidal cell growth and albumin secretion of HepG2 serving as a representative cell model.
Collapse
Affiliation(s)
- Anna Fritschen
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Nils Lindner
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Sebastian Scholpp
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Philipp Richthof
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Jonas Dietz
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | | | | | - Andreas Blaeser
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
- Centre for Synthetic BiologyTechnical University of Darmstadt64289DarmstadtGermany
| |
Collapse
|
10
|
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
|
11
|
Quintard C, Tubbs E, Jonsson G, Jiao J, Wang J, Werschler N, Laporte C, Pitaval A, Bah TS, Pomeranz G, Bissardon C, Kaal J, Leopoldi A, Long DA, Blandin P, Achard JL, Battail C, Hagelkruys A, Navarro F, Fouillet Y, Penninger JM, Gidrol X. A microfluidic platform integrating functional vascularized organoids-on-chip. Nat Commun 2024; 15:1452. [PMID: 38365780 PMCID: PMC10873332 DOI: 10.1038/s41467-024-45710-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 02/02/2024] [Indexed: 02/18/2024] Open
Abstract
The development of vascular networks in microfluidic chips is crucial for the long-term culture of three-dimensional cell aggregates such as spheroids, organoids, tumoroids, or tissue explants. Despite rapid advancement in microvascular network systems and organoid technologies, vascularizing organoids-on-chips remains a challenge in tissue engineering. Most existing microfluidic devices poorly reflect the complexity of in vivo flows and require complex technical set-ups. Considering these constraints, we develop a platform to establish and monitor the formation of endothelial networks around mesenchymal and pancreatic islet spheroids, as well as blood vessel organoids generated from pluripotent stem cells, cultured for up to 30 days on-chip. We show that these networks establish functional connections with the endothelium-rich spheroids and vascular organoids, as they successfully provide intravascular perfusion to these structures. We find that organoid growth, maturation, and function are enhanced when cultured on-chip using our vascularization method. This microphysiological system represents a viable organ-on-chip model to vascularize diverse biological 3D tissues and sets the stage to establish organoid perfusions using advanced microfluidics.
Collapse
Affiliation(s)
- Clément Quintard
- Univ. Grenoble Alpes, CEA, IRIG/BGE, BIOMICS, 38000, Grenoble, France
- Univ. Grenoble Alpes, CEA, LETI, DTBS, 38000, Grenoble, France
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia (BC), Canada
| | - Emily Tubbs
- Univ. Grenoble Alpes, CEA, IRIG/BGE, BIOMICS, 38000, Grenoble, France
| | - Gustav Jonsson
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, IMBA, Dr. Bohr-Gasse 3, 1030, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, 1030, Vienna, Austria
- Eric Kandel Institute, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Jie Jiao
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia (BC), Canada
| | - Jun Wang
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia (BC), Canada
| | - Nicolas Werschler
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia (BC), Canada
| | - Camille Laporte
- Univ. Grenoble Alpes, CEA, IRIG/BGE, BIOMICS, 38000, Grenoble, France
- Univ. Grenoble Alpes, CEA, LETI, DTBS, 38000, Grenoble, France
| | - Amandine Pitaval
- Univ. Grenoble Alpes, CEA, IRIG/BGE, BIOMICS, 38000, Grenoble, France
| | - Thierno-Sidy Bah
- Univ. Grenoble Alpes, CEA, IRIG, BGE, Gen&Chem, 38000, Grenoble, France
| | - Gideon Pomeranz
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | | | - Joris Kaal
- Univ. Grenoble Alpes, CEA, LETI, DTBS, 38000, Grenoble, France
| | - Alexandra Leopoldi
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, IMBA, Dr. Bohr-Gasse 3, 1030, Vienna, Austria
- Eric Kandel Institute, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - David A Long
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Pierre Blandin
- Univ. Grenoble Alpes, CEA, LETI, DTBS, 38000, Grenoble, France
| | - Jean-Luc Achard
- Université Grenoble Alpes, CNRS, Grenoble INP, LEGI, 38000, Grenoble, France
| | | | - Astrid Hagelkruys
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, IMBA, Dr. Bohr-Gasse 3, 1030, Vienna, Austria
- Eric Kandel Institute, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Fabrice Navarro
- Univ. Grenoble Alpes, CEA, LETI, DTBS, 38000, Grenoble, France
| | - Yves Fouillet
- Univ. Grenoble Alpes, CEA, LETI, DTBS, 38000, Grenoble, France
| | - Josef M Penninger
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia (BC), Canada.
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, IMBA, Dr. Bohr-Gasse 3, 1030, Vienna, Austria.
- Eric Kandel Institute, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
- Helmholtz Centre for Infection Research, Braunschweig, Germany.
| | - Xavier Gidrol
- Univ. Grenoble Alpes, CEA, IRIG/BGE, BIOMICS, 38000, Grenoble, France.
| |
Collapse
|
12
|
Tronolone JJ, Mathur T, Chaftari CP, Sun Y, Jain A. Machine learning chained neural network analysis of oxygen transport amplifies the physiological relevance of vascularized microphysiological systems. Bioeng Transl Med 2023; 8:e10582. [PMID: 38023704 PMCID: PMC10658488 DOI: 10.1002/btm2.10582] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/04/2023] [Accepted: 07/14/2023] [Indexed: 12/01/2023] Open
Abstract
Since every biological system requires capillaries to support its oxygenation, design of engineered preclinical models of such systems, for example, vascularized microphysiological systems (vMPS) have gained attention enhancing the physiological relevance of human biology and therapies. But the physiology and function of formed vessels in the vMPS is currently assessed by non-standardized, user-dependent, and simple morphological metrics that poorly relate to the fundamental function of oxygenation of organs. Here, a chained neural network is engineered and trained using morphological metrics derived from a diverse set of vMPS representing random combinations of factors that influence the vascular network architecture of a tissue. This machine-learned algorithm outputs a singular measure, termed as vascular network quality index (VNQI). Cross-correlation of morphological metrics and VNQI against measured oxygen levels within vMPS revealed that VNQI correlated the most with oxygen measurements. VNQI is sensitive to the determinants of vascular networks and it consistently correlates better to the measured oxygen than morphological metrics alone. Finally, the VNQI is positively associated with the functional outcomes of cell transplantation therapies, shown in the vascularized islet-chip challenged with hypoxia. Therefore, adoption of this tool will amplify the predictions and enable standardization of organ-chips, transplant models, and other cell biosystems.
Collapse
Affiliation(s)
- James J. Tronolone
- Department of Biomedical Engineering, College of EngineeringTexas A&M UniversityCollege StationTexasUSA
| | - Tanmay Mathur
- Department of Biomedical Engineering, College of EngineeringTexas A&M UniversityCollege StationTexasUSA
| | - Christopher P. Chaftari
- Department of Biomedical Engineering, College of EngineeringTexas A&M UniversityCollege StationTexasUSA
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Abhishek Jain
- Department of Biomedical Engineering, College of EngineeringTexas A&M UniversityCollege StationTexasUSA
- Department of Medical Physiology, School of MedicineTexas A&M Health Science CenterBryanTexasUSA
- Department of Cardiovascular ScienceHouston Methodist Academic InstituteHoustonTexasUSA
| |
Collapse
|
13
|
Tronolone JJ, Mathur T, Chaftari CP, Jain A. Evaluation of the Morphological and Biological Functions of Vascularized Microphysiological Systems with Supervised Machine Learning. Ann Biomed Eng 2023; 51:1723-1737. [PMID: 36913087 DOI: 10.1007/s10439-023-03177-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/22/2023] [Indexed: 03/14/2023]
Abstract
Vascularized microphysiological systems and organoids are contemporary preclinical experimental platforms representing human tissue or organ function in health and disease. While vascularization is emerging as a necessary physiological organ-level feature required in most such systems, there is no standard tool or morphological metric to measure the performance or biological function of vascularized networks within these models. Further, the commonly reported morphological metrics may not correlate to the network's biological function-oxygen transport. Here, a large library of vascular network images was analyzed by the measure of each sample's morphology and oxygen transport potential. The oxygen transport quantification is computationally expensive and user-dependent, so machine learning techniques were examined to generate regression models relating morphology to function. Principal component and factor analyses were applied to reduce dimensionality of the multivariate dataset, followed by multiple linear regression and tree-based regression analyses. These examinations reveal that while several morphological data relate poorly to the biological function, some machine learning models possess a relatively improved, but still moderate predictive potential. Overall, random forest regression model correlates to the biological function of vascular networks with relatively higher accuracy than other regression models.
Collapse
Affiliation(s)
- James J Tronolone
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX, 77843, USA
| | - Tanmay Mathur
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX, 77843, USA
| | - Christopher P Chaftari
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX, 77843, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX, 77843, USA.
- Department of Medical Physiology, School of Medicine, Texas A&M University, Bryan, TX, USA.
- Department of Cardiovascular Sciences, Houston Methodist Academic Institute, Houston, USA.
| |
Collapse
|
14
|
Beydag-Tasöz BS, Yennek S, Grapin-Botton A. Towards a better understanding of diabetes mellitus using organoid models. Nat Rev Endocrinol 2023; 19:232-248. [PMID: 36670309 PMCID: PMC9857923 DOI: 10.1038/s41574-022-00797-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 01/22/2023]
Abstract
Our understanding of diabetes mellitus has benefited from a combination of clinical investigations and work in model organisms and cell lines. Organoid models for a wide range of tissues are emerging as an additional tool enabling the study of diabetes mellitus. The applications for organoid models include studying human pancreatic cell development, pancreatic physiology, the response of target organs to pancreatic hormones and how glucose toxicity can affect tissues such as the blood vessels, retina, kidney and nerves. Organoids can be derived from human tissue cells or pluripotent stem cells and enable the production of human cell assemblies mimicking human organs. Many organ mimics relevant to diabetes mellitus are already available, but only a few relevant studies have been performed. We discuss the models that have been developed for the pancreas, liver, kidney, nerves and vasculature, how they complement other models, and their limitations. In addition, as diabetes mellitus is a multi-organ disease, we highlight how a merger between the organoid and bioengineering fields will provide integrative models.
Collapse
Affiliation(s)
- Belin Selcen Beydag-Tasöz
- The Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen, Denmark
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Siham Yennek
- The Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen, Denmark
| | - Anne Grapin-Botton
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
- Paul Langerhans Institute Dresden, Dresden, Germany.
| |
Collapse
|
15
|
Tronolone JJ, Mathur T, Chaftari CP, Jain A. Evaluation of the morphological and biological functions of vascularized microphysiological systems with supervised machine learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523755. [PMID: 36711458 PMCID: PMC9882172 DOI: 10.1101/2023.01.12.523755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Vascularized microphysiological systems and organoids are contemporary preclinical experimental platforms representing human tissue or organ function in health and disease. While vascularization is emerging as a necessary physiological organ-level feature required in most such systems, there is no standard tool or morphological metric to measure the performance or biological function of vascularized networks within these models. Further, the commonly reported morphological metrics may not correlate to the network's biological function - oxygen transport. Here, a large library of vascular network images was analyzed by the measure of each sample's morphology and oxygen transport potential. The oxygen transport quantification is computationally expensive and user-dependent, so machine learning techniques were examined to generate regression models relating morphology to function. Principal component and factor analyses were applied to reduce dimensionality of the multivariate dataset, followed by multiple linear regression and tree-based regression analyses. These examinations reveal that while several morphological data relate poorly to the biological function, some machine learning models possess a relatively improved, but still moderate predictive potential. Overall, random forest regression model correlates to the biological function of vascular networks with relatively higher accuracy than other regression models.
Collapse
|
16
|
Microfluidic Technology for Evaluating and Preserving Islet Function for Islet Transplant in Type 1 Diabetes. CURRENT TRANSPLANTATION REPORTS 2022. [DOI: 10.1007/s40472-022-00377-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
17
|
Urbanczyk M, Zbinden A, Schenke-Layland K. Organ-specific endothelial cell heterogenicity and its impact on regenerative medicine and biomedical engineering applications. Adv Drug Deliv Rev 2022; 186:114323. [PMID: 35568103 DOI: 10.1016/j.addr.2022.114323] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/23/2022] [Accepted: 05/05/2022] [Indexed: 02/08/2023]
Abstract
Endothelial cells (ECs) are a key cellular component of the vascular system as they form the inner lining of the blood vessels. Recent findings highlight that ECs express extensive phenotypic heterogenicity when following the vascular tree from the major vasculature down to the organ capillaries. However, in vitro models, used for drug development and testing, or to study the role of ECs in health and disease, rarely acknowledge this EC heterogenicity. In this review, we highlight the main differences between different EC types, briefly summarize their different characteristics and focus on the use of ECs in in vitro models. We introduce different approaches on how ECs can be utilized in co-culture test systems in the field of brain, pancreas, and liver research to study the role of the endothelium in health and disease. Finally, we discuss potential improvements to current state-of-the-art in vitro models and future directions.
Collapse
|
18
|
Wan Z, Zhong AX, Zhang S, Pavlou G, Coughlin MF, Shelton SE, Nguyen HT, Lorch JH, Barbie DA, Kamm RD. A Robust Method for Perfusable Microvascular Network Formation In Vitro. SMALL METHODS 2022; 6:e2200143. [PMID: 35373502 PMCID: PMC9844969 DOI: 10.1002/smtd.202200143] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/03/2022] [Indexed: 05/02/2023]
Abstract
Micropost-based microfluidic devices are widely used for microvascular network (MVN) formation in diverse research fields. However, consistently generating perfusable MVNs of physiological morphology and dimension has proven to be challenging. Here, how initial seeding parameters determine key characteristics of MVN formation is investigated and a robust two-step seeding strategy to generate perfusable physiological MVNs in microfluidic devices is established.
Collapse
Affiliation(s)
- Zhengpeng Wan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Amy X Zhong
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Shun Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Georgios Pavlou
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Mark F Coughlin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Sarah E Shelton
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Huu Tuan Nguyen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Jochen H Lorch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - David A Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
19
|
Mykuliak A, Yrjänäinen A, Mäki AJ, Gebraad A, Lampela E, Kääriäinen M, Pakarinen TK, Kallio P, Miettinen S, Vuorenpää H. Vasculogenic Potency of Bone Marrow- and Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells Results in Differing Vascular Network Phenotypes in a Microfluidic Chip. Front Bioeng Biotechnol 2022; 10:764237. [PMID: 35211462 PMCID: PMC8861308 DOI: 10.3389/fbioe.2022.764237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/11/2022] [Indexed: 12/27/2022] Open
Abstract
The vasculature is an essential, physiological element in virtually all human tissues. Formation of perfusable vasculature is therefore crucial for reliable tissue modeling. Three-dimensional vascular networks can be formed through the co-culture of endothelial cells (ECs) with stromal cells embedded in hydrogel. Mesenchymal stem/stromal cells (MSCs) derived from bone marrow (BMSCs) and adipose tissue (ASCs) are an attractive choice as stromal cells due to their natural perivascular localization and ability to support formation of mature and stable microvessels in vitro. So far, BMSCs and ASCs have been compared as vasculature-supporting cells in static cultures. In this study, BMSCs and ASCs were co-cultured with endothelial cells in a fibrin hydrogel in a perfusable microfluidic chip. We demonstrated that using MSCs of different origin resulted in vascular networks with distinct phenotypes. Both types of MSCs supported formation of mature and interconnected microvascular networks-on-a-chip. However, BMSCs induced formation of fully perfusable microvasculature with larger vessel area and length whereas ASCs resulted in partially perfusable microvascular networks. Immunostainings revealed that BMSCs outperformed ASCs in pericytic characteristics. Moreover, co-culture with BMSCs resulted in significantly higher expression levels of endothelial and pericyte-specific genes, as well as genes involved in vasculature maturation. Overall, our study provides valuable knowledge on the properties of MSCs as vasculature-supporting cells and highlights the importance of choosing the application-specific stromal cell source for vascularized organotypic models.
Collapse
Affiliation(s)
- Anastasiia Mykuliak
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Alma Yrjänäinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Antti-Juhana Mäki
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Arjen Gebraad
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Ella Lampela
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Minna Kääriäinen
- Department of Plastic and Reconstructive Surgery, Tampere University Hospital, Tampere, Finland
| | | | - Pasi Kallio
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Susanna Miettinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Hanna Vuorenpää
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
20
|
Rodríguez-Comas J, Ramón-Azcón J. Islet-on-a-chip for the study of pancreatic β-cell function. IN VITRO MODELS 2022; 1:41-57. [PMID: 39872972 PMCID: PMC11749753 DOI: 10.1007/s44164-021-00005-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/25/2021] [Accepted: 11/08/2021] [Indexed: 01/30/2025]
Abstract
Diabetes mellitus is a significant public health problem worldwide. It encompasses a group of chronic disorders characterized by hyperglycemia, resulting from pancreatic islet dysfunction or as a consequence of insulin-producing β-cell death. Organ-on-a-chip platforms have emerged as technological systems combining cell biology, engineering, and biomaterial technological advances with microfluidics to recapitulate a specific organ's physiological or pathophysiological environment. These devices offer a novel model for the screening of pharmaceutical agents and to study a particular disease. In the field of diabetes, a variety of microfluidic devices have been introduced to recreate native islet microenvironments and to understand pancreatic β-cell kinetics in vitro. This kind of platforms has been shown fundamental for the study of the islet function and to assess the quality of these islets for subsequent in vivo transplantation. However, islet physiological systems are still limited compared to other organs and tissues, evidencing the difficulty to study this "organ" and the need for further technological advances. In this review, we summarize the current state of islet-on-a-chip platforms that have been developed so far. We recapitulate the most relevant studies involving pancreatic islets and microfluidics, focusing on the molecular and cellular-scale activities that underlie pancreatic β-cell function.
Collapse
Affiliation(s)
- Júlia Rodríguez-Comas
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac, 10-12, 08028 Barcelona, Spain
| | - Javier Ramón-Azcón
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac, 10-12, 08028 Barcelona, Spain
- ICREA-Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| |
Collapse
|
21
|
Papoz A, Clément F, Laporte C, Tubbs E, Gidrol X, Pitaval A. [Generating pancreatic islets organoids: Langerhanoids]. Med Sci (Paris) 2022; 38:52-58. [PMID: 35060887 DOI: 10.1051/medsci/2021244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The extension of islet transplantation to a wider number of Type 1 diabetic patients is compromised by the scarcity of donors, the reduced ex vivo survival of pancreatic islets and the use of immunosuppressive treatments. Islets of Langerhans isolated from brain-dead donors are currently the only cell source for transplantation. Thus, it is crucial to find an alternative and an abundant source of functional insulin secreting cells not only for clinical use but also for the development of research dedicated to the screening of drugs and to the development of new therapeutic targets. Several groups around the world, including ours, develop 3D culture models as Langerhanoids that closely mimick human pancreatic islets physiology. In this review, we describe recent advances to mimic the pancreatic niche (extracellular matrix, vascularization, microfluidics) allowing better functionality of Langerhanoids.
Collapse
Affiliation(s)
- Anastasia Papoz
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France
| | - Flora Clément
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France
| | - Camille Laporte
- Univ. Grenoble Alpes, CEA, Leti, Division for biology and healthcare technologies, Microfluidic systems and bioengineering Lab, F-38000, Grenoble, France
| | - Emily Tubbs
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France - Univ. Grenoble Alpes, LBFA et BEeSy, Inserm U1055, F-38000, Grenoble, France
| | - Xavier Gidrol
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France
| | - Amandine Pitaval
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France
| |
Collapse
|
22
|
Lee JS, Kim J, Cui B, Kim SK, Cho SA, An S, Cho SW. Hybrid skin chips for toxicological evaluation of chemical drugs and cosmetic compounds. LAB ON A CHIP 2022; 22:343-353. [PMID: 34904990 DOI: 10.1039/d1lc00550b] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Development of drugs and cosmetics for topical application require safety tests in skin models. However, current skin models, such as skin cell sheets and artificial tissue-engineered skin, do not allow sophisticated toxicological evaluations (e.g., sensory irritation, hepatotoxicity). Animal models are prohibited worldwide for testing cosmetics. Therefore, reliable human skin models that recapitulate physiological events in skin tissue need to be established under in vitro settings. In this study, hybrid human skin models that enable delicate toxicological evaluations of drugs and cosmetic compounds are demonstrated. To recapitulate skin cornification, keratinocytes in the top layer of a vertical microfluidic chip were cultured at the air-liquid interface. For the skin-nerve hybrid model, differentiated neural stem cells in 3D collagen were positioned adjacent to and right below the skin layer. This model enables real-time quantitative skin sensitization analysis following chemical treatments by detecting alterations in neuronal activity in combination with a calcium imaging technique. For the skin-liver model, hepatic cells derived from pluripotent stem cells were cultured in 3D collagen distant from the skin layer. Potential hepatotoxicity of cutaneously applied chemicals in this model can be evaluated by quantification of glutathione and reactive oxygen species. Our study suggests that 3D hybrid skin chips would provide useful human skin models in pharmaceutical and cosmetic industries.
Collapse
Affiliation(s)
- Jong Seung Lee
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea.
| | - Jin Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea.
| | - Baofang Cui
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea.
| | - Su Kyeom Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea.
| | - Sun-A Cho
- Safety & Microbiology Lab, Amorepacific Co. R&D Unit, Yongin 17038, Republic of Korea
| | - Susun An
- Safety & Microbiology Lab, Amorepacific Co. R&D Unit, Yongin 17038, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
23
|
Shafiee S, Shariatzadeh S, Zafari A, Majd A, Niknejad H. Recent Advances on Cell-Based Co-Culture Strategies for Prevascularization in Tissue Engineering. Front Bioeng Biotechnol 2021; 9:745314. [PMID: 34900955 PMCID: PMC8655789 DOI: 10.3389/fbioe.2021.745314] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/02/2021] [Indexed: 12/14/2022] Open
Abstract
Currently, the fabrication of a functional vascular network to maintain the viability of engineered tissues is a major bottleneck in the way of developing a more advanced engineered construct. Inspired by vasculogenesis during the embryonic period, the in vitro prevascularization strategies have focused on optimizing communications and interactions of cells, biomaterial and culture conditions to develop a capillary-like network to tackle the aforementioned issue. Many of these studies employ a combination of endothelial lineage cells and supporting cells such as mesenchymal stem cells, fibroblasts, and perivascular cells to create a lumenized endothelial network. These supporting cells are necessary for the stabilization of the newly developed endothelial network. Moreover, to optimize endothelial network development without impairing biomechanical properties of scaffolds or differentiation of target tissue cells, several other factors, including target tissue, endothelial cell origins, the choice of supporting cell, culture condition, incorporated pro-angiogenic factors, and choice of biomaterial must be taken into account. The prevascularization method can also influence the endothelial lineage cell/supporting cell co-culture system to vascularize the bioengineered constructs. This review aims to investigate the recent advances on standard cells used in in vitro prevascularization methods, their co-culture systems, and conditions in which they form an organized and functional vascular network.
Collapse
Affiliation(s)
- Sepehr Shafiee
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siavash Shariatzadeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Zafari
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Majd
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Kim M, Jang J. Construction of 3D hierarchical tissue platforms for modeling diabetes. APL Bioeng 2021; 5:041506. [PMID: 34703970 PMCID: PMC8530538 DOI: 10.1063/5.0055128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus (DM) is one of the most serious systemic diseases worldwide, and the majority of DM patients face severe complications. However, many of underlying disease mechanisms related to these complications are difficult to understand with the use of currently available animal models. With the urgent need to fundamentally understand DM pathology, a variety of 3D biomimetic platforms have been generated by the convergence of biofabrication and tissue engineering strategies for the potent drug screening platform of pre-clinical research. Here, we suggest key requirements for the fabrication of physiomimetic tissue models in terms of recapitulating the cellular organization, creating native 3D microenvironmental niches for targeted tissue using biomaterials, and applying biofabrication technologies to implement tissue-specific geometries. We also provide an overview of various in vitro DM models, from a cellular level to complex living systems, which have been developed using various bioengineering approaches. Moreover, we aim to discuss the roadblocks facing in vitro tissue models and end with an outlook for future DM research.
Collapse
Affiliation(s)
- Myungji Kim
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH, 77 Cheongam-ro, Namgu, Pohang, Kyungbuk, 37673, Republic of Korea
| | - Jinah Jang
- Author to whom correspondence should be addressed:
| |
Collapse
|
25
|
Hawthorne WJ, Fuller E, Thomas A, Rao JS, Burlak C. Updateon xenotransplantation for May/June 2021. Xenotransplantation 2021; 28:e12710. [PMID: 34617623 DOI: 10.1111/xen.12710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/02/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Wayne J Hawthorne
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia.,Department of Surgery, Westmead Clinical School, Westmead Hospital, University of Sydney, Westmead, New South Wales, Australia
| | - Erin Fuller
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Adwin Thomas
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Joseph Sushil Rao
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA.,Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Christopher Burlak
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
26
|
Ewald ML, Chen YH, Lee AP, Hughes CCW. The vascular niche in next generation microphysiological systems. LAB ON A CHIP 2021; 21:3244-3262. [PMID: 34396383 PMCID: PMC8635227 DOI: 10.1039/d1lc00530h] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
In recent years, microphysiological system (MPS, also known as, organ-on-a-chip or tissue chip) platforms have emerged with great promise to improve the predictive capacity of preclinical modeling thereby reducing the high attrition rates when drugs move into trials. While their designs can vary quite significantly, in general MPS are bioengineered in vitro microenvironments that recapitulate key functional units of human organs, and that have broad applications in human physiology, pathophysiology, and clinical pharmacology. A critical next step in the evolution of MPS devices is the widespread incorporation of functional vasculature within tissues. The vasculature itself is a major organ that carries nutrients, immune cells, signaling molecules and therapeutics to all other organs. It also plays critical roles in inducing and maintaining tissue identity through expression of angiocrine factors, and in providing tissue-specific milieus (i.e., the vascular niche) that can support the survival and function of stem cells. Thus, organs are patterned, maintained and supported by the vasculature, which in turn receives signals that drive tissue specific gene expression. In this review, we will discuss published vascularized MPS platforms and present considerations for next-generation devices looking to incorporate this critical constituent. Finally, we will highlight the organ-patterning processes governed by the vasculature, and how the incorporation of a vascular niche within MPS platforms will establish a unique opportunity to study stem cell development.
Collapse
Affiliation(s)
- Makena L Ewald
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA.
| | | | | | | |
Collapse
|
27
|
Abstract
Recreating human organ-level function in vitro is a rapidly evolving field that integrates tissue engineering, stem cell biology, and microfluidic technology to produce 3D organoids. A critical component of all organs is the vasculature. Herein, we discuss general strategies to create vascularized organoids, including common source materials, and survey previous work using vascularized organoids to recreate specific organ functions and simulate tumor progression. Vascularization is not only an essential component of individual organ function but also responsible for coupling the fate of all organs and their functions. While some success in coupling two or more organs together on a single platform has been demonstrated, we argue that the future of vascularized organoid technology lies in creating organoid systems complete with tissue-specific microvasculature and in coupling multiple organs through a dynamic vascular network to create systems that can respond to changing physiological conditions.
Collapse
Affiliation(s)
- Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA;
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California 92697, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA;
| |
Collapse
|
28
|
Kuwabara R, Hu S, Smink AM, Orive G, Lakey JRT, de Vos P. Applying Immunomodulation to Promote Longevity of Immunoisolated Pancreatic Islet Grafts. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:129-140. [PMID: 33397201 DOI: 10.1089/ten.teb.2020.0326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Islet transplantation is a promising therapy for insulin-dependent diabetes, but large-scale application is hampered by the lack of a consistent source of insulin-producing cells and need for lifelong administration of immunosuppressive drugs, which are associated with severe side effects. To avoid chronic immunosuppression, islet grafts can be enveloped in immunoisolating polymeric membranes. These immunoisolating polymeric membranes protect islet grafts from cell-mediated rejection while allowing diffusion of oxygen, nutrients, and insulin. Although clinical trials have shown the safety and feasibility of encapsulated islets to control glucose homeostasis, the strategy does up till now not support long-term graft survival. This partly can be explained by a significant loss of insulin-producing cells in the immediate period after implantation. The loss can be prevented by combining immunoisolation with immunomodulation, such as combined administration of immunomodulating cytokines or coencapsulation of immunomodulating cell types such as regulatory T cells, mesenchymal stem cells, or Sertoli cells. Also, administration of specific antibodies or apoptotic donor leucocytes is considered to create a tolerant microenvironment around immunoisolated grafts. In this review, we describe the outcomes and limitations of these approaches, as well as the recent progress in immunoisolating devices. Impact statement Immunoisolation by enveloping islets in semipermeable membranes allows for successful transplantation of islet grafts in the absence of chronic immunosuppression, but the duration of graft survival is still not permanent. The reasons for long-term final graft failure is not fully understood, but combining immunoisolation with immunomodulation of tissues or host immune system has been proposed to enhance the longevity of grafts. This article reviews the recent progress and challenges of immunoisolation, as well as the benefits and feasibility of combining encapsulation approaches with immunomodulation to promote longevity of encapsulated grafts.
Collapse
Affiliation(s)
- Rei Kuwabara
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Biomaterials, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shuxian Hu
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexandra M Smink
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Jonathan R T Lakey
- Department of Surgery and Biomedical Engineering, University of California Irvine, Irvine, California, USA
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
29
|
Lewis PL, Wells JM. Engineering-inspired approaches to study β-cell function and diabetes. Stem Cells 2021; 39:522-535. [PMID: 33497522 DOI: 10.1002/stem.3340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/13/2021] [Indexed: 12/21/2022]
Abstract
Strategies to mitigate the pathologies from diabetes range from simply administering insulin to prescribing complex drug/biologic regimens combined with lifestyle changes. There is a substantial effort to better understand β-cell physiology during diabetes pathogenesis as a means to develop improved therapies. The convergence of multiple fields ranging from developmental biology to microfluidic engineering has led to the development of new experimental systems to better study complex aspects of diabetes and β-cell biology. Here we discuss the available insulin-secreting cell types used in research, ranging from primary human β-cells, to cell lines, to pluripotent stem cell-derived β-like cells. Each of these sources possess inherent strengths and weaknesses pertinent to specific applications, especially in the context of engineered platforms. We then outline how insulin-expressing cells have been used in engineered platforms and how recent advances allow for better mimicry of in vivo conditions. Chief among these conditions are β-cell interactions with other endocrine organs. This facet is beginning to be thoroughly addressed by the organ-on-a-chip community, but holds enormous potential in the development of novel diabetes therapeutics. Furthermore, high throughput strategies focused on studying β-cell biology, improving β-cell differentiation, or proliferation have led to enormous contributions in the field and will no doubt be instrumental in bringing new diabetes therapeutics to the clinic.
Collapse
Affiliation(s)
- Phillip L Lewis
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - James M Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
30
|
Abadpour S, Aizenshtadt A, Olsen PA, Shoji K, Wilson SR, Krauss S, Scholz H. Pancreas-on-a-Chip Technology for Transplantation Applications. Curr Diab Rep 2020; 20:72. [PMID: 33206261 PMCID: PMC7674381 DOI: 10.1007/s11892-020-01357-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Human pancreas-on-a-chip (PoC) technology is quickly advancing as a platform for complex in vitro modeling of islet physiology. This review summarizes the current progress and evaluates the possibility of using this technology for clinical islet transplantation. RECENT FINDINGS PoC microfluidic platforms have mainly shown proof of principle for long-term culturing of islets to study islet function in a standardized format. Advancement in microfluidic design by using imaging-compatible biomaterials and biosensor technology might provide a novel future tool for predicting islet transplantation outcome. Progress in combining islets with other tissue types gives a possibility to study diabetic interventions in a minimal equivalent in vitro environment. Although the field of PoC is still in its infancy, considerable progress in the development of functional systems has brought the technology on the verge of a general applicable tool that may be used to study islet quality and to replace animal testing in the development of diabetes interventions.
Collapse
Affiliation(s)
- Shadab Abadpour
- Department of Transplant Medicine and Institute for Surgical Research, Oslo University Hospital, Post Box 4950, Nydalen, N-0424 Oslo, Norway
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Aleksandra Aizenshtadt
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Petter Angell Olsen
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kayoko Shoji
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Steven Ray Wilson
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Chemistry, University of Oslo, Oslo, Norway
| | - Stefan Krauss
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Institute of Immunology, Oslo University Hospital, Oslo, Norway
| | - Hanne Scholz
- Department of Transplant Medicine and Institute for Surgical Research, Oslo University Hospital, Post Box 4950, Nydalen, N-0424 Oslo, Norway
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
31
|
Gnecco JS, Brown AT, Kan EL, Baugh L, Ives C, Loring M, Griffith LG. Physiomimetic Models of Adenomyosis. Semin Reprod Med 2020; 38:179-196. [PMID: 33176387 PMCID: PMC7803459 DOI: 10.1055/s-0040-1719084] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adenomyosis remains an enigmatic disease in the clinical and research communities. The high prevalence, diversity of morphological and symptomatic presentations, array of potential etiological explanations, and variable response to existing interventions suggest that different subgroups of patients with distinguishable mechanistic drivers of disease may exist. These factors, combined with the weak links to genetic predisposition, make the entire spectrum of the human condition challenging to model in animals. Here, after an overview of current approaches, a vision for applying physiomimetic modeling to adenomyosis is presented. Physiomimetics combines a system's biology analysis of patient populations to generate hypotheses about mechanistic bases for stratification with in vitro patient avatars to test these hypotheses. A substantial foundation for three-dimensional (3D) tissue engineering of adenomyosis lesions exists in several disparate areas: epithelial organoid technology; synthetic biomaterials matrices for epithelial–stromal coculture; smooth muscle 3D tissue engineering; and microvascular tissue engineering. These approaches can potentially be combined with microfluidic platform technologies to model the lesion microenvironment and can potentially be coupled to other microorgan systems to examine systemic effects. In vitro patient-derived models are constructed to answer specific questions leading to target identification and validation in a manner that informs preclinical research and ultimately clinical trial design.
Collapse
Affiliation(s)
- Juan S Gnecco
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Alex T Brown
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Ellen L Kan
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Lauren Baugh
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Clara Ives
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Megan Loring
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Endometriosis and Adenomyosis Care Collaborative, Center for Minimally Invasive Gynecologic Surgery, Newton Wellesley Hospital, Newton, Massachusetts
| | - Linda G Griffith
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
32
|
Zbinden A, Urbanczyk M, Layland SL, Becker L, Marzi J, Bosch M, Loskill P, Duffy GP, Schenke-Layland K. Collagen and Endothelial Cell Coculture Improves β-Cell Functionality and Rescues Pancreatic Extracellular Matrix. Tissue Eng Part A 2020; 27:977-991. [PMID: 33023407 DOI: 10.1089/ten.tea.2020.0250] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The use of biomaterials and biomaterial functionalization is a promising approach to support pancreatic islet viability posttransplantation in an effort to reduce insulin dependence for patients afflicted with diabetes mellitus type 1. Extracellular matrix (ECM) proteins are known to impact numerous reparative functions in the body. Assessing how endogenously expressed pancreatic ECM proteins are affected by posttransplant-like hypoxic conditions may provide significant insights toward the development of tissue-engineered therapeutic strategies to positively influence β-cell survival, proliferation, and functionality. Here, we investigated the expression of three relevant groups of pancreatic ECM proteins in human native tissue, including basement membrane (BM) proteins (collagen type 4 [COL4], laminins [LAM]), proteoglycans (decorin [DCN], nidogen-1 [NID1]), and fibril-forming proteins (fibronectin [FN], collagen type 1 [COL1]). In an in vitro hypoxia model, we identified that ECM proteins were differently affected by hypoxic conditions, contributing to an overall loss of β-cell functionality. The use of a COL1 hydrogel as carrier material demonstrated a protective effect on β-cells mitigating the effect of hypoxia on proteoglycans as well as fibril-forming protein expression, supporting β-cell functionality in hypoxia. We further showed that providing endothelial cells (ECs) into the COL1 hydrogel improves β-cell response as well as the expression of relevant BM proteins. Our data show that β-cells benefit from a microenvironment composed of structure-providing COL1 with the incorporation of ECs to withstand the harsh conditions of hypoxia. Such hydrogels support β-cell survival and can serve as an initial source of ECM proteins to allow cell engraftment while preserving cell functionality posttransplantation. Impact statement Expression analysis identifies hypoxia-induced pathological changes in extracellular matrix (ECM) homeostasis as potential targets to support β-cell transplants by encapsulation in biomaterials for the treatment of diabetes mellitus. A collagen-1 hydrogel is shown to attenuate the effect of hypoxia on β-cells and their ECM expression. The functionalization of the hydrogel with endothelial cells increases the β-cell response to glucose and rescues essential basement membrane proteins.
Collapse
Affiliation(s)
- Aline Zbinden
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Max Urbanczyk
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Shannon L Layland
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Lucas Becker
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard Karls University Tübingen, Tübingen, Germany
| | - Julia Marzi
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard Karls University Tübingen, Tübingen, Germany.,NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Mariella Bosch
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Peter Loskill
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany.,Fraunhofer IGB, Stuttgart, Germany
| | - Garry P Duffy
- Anatomy & Regenerative Medicine Institute, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Katja Schenke-Layland
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard Karls University Tübingen, Tübingen, Germany.,NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany.,Anatomy & Regenerative Medicine Institute, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland.,Cardiovascular Research Laboratories, Department of Medicine/Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|