1
|
Ribeiro VL, Dernowsek JA, Fernandes RR, Pitol DL, Issa JPM, Mazzi-Chaves JF, Bombonato-Prado KF, Sousa-Neto MD, Passos GA. Repopulation of a 3D simulated periapical lesion cavity with dental pulp stem cell spheroids with triggered osteoblastic differentiation. Braz Dent J 2024; 35:e235847. [PMID: 39699491 DOI: 10.1590/0103-644020235847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/02/2024] [Indexed: 12/20/2024] Open
Abstract
We established a proof-of-concept model system for the biological healing of periapical lesions using stem cell spheroids. Mesenchymal stem cells from human exfoliated deciduous teeth (SHED) were cultured in a 2D monolayer and then as 3D multicellular spheroids. An image of a periapical lesion of an upper lateral incisor tooth was obtained by computed tomography and was used as a model for photopolymer resin 3D printing to generate a negative frame of the lesion. The negative model served to prepare a positive model of the periapical lesion cavity in an agarose gel. SHED that were cultured in monolayers or as spheroids were seeded in the positive lesion mold before or after osteoblastic differentiation. The results showed that compared to cells cultured in monolayers, spheroids exhibited uniform cellularity and a greater viability within the lesion cavity, which was accompanied by a temporal reduction in the expression of CD13, CD29, CD44, CD73, and CD90 mRNAs that are typically expressed by stem cells. Concomitantly, the expression of markers that characterize osteoblastic differentiation (RUNX2, ALP, and BGLAP) increased. These results provide a new perspective for regenerative endodontics with the use of SHED-derived spheroids to repair periapical lesions.
Collapse
Affiliation(s)
- Vítor Luís Ribeiro
- Program in Restorative Dentistry, Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
- Center for Cell-Based Therapy in Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
| | - Janaína A Dernowsek
- Biotechnology Center, Energy and Nuclear Research Institute (IPEN), University of São Paulo, São Paulo, SP, Brazil
| | - Roger R Fernandes
- Department of Bucomaxilofacial Surgery, School of Dentistry of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Dimitrius L Pitol
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
| | - João Paulo Mardegan Issa
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
| | - Jardel F Mazzi-Chaves
- Program in Restorative Dentistry, Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
- Center for Cell-Based Therapy in Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
| | - Karina Fittipaldi Bombonato-Prado
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
- Center for Cell-Based Therapy in Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
| | - Manoel Damião Sousa-Neto
- Program in Restorative Dentistry, Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
- Center for Cell-Based Therapy in Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
| | - Geraldo Aleixo Passos
- Program in Restorative Dentistry, Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
- Center for Cell-Based Therapy in Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
- Laboratory of Genetics and Molecular Biology, Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo(USP), Ribeirão Preto, SP, Brazil
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, USP, Ribeirão Preto, SP, Brazil
| |
Collapse
|
2
|
Sasaki T, Hisada S, Kanki H, Nunomura K, Lin B, Nishiyama K, Kawano T, Matsumura S, Mochizuki H. Modulation of Ca 2+ oscillation following ischemia and nicotinic acetylcholine receptors in primary cortical neurons by high-throughput analysis. Sci Rep 2024; 14:27667. [PMID: 39532929 PMCID: PMC11557898 DOI: 10.1038/s41598-024-77882-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: 07/27/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Calcium oscillations in primary neuronal cultures and iPSCs have been employed to investigate arrhythmogenicity and epileptogenicity in drug development. Previous studies have demonstrated that Ca2+ influx via NMDA and nicotinic acetylcholine receptors (nAChRs) modulates Ca2+ oscillations. Nevertheless, there has been no comprehensive investigation into the impact of ischemia or nAChR-positive allosteric modulators (PAM) drugs on Ca2+ oscillations at a level that would facilitate high-throughput screening. We investigated the effects of ischemia and nAChR subtypes or nAChR PAM agonists on Ca2+ oscillations in high-density 2D and 3D-sphere primary neuronal cultures using 384-well plates with FDSS-7000. Ischemia for 1 and 2 h resulted in an increase in the frequency of Ca2+ oscillations and a decrease in their amplitude in a time-dependent manner. The NMDA and AMPA receptor inhibition significantly suppressed Ca2+ oscillation. Inhibition of NR2A or NR2B had the opposite effect on Ca oscillations. The potentiation of ischemia-induced Ca2+ oscillations was significantly inhibited by the NMDA receptor antagonist, MK-801, and the frequency of these oscillations was suppressed by the NR2B inhibitor, Ro-256981. In the 3D-neurosphere, the application of an α7nAChR agonist increased the frequency of Ca2+ oscillations, whereas the activation of α4β2 had no effect. The combination of nicotine and PNU-120596 (type II PAM) affected the frequency and amplitude of Ca2+ oscillations in a manner distinct from that of type I PAM. These systems may be useful not only for detecting epileptogenicity but also in the search for neuroprotective agents against cerebral ischemia.
Collapse
Affiliation(s)
- Tsutomu Sasaki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan.
- StemRIM Institute of Regeneration-Inducing Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan.
| | - Sunao Hisada
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Hideaki Kanki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Kazuto Nunomura
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, 1‑6 Yamadaoka, Suita, Osaka, 565‑0871, Japan
| | - Bangzhong Lin
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, 1‑6 Yamadaoka, Suita, Osaka, 565‑0871, Japan
| | - Kumiko Nishiyama
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Tomohito Kawano
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Shigenobu Matsumura
- Graduate School of Comprehensive Rehabilitation, Osaka Prefecture University, Osaka, 583-8555, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
3
|
Gopallawa I, Gupta C, Jawa R, Cyril A, Jawa V, Chirmule N, Gujar V. Applications of Organoids in Advancing Drug Discovery and Development. J Pharm Sci 2024; 113:2659-2667. [PMID: 39002723 DOI: 10.1016/j.xphs.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 07/15/2024]
Abstract
Organoids are small, self-organizing three-dimensional cell cultures that are derived from stem cells or primary organs. These cultures replicate the complexity of an organ, which cannot be achieved by single-cell culture systems. Organoids can be used in testing of new drugs instead of animals. Development and validation of organoids is thus important to reduce the reliance on animals for drug testing. In this review, we have discussed the developmental and regulatory aspects of organoids and highlighted their importance in drug development. We have first summarized different types of culture-based organoid systems such as submerged Matrigel, micro-fluidic 3D cultures, inducible pluripotent stem cells, and air-liquid interface cultures. These systems help us understand the intricate interplay between cells and their surrounding milieu for identifying functions of target receptors, soluble factors, and spatial interactions. Further, we have discussed the advances in humanized severe-combined immunodeficiency mouse models and their applications in the pharmacology of immune-oncology. Since regulatory aspects are important in using organoids for drug development, we have summarized FDA and EMA regulations on organoid research to support pre-clinical studies. Finally, we have included some unique studies highlighting the use of organoids in studying infectious diseases, cancer, and fundamental biology. These studies also exemplify the latest technological advances in organoid development resulting in improved efficiency. Overall, this review comprehensively summarizes the applications of organoids in early drug development during discovery and pre-clinical studies.
Collapse
Affiliation(s)
- Indiwari Gopallawa
- Clinical Pharmacology & Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | | | - Rayan Jawa
- University of Pennsylvania, Philadelphia, PA, USA
| | - Arya Cyril
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Vibha Jawa
- Bristol Myers Squibb, Lawrenceville, NY, USA.
| | | | - Vikramsingh Gujar
- Anatomy and Cell Biology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| |
Collapse
|
4
|
Wan S, Aregueta Robles U, Poole-Warren L, Esrafilzadeh D. Advances in 3D tissue models for neural engineering: self-assembled versus engineered tissue models. Biomater Sci 2024; 12:3522-3549. [PMID: 38829222 DOI: 10.1039/d4bm00317a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Neural tissue engineering has emerged as a promising field that aims to create functional neural tissue for therapeutic applications, drug screening, and disease modelling. It is becoming evident in the literature that this goal requires development of three-dimensional (3D) constructs that can mimic the complex microenvironment of native neural tissue, including its biochemical, mechanical, physical, and electrical properties. These 3D models can be broadly classified as self-assembled models, which include spheroids, organoids, and assembloids, and engineered models, such as those based on decellularized or polymeric scaffolds. Self-assembled models offer advantages such as the ability to recapitulate neural development and disease processes in vitro, and the capacity to study the behaviour and interactions of different cell types in a more realistic environment. However, self-assembled constructs have limitations such as lack of standardised protocols, inability to control the cellular microenvironment, difficulty in controlling structural characteristics, reproducibility, scalability, and lengthy developmental timeframes. Integrating biomimetic materials and advanced manufacturing approaches to present cells with relevant biochemical, mechanical, physical, and electrical cues in a controlled tissue architecture requires alternate engineering approaches. Engineered scaffolds, and specifically 3D hydrogel-based constructs, have desirable properties, lower cost, higher reproducibility, long-term stability, and they can be rapidly tailored to mimic the native microenvironment and structure. This review explores 3D models in neural tissue engineering, with a particular focus on analysing the benefits and limitations of self-assembled organoids compared with hydrogel-based engineered 3D models. Moreover, this paper will focus on hydrogel based engineered models and probe their biomaterial components, tuneable properties, and fabrication techniques that allow them to mimic native neural tissue structures and environment. Finally, the current challenges and future research prospects of 3D neural models for both self-assembled and engineered models in neural tissue engineering will be discussed.
Collapse
Affiliation(s)
- Shuqian Wan
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| | - Ulises Aregueta Robles
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| | - Laura Poole-Warren
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
- Tyree Foundation Institute of Health Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Dorna Esrafilzadeh
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
5
|
Abuwatfa WH, Pitt WG, Husseini GA. Scaffold-based 3D cell culture models in cancer research. J Biomed Sci 2024; 31:7. [PMID: 38221607 PMCID: PMC10789053 DOI: 10.1186/s12929-024-00994-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/04/2024] [Indexed: 01/16/2024] Open
Abstract
Three-dimensional (3D) cell cultures have emerged as valuable tools in cancer research, offering significant advantages over traditional two-dimensional (2D) cell culture systems. In 3D cell cultures, cancer cells are grown in an environment that more closely mimics the 3D architecture and complexity of in vivo tumors. This approach has revolutionized cancer research by providing a more accurate representation of the tumor microenvironment (TME) and enabling the study of tumor behavior and response to therapies in a more physiologically relevant context. One of the key benefits of 3D cell culture in cancer research is the ability to recapitulate the complex interactions between cancer cells and their surrounding stroma. Tumors consist not only of cancer cells but also various other cell types, including stromal cells, immune cells, and blood vessels. These models bridge traditional 2D cell cultures and animal models, offering a cost-effective, scalable, and ethical alternative for preclinical research. As the field advances, 3D cell cultures are poised to play a pivotal role in understanding cancer biology and accelerating the development of effective anticancer therapies. This review article highlights the key advantages of 3D cell cultures, progress in the most common scaffold-based culturing techniques, pertinent literature on their applications in cancer research, and the ongoing challenges.
Collapse
Affiliation(s)
- Waad H Abuwatfa
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, P.O. Box. 26666, Sharjah, United Arab Emirates
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, P.O. Box 26666, Sharjah, United Arab Emirates
| | - William G Pitt
- Department of Chemical Engineering, Brigham Young University, Provo, UT, 84602, USA
| | - Ghaleb A Husseini
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, P.O. Box. 26666, Sharjah, United Arab Emirates.
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, P.O. Box 26666, Sharjah, United Arab Emirates.
| |
Collapse
|
6
|
Xie R, Pal V, Yu Y, Lu X, Gao M, Liang S, Huang M, Peng W, Ozbolat IT. A comprehensive review on 3D tissue models: Biofabrication technologies and preclinical applications. Biomaterials 2024; 304:122408. [PMID: 38041911 PMCID: PMC10843844 DOI: 10.1016/j.biomaterials.2023.122408] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/09/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
The limitations of traditional two-dimensional (2D) cultures and animal testing, when it comes to precisely foreseeing the toxicity and clinical effectiveness of potential drug candidates, have resulted in a notable increase in the rate of failure during the process of drug discovery and development. Three-dimensional (3D) in-vitro models have arisen as substitute platforms with the capacity to accurately depict in-vivo conditions and increasing the predictivity of clinical effects and toxicity of drug candidates. It has been found that 3D models can accurately represent complex tissue structure of human body and can be used for a wide range of disease modeling purposes. Recently, substantial progress in biomedicine, materials and engineering have been made to fabricate various 3D in-vitro models, which have been exhibited better disease progression predictivity and drug effects than convention models, suggesting a promising direction in pharmaceutics. This comprehensive review highlights the recent developments in 3D in-vitro tissue models for preclinical applications including drug screening and disease modeling targeting multiple organs and tissues, like liver, bone, gastrointestinal tract, kidney, heart, brain, and cartilage. We discuss current strategies for fabricating 3D models for specific organs with their strengths and pitfalls. We expand future considerations for establishing a physiologically-relevant microenvironment for growing 3D models and also provide readers with a perspective on intellectual property, industry, and regulatory landscape.
Collapse
Affiliation(s)
- Renjian Xie
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China
| | - Vaibhav Pal
- Department of Chemistry, Pennsylvania State University, University Park, PA, USA; The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Yanrong Yu
- School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China
| | - Xiaolu Lu
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China
| | - Mengwei Gao
- School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China
| | - Shijie Liang
- School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China
| | - Miao Huang
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China
| | - Weijie Peng
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China; School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China.
| | - Ibrahim T Ozbolat
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA; Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Materials Research Institute, Pennsylvania State University, University Park, PA, USA; Department of Neurosurgery, Pennsylvania State College of Medicine, Hershey, PA, USA; Penn State Cancer Institute, Penn State University, Hershey, PA, 17033, USA; Department of Medical Oncology, Cukurova University, Adana, 01130, Turkey; Biotechnology Research and Application Center, Cukurova University, Adana, 01130, Turkey.
| |
Collapse
|
7
|
Pereira I, Lopez-Martinez MJ, Samitier J. Advances in current in vitro models on neurodegenerative diseases. Front Bioeng Biotechnol 2023; 11:1260397. [PMID: 38026882 PMCID: PMC10658011 DOI: 10.3389/fbioe.2023.1260397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Many neurodegenerative diseases are identified but their causes and cure are far from being well-known. The problem resides in the complexity of the neural tissue and its location which hinders its easy evaluation. Although necessary in the drug discovery process, in vivo animal models need to be reduced and show relevant differences with the human tissues that guide scientists to inquire about other possible options which lead to in vitro models being explored. From organoids to organ-on-a-chips, 3D models are considered the cutting-edge technology in cell culture. Cell choice is a big parameter to take into consideration when planning an in vitro model and cells capable of mimicking both healthy and diseased tissue, such as induced pluripotent stem cells (iPSC), are recognized as good candidates. Hence, we present a critical review of the latest models used to study neurodegenerative disease, how these models have evolved introducing microfluidics platforms, 3D cell cultures, and the use of induced pluripotent cells to better mimic the neural tissue environment in pathological conditions.
Collapse
Affiliation(s)
- Inês Pereira
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Maria J. Lopez-Martinez
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| |
Collapse
|
8
|
Pistollato F, Campia I, Daskalopoulos EP, Bernasconi C, Desaintes C, Di Virgilio S, Kyriakopoulou C, Whelan M, Deceuninck P. Gauging innovation and health impact from biomedical research: survey results and interviews with recipients of EU-funding in the fields of Alzheimer's disease, breast cancer and prostate cancer. Health Res Policy Syst 2023; 21:66. [PMID: 37386455 DOI: 10.1186/s12961-023-00981-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/05/2023] [Indexed: 07/01/2023] Open
Abstract
Biomedical research on Alzheimer's disease (AD), breast cancer (BC) and prostate cancer (PC) has globally improved our understanding of the etiopathological mechanisms underlying the onset of these diseases, often with the goal to identify associated genetic and environmental risk factors and develop new medicines. However, the prevalence of these diseases and failure rate in drug development remain high. Being able to retrospectively monitor the major scientific breakthroughs and impact of such investment endeavors is important to re-address funding strategies if and when needed. The EU has supported research into those diseases via its successive framework programmes for research, technological development and innovation. The European Commission (EC) has already undertaken several activities to monitor research impact. As an additional contribution, the EC Joint Research Centre (JRC) launched in 2020 a survey addressed to former and current participants of EU-funded research projects in the fields of AD, BC and PC, with the aim to understand how EU-funded research has contributed to scientific innovation and societal impact, and how the selection of the experimental models may have underpinned the advances made. Further feedback was also gathered through in-depth interviews with some selected survey participants representative of the diverse pre-clinical models used in the EU-funded projects. A comprehensive analysis of survey replies, complemented with the information derived from the interviews, has recently been published in a Synopsis report. Here we discuss the main findings of this analysis and propose a set of priority actions that could be considered to help improving the translation of scientific innovation of biomedical research into societal impact.
Collapse
Affiliation(s)
- Francesca Pistollato
- European Commission, Joint Research Centre (JRC), Directorate F-Health, Consumers and Reference Materials, Via E. Fermi 2749, 21027, Ispra, VA, Italy
| | - Ivana Campia
- European Commission, Joint Research Centre (JRC), Directorate F-Health, Consumers and Reference Materials, Via E. Fermi 2749, 21027, Ispra, VA, Italy
| | - Evangelos P Daskalopoulos
- European Commission, Joint Research Centre (JRC), Directorate F-Health, Consumers and Reference Materials, Via E. Fermi 2749, 21027, Ispra, VA, Italy
| | - Camilla Bernasconi
- European Commission, Joint Research Centre (JRC), Directorate F-Health, Consumers and Reference Materials, Via E. Fermi 2749, 21027, Ispra, VA, Italy
| | | | - Sergio Di Virgilio
- European Commission, DG Research & Innovation (DG RTD), Brussels, Belgium
| | | | - Maurice Whelan
- European Commission, Joint Research Centre (JRC), Directorate F-Health, Consumers and Reference Materials, Via E. Fermi 2749, 21027, Ispra, VA, Italy
| | - Pierre Deceuninck
- European Commission, Joint Research Centre (JRC), Directorate F-Health, Consumers and Reference Materials, Via E. Fermi 2749, 21027, Ispra, VA, Italy.
| |
Collapse
|
9
|
Correia CD, Ferreira A, Fernandes MT, Silva BM, Esteves F, Leitão HS, Bragança J, Calado SM. Human Stem Cells for Cardiac Disease Modeling and Preclinical and Clinical Applications—Are We on the Road to Success? Cells 2023; 12:1727. [DOI: https:/doi.org/10.3390/cells12131727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
Cardiovascular diseases (CVDs) are pointed out by the World Health Organization (WHO) as the leading cause of death, contributing to a significant and growing global health and economic burden. Despite advancements in clinical approaches, there is a critical need for innovative cardiovascular treatments to improve patient outcomes. Therapies based on adult stem cells (ASCs) and embryonic stem cells (ESCs) have emerged as promising strategies to regenerate damaged cardiac tissue and restore cardiac function. Moreover, the generation of human induced pluripotent stem cells (iPSCs) from somatic cells has opened new avenues for disease modeling, drug discovery, and regenerative medicine applications, with fewer ethical concerns than those associated with ESCs. Herein, we provide a state-of-the-art review on the application of human pluripotent stem cells in CVD research and clinics. We describe the types and sources of stem cells that have been tested in preclinical and clinical trials for the treatment of CVDs as well as the applications of pluripotent stem-cell-derived in vitro systems to mimic disease phenotypes. How human stem-cell-based in vitro systems can overcome the limitations of current toxicological studies is also discussed. Finally, the current state of clinical trials involving stem-cell-based approaches to treat CVDs are presented, and the strengths and weaknesses are critically discussed to assess whether researchers and clinicians are getting closer to success.
Collapse
Affiliation(s)
- Cátia D. Correia
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Anita Ferreira
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- School of Health, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Bárbara M. Silva
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Doctoral Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Filipa Esteves
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Helena S. Leitão
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - José Bragança
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Sofia M. Calado
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
10
|
Correia CD, Ferreira A, Fernandes MT, Silva BM, Esteves F, Leitão HS, Bragança J, Calado SM. Human Stem Cells for Cardiac Disease Modeling and Preclinical and Clinical Applications-Are We on the Road to Success? Cells 2023; 12:1727. [PMID: 37443761 PMCID: PMC10341347 DOI: 10.3390/cells12131727] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Abstract
Cardiovascular diseases (CVDs) are pointed out by the World Health Organization (WHO) as the leading cause of death, contributing to a significant and growing global health and economic burden. Despite advancements in clinical approaches, there is a critical need for innovative cardiovascular treatments to improve patient outcomes. Therapies based on adult stem cells (ASCs) and embryonic stem cells (ESCs) have emerged as promising strategies to regenerate damaged cardiac tissue and restore cardiac function. Moreover, the generation of human induced pluripotent stem cells (iPSCs) from somatic cells has opened new avenues for disease modeling, drug discovery, and regenerative medicine applications, with fewer ethical concerns than those associated with ESCs. Herein, we provide a state-of-the-art review on the application of human pluripotent stem cells in CVD research and clinics. We describe the types and sources of stem cells that have been tested in preclinical and clinical trials for the treatment of CVDs as well as the applications of pluripotent stem-cell-derived in vitro systems to mimic disease phenotypes. How human stem-cell-based in vitro systems can overcome the limitations of current toxicological studies is also discussed. Finally, the current state of clinical trials involving stem-cell-based approaches to treat CVDs are presented, and the strengths and weaknesses are critically discussed to assess whether researchers and clinicians are getting closer to success.
Collapse
Affiliation(s)
- Cátia D. Correia
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Anita Ferreira
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- School of Health, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Bárbara M. Silva
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Doctoral Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Filipa Esteves
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Helena S. Leitão
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - José Bragança
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Sofia M. Calado
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
11
|
Zhang Z, Liu Y, Tao X, Du P, Enkhbat M, Lim KS, Wang H, Wang PY. Engineering Cell Microenvironment Using Nanopattern-Derived Multicellular Spheroids and Photo-Crosslinked Gelatin/Hyaluronan Hydrogels. Polymers (Basel) 2023; 15:polym15081925. [PMID: 37112072 PMCID: PMC10144125 DOI: 10.3390/polym15081925] [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: 03/21/2023] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Cell cultures of dispersed cells within hydrogels depict the interaction of the cell-extracellular matrix (ECM) in 3D, while the coculture of different cells within spheroids combines both the effects of cell-cell and cell-ECM interactions. In this study, the cell co-spheroids of human bone mesenchymal stem cells/human umbilical vein endothelial cells (HBMSC/HUVECs) are prepared with the assistance of a nanopattern, named colloidal self-assembled patterns (cSAPs), which is superior to low-adhesion surfaces. A phenol-modified gelatin/hyaluronan (Gel-Ph/HA-Ph) hydrogel is used to encapsulate the multicellular spheroids and the constructs are photo-crosslinked using blue light. The results show that Gel-Ph/HA-Ph hydrogels with a 5%-to-0.3% ratio have the best properties. Cells in HBMSC/HUVEC co-spheroids are more favorable for osteogenic differentiation (Runx2, ALP, Col1a1 and OPN) and vascular network formation (CD31+ cells) compared to HBMSC spheroids. In a subcutaneous nude mouse model, the HBMSC/HUVEC co-spheroids showed better performance than HBMSC spheroids in angiogenesis and the development of blood vessels. Overall, this study paves a new way for using nanopatterns, cell coculturing and hydrogel technology for the generation and application of multicellular spheroids.
Collapse
Affiliation(s)
- Zhen Zhang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Liu
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xuelian Tao
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ping Du
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Myagmartsend Enkhbat
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Khoon S Lim
- School of Medical Sciences, University of Sydney, Sydney, NSW 2052, Australia
| | - Huaiyu Wang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng-Yuan Wang
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
12
|
Zhang D, Eguchi N, Okazaki S, Sora I, Hishimoto A. Telencephalon Organoids Derived from an Individual with ADHD Show Altered Neurodevelopment of Early Cortical Layer Structure. Stem Cell Rev Rep 2023:10.1007/s12015-023-10519-z. [PMID: 36872412 PMCID: PMC10366301 DOI: 10.1007/s12015-023-10519-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 03/07/2023]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder that occurs in early childhood and can persist to adulthood. It can affect many aspects of a patient's daily life, so it is necessary to explore the mechanism and pathological alterations. For this purpose, we applied induced pluripotent stem cell (iPSC)-derived telencephalon organoids to recapitulate the alterations occurring in the early cerebral cortex of ADHD patients. We found that telencephalon organoids of ADHD showed less growth of layer structures than control-derived organoids. On day 35 of differentiation, the thinner cortex layer structures of ADHD-derived organoids contained more neurons than those of control-derived organoids. Furthermore, ADHD-derived organoids showed a decrease in cell proliferation during development from day 35 to 56. On day 56 of differentiation, there was a significant difference in the proportion of symmetric and asymmetric cell division between the ADHD and control groups. In addition, we observed increased cell apoptosis in ADHD during early development. These results show alterations in the characteristics of neural stem cells and the formation of layer structures, which might indicate key roles in the pathogenesis of ADHD. Our organoids exhibit the cortical developmental alterations observed in neuroimaging studies, providing an experimental foundation for understanding the pathological mechanisms of ADHD.
Collapse
Affiliation(s)
- Danmeng Zhang
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noriomi Eguchi
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satoshi Okazaki
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ichiro Sora
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Akitoyo Hishimoto
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
13
|
Rathore RS, R Ayyannan S, Mahto SK. Emerging three-dimensional neuronal culture assays for neurotherapeutics drug discovery. Expert Opin Drug Discov 2022; 17:619-628. [DOI: 10.1080/17460441.2022.2061458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Rahul S Rathore
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi-221005, UP, India
| | - Senthil R Ayyannan
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi-221005, UP, India
| | - Sanjeev K Mahto
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi-221005, UP, India
| |
Collapse
|
14
|
Akbaba TH, Bekircan-Kurt CE, Balci-Peynircioglu B, Balci-Hayta B. Biologia Futura: the importance of 3D organoids-a new approach for research on neurological and rare diseases. Biol Futur 2021; 72:281-290. [PMID: 34554549 DOI: 10.1007/s42977-021-00070-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022]
Abstract
3D cell cultures and organoid approach are increasingly being used for basic research and drug discovery of several diseases. Recent advances in these technologies, enabling research on tissue-like structures created in vitro is very important for the value of the data produced. Application of 3D cultures will not only contribute to advancing basic research, but also help to reduce animal usage in biomedical science. The 3D organoid approach is important for research on diseases where patient tissue is difficult to obtain. Therefore, this review aims to show recent advances in the 3D organoid technology in disease modeling and potential usage in translational and personalized medicine of diseases with limited patient material such as neurological diseases and rare diseases.
Collapse
Affiliation(s)
- Tayfun Hilmi Akbaba
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey
| | - Can Ebru Bekircan-Kurt
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey
| | - Banu Balci-Peynircioglu
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey
| | - Burcu Balci-Hayta
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey.
| |
Collapse
|
15
|
Co-Stimulation of Oxytocin and Arginine-Vasopressin Receptors Affect Hypothalamic Neurospheroid Size. Int J Mol Sci 2021; 22:ijms22168464. [PMID: 34445168 PMCID: PMC8395152 DOI: 10.3390/ijms22168464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022] Open
Abstract
Oxytocin (OXT) is a neuropeptide involved in a plethora of behavioral and physiological processes. However, there is a prominent lack of 3D cell culture models that investigate the effects of OXT on a cellular/molecular level. In this study, we established a hypothalamic neuronal spheroid model to investigate the cellular response in a more realistic 3D setting. Our data indicate that the formation of spheroids itself does not alter the basic characteristics of the cell line and that markers of cellular morphology and connectivity are stably expressed. We found that both OXT and arginine vasopressin (AVP) treatment increase spheroid size (surface area and volume), as well as individual nucleus size, which serves as an indicator for cellular proliferation. The cellular response to both OXT and AVP seems mainly to be mediated by the AVP receptor 1a (V1aR); however, the OXT receptor (OXTR) contributes significantly to the observed proliferative effect. When we blocked the OXTR pharmacologically or knocked down the OXTR by siRNA, the OXT- or AVP-induced cellular proliferation decreased. In summary, we established a 3D cell culture model of the neuronal response to OXT and AVP and found that spheroids react to the treatment via their respective receptors but also via cross-talk between the two receptor types.
Collapse
|
16
|
Jorgensen M, Gibbons A, Sui K, Carpenter R, Zhang X, Xie Y. Predictable fabrication of pre-made alginate hydrogel microtubes for stem cell aggregation using needle-in-needle devices. Biofabrication 2021; 13:035043. [PMID: 33930885 DOI: 10.1088/1758-5090/abfd7b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/30/2021] [Indexed: 12/18/2022]
Abstract
Alginate hydrogels in microtubular structures have great potential to advance three-dimensional (3D) culture, organoid formation, tissue engineering, and cell therapy. To address the need of fabricating consistent, stable hydrogel microtubes for efficient large organoid generation in a simple and quick manner, we have designed needle-in-needle devices to fabricate alginate hydrogel microtubes without any dead volume of the cell-alginate mixture and demonstrated the feasibility of injecting and culturing embryoid bodies in these pre-made hydrogel microtubes. We further used a reverse engineering approach to find out the optimal flow rates and alginate concentration for fabricating pre-made hydrogel microtubes with desired diameter using particular sets of needle-in-needle devices. We established the relationship of the alginate flow rate with diameter and wall thickness of the microtube using mathematic modeling. It offers a way to determine the flow rate for making microtubes with the desired dimension. Additionally, we evaluated the effect of CaCl2concentration on the diameter as well as stem cell viability. At last, we demonstrated the capacity of fabricating hydrogel microtubes of varying diameters using three sets of needle-in-needle devices and evaluated stem cell growth in these hydrogel microtubes. It provides a new avenue to accessible, repeatable, scalable, and easy to use pre-made 'off-the-shelf' hydrogel microtubes for 3D cell culture including, but not limiting to stem cells.
Collapse
Affiliation(s)
- Matthew Jorgensen
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, United States of America
| | - Ashley Gibbons
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, United States of America
| | - Kevin Sui
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, United States of America
| | - Rebecca Carpenter
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, United States of America
| | - Xulang Zhang
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, United States of America
| | - Yubing Xie
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, United States of America
| |
Collapse
|
17
|
Riss T, Trask OJ. Factors to consider when interrogating 3D culture models with plate readers or automated microscopes. In Vitro Cell Dev Biol Anim 2021; 57:238-256. [PMID: 33564998 PMCID: PMC7946695 DOI: 10.1007/s11626-020-00537-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/02/2020] [Indexed: 11/27/2022]
Abstract
Along with the increased use of more physiologically relevant three-dimensional cell culture models comes the responsibility of researchers to validate new assay methods that measure events in structures that are physically larger and more complex compared to monolayers of cells. It should not be assumed that assays designed using monolayers of cells will work for cells cultured as larger three-dimensional masses. The size and barriers for penetration of molecules through the layers of cells result in a different microenvironment for the cells in the outer layer compared to the center of three-dimensional structures. Diffusion rates for nutrients and oxygen may limit metabolic activity which is often measured as a marker for cell viability. For assays that lyse cells, the penetration of reagents to achieve uniform cell lysis must be considered. For live cell fluorescent imaging assays, the diffusion of fluorescent probes and penetration of photons of light for probe excitation and fluorescent emission must be considered. This review will provide an overview of factors to consider when implementing assays to interrogate three dimensional cell culture models.
Collapse
Affiliation(s)
- Terry Riss
- Promega Corporation, Cell Health, 2800 Woods Hollow Road, Fitchburg, WI, 53711, USA.
| | - O Joseph Trask
- PerkinElmer Inc., Life Sciences and Technology, 940 Winter Street, Waltham, MA, 02451, USA
| |
Collapse
|
18
|
Hereditary Optic Neuropathies: Induced Pluripotent Stem Cell-Based 2D/3D Approaches. Genes (Basel) 2021; 12:genes12010112. [PMID: 33477675 PMCID: PMC7831942 DOI: 10.3390/genes12010112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/10/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
Inherited optic neuropathies share visual impairment due to the degeneration of retinal ganglion cells (RGCs) as the hallmark of the disease. This group of genetic disorders are caused by mutations in nuclear genes or in the mitochondrial DNA (mtDNA). An impaired mitochondrial function is the underlying mechanism of these diseases. Currently, optic neuropathies lack an effective treatment, and the implementation of induced pluripotent stem cell (iPSC) technology would entail a huge step forward. The generation of iPSC-derived RGCs would allow faithfully modeling these disorders, and these RGCs would represent an appealing platform for drug screening as well, paving the way for a proper therapy. Here, we review the ongoing two-dimensional (2D) and three-dimensional (3D) approaches based on iPSCs and their applications, taking into account the more innovative technologies, which include tissue engineering or microfluidics.
Collapse
|
19
|
Arzua T, Yan Y, Jiang C, Logan S, Allison RL, Wells C, Kumar SN, Schäfer R, Bai X. Modeling alcohol-induced neurotoxicity using human induced pluripotent stem cell-derived three-dimensional cerebral organoids. Transl Psychiatry 2020; 10:347. [PMID: 33051447 PMCID: PMC7553959 DOI: 10.1038/s41398-020-01029-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/11/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Maternal alcohol exposure during pregnancy can substantially impact the development of the fetus, causing a range of symptoms, known as fetal alcohol spectrum disorders (FASDs), such as cognitive dysfunction and psychiatric disorders, with the pathophysiology and mechanisms largely unknown. Recently developed human cerebral organoids from induced pluripotent stem cells are similar to fetal brains in the aspects of development and structure. These models allow more relevant in vitro systems to be developed for studying FASDs than animal models. Modeling binge drinking using human cerebral organoids, we sought to quantify the downstream toxic effects of alcohol (ethanol) on neural pathology phenotypes and signaling pathways within the organoids. The results revealed that alcohol exposure resulted in unhealthy organoids at cellular, subcellular, bioenergetic metabolism, and gene expression levels. Alcohol induced apoptosis on organoids. The apoptotic effects of alcohol on the organoids depended on the alcohol concentration and varied between cell types. Specifically, neurons were more vulnerable to alcohol-induced apoptosis than astrocytes. The alcohol-treated organoids exhibit ultrastructural changes such as disruption of mitochondria cristae, decreased intensity of mitochondrial matrix, and disorganized cytoskeleton. Alcohol exposure also resulted in mitochondrial dysfunction and metabolic stress in the organoids as evidenced by (1) decreased mitochondrial oxygen consumption rates being linked to basal respiration, ATP production, proton leak, maximal respiration and spare respiratory capacity, and (2) increase of non-mitochondrial respiration in alcohol-treated organoids compared with control groups. Furthermore, we found that alcohol treatment affected the expression of 199 genes out of 17,195 genes analyzed. Bioinformatic analyses showed the association of these dysregulated genes with 37 pathways related to clinically relevant pathologies such as psychiatric disorders, behavior, nervous system development and function, organismal injury and abnormalities, and cellular development. Notably, 187 of these genes are critically involved in neurodevelopment, and/or implicated in nervous system physiology and neurodegeneration. Furthermore, the identified genes are key regulators of multiple pathways linked in networks. This study extends for the first time animal models of binge drinking-related FASDs to a human model, allowing in-depth analyses of neurotoxicity at tissue, cellular, subcellular, metabolism, and gene levels. Hereby, we provide novel insights into alcohol-induced pathologic phenotypes, cell type-specific vulnerability, and affected signaling pathways and molecular networks, that can contribute to a better understanding of the developmental neurotoxic effects of binge drinking during pregnancy.
Collapse
Affiliation(s)
- Thiago Arzua
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Yasheng Yan
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Congshan Jiang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Sarah Logan
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Reilly L Allison
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Clive Wells
- Department of Microbiology, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Suresh N Kumar
- Department of Pathology, Children's Research Institute Imaging Core, Neuroscience Imaging Facility, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohaematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen gGmbH, Goethe University Hospital, 60438, Frankfurt am Main, Germany
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, 53226, WI, USA.
| |
Collapse
|
20
|
Pistollato F, Bernasconi C, McCarthy J, Campia I, Desaintes C, Wittwehr C, Deceuninck P, Whelan M. Alzheimer's Disease, and Breast and Prostate Cancer Research: Translational Failures and the Importance to Monitor Outputs and Impact of Funded Research. Animals (Basel) 2020; 10:E1194. [PMID: 32674379 PMCID: PMC7401638 DOI: 10.3390/ani10071194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022] Open
Abstract
Dementia and cancer are becoming increasingly prevalent in Western countries. In the last two decades, research focused on Alzheimer's disease (AD) and cancer, in particular, breast cancer (BC) and prostate cancer (PC), has been substantially funded both in Europe and worldwide. While scientific research outcomes have contributed to increase our understanding of the disease etiopathology, still the prevalence of these chronic degenerative conditions remains very high across the globe. By definition, no model is perfect. In particular, animal models of AD, BC, and PC have been and still are traditionally used in basic/fundamental, translational, and preclinical research to study human disease mechanisms, identify new therapeutic targets, and develop new drugs. However, animals do not adequately model some essential features of human disease; therefore, they are often unable to pave the way to the development of drugs effective in human patients. The rise of new technological tools and models in life science, and the increasing need for multidisciplinary approaches have encouraged many interdisciplinary research initiatives. With considerable funds being invested in biomedical research, it is becoming pivotal to define and apply indicators to monitor the contribution to innovation and impact of funded research. Here, we discuss some of the issues underlying translational failure in AD, BC, and PC research, and describe how indicators could be applied to retrospectively measure outputs and impact of funded biomedical research.
Collapse
Affiliation(s)
- Francesca Pistollato
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Camilla Bernasconi
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Janine McCarthy
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
- Physicians Committee for Responsible Medicine (PCRM), Washington, DC 20016, USA;
| | - Ivana Campia
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Christian Desaintes
- European Commission, Directorate General for Research and Innovation (RTD), 1000 Brussels, Belgium;
| | - Clemens Wittwehr
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Pierre Deceuninck
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Maurice Whelan
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| |
Collapse
|
21
|
Betz JF, Ho VB, Gaston JD. 3D Bioprinting and Its Application to Military Medicine. Mil Med 2020; 185:e1510-e1519. [DOI: 10.1093/milmed/usaa121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/13/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022] Open
Abstract
Abstract
Introduction
Traditionally, tissue engineering techniques have largely focused on 2D cell culture models—monolayers of immortalized or primary cells growing on tissue culture plastic. Although these techniques have proven useful in research, they often lack physiological validity, because of the absence of fundamental tissue properties, such as multicellular organization, specialized extracellular matrix structures, and molecular or force gradients essential to proper physiological function. More recent advances in 3D cell culture methods have facilitated the development of more complex physiological models and tissue constructs; however, these often rely on self-organization of cells (bottom-up design), and the range of tissue construct size and complexity generated by these methods remains relatively limited. By borrowing from advances in the additive manufacturing field, 3D bioprinting techniques are enabling top-down design and fabrication of cellular constructs with controlled sizing, spacing, and chemical functionality. The high degree of control over engineered tissue architecture, previously unavailable to researchers, enables the generation of more complex, physiologically relevant 3D tissue constructs. Three main 3D bioprinting techniques are reviewed—extrusion, droplet-based, and laser-assisted bioprinting techniques are among the more robust 3D bioprinting techniques, each with its own strengths and weaknesses. High complexity tissue constructs created through 3D bioprinting are opening up new avenues in tissue engineering, regenerative medicine, and physiological model systems for researchers in the military medicine community.
Materials and Methods
Recent primary literature and reviews were selected to provide a broad overview of the field of 3D bioprinting and illustrate techniques and examples of 3D bioprinting relevant to military medicine. References were selected to illustrate specific examples of advances and potential military medicine applications in the 3D bioprinting field, rather than to serve as a comprehensive review.
Results
Three classes of 3D bioprinting techniques were reviewed: extrusion, droplet-based, and laser-assisted bioprinting. Advantages, disadvantages, important considerations, and constraints of each technique were discussed. Examples from the primary literature were given to illustrate the techniques. Relevant applications of 3D bioprinting to military medicine, namely tissue engineering/regenerative medicine and new models of physiological systems, are discussed in the context of advancing military medicine.
Conclusions
3D bioprinting is a rapidly evolving field that provides researchers the ability to build tissue constructs that are more complex and physiologically relevant than traditional 2D culture methods. Advances in bioprinting techniques, bioink formulation, and cell culture methods are being translated into new paradigms in tissue engineering and physiological system modeling, advancing the state of the art, and increasing construct availability to the military medicine research community.
Collapse
Affiliation(s)
- Jordan F Betz
- Geneva Foundation, 917 Pacific Ave, Tacoma, WA 98402
- Department of Radiology and Radiological Sciences, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814
| | - Vincent B Ho
- Department of Radiology and Radiological Sciences, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814
| | - Joel D Gaston
- Geneva Foundation, 917 Pacific Ave, Tacoma, WA 98402
- Department of Radiology and Radiological Sciences, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814
| |
Collapse
|
22
|
Chow EKH. The 2020 SLAS Technology Ten: Translating Life Sciences Innovation. SLAS Technol 2020; 25:1-5. [PMID: 31958032 DOI: 10.1177/2472630319896750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
23
|
Eglen RM. Convergence of Three-Dimensional Cell Culture and Human iPS Cells: Improving Clinical Relevance in Drug Discovery. SLAS Technol 2019; 24:1-2. [PMID: 30798681 DOI: 10.1177/2472630318810819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|