1
|
Kumar D, Gupta S, Gupta V, Tanwar R, Chandel A. Engineering the Future of Regenerative Medicines in Gut Health with Stem Cell-Derived Intestinal Organoids. Stem Cell Rev Rep 2025:10.1007/s12015-025-10893-w. [PMID: 40380985 DOI: 10.1007/s12015-025-10893-w] [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: 05/05/2025] [Indexed: 05/19/2025]
Abstract
The advent of intestinal organoids, three-dimensional structures derived from stem cells, has significantly advanced the field of biology by providing robust in vitro models that closely mimic the architecture and functionality of the human intestine. These organoids, generated from induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), or adult stem cells, possess remarkable capabilities for self-renewal, differentiation into diverse intestinal cell types, and functional recapitulation of physiological processes, including nutrient absorption, epithelial barrier integrity, and host-microbe interactions. The utility of intestinal organoids has been extensively demonstrated in disease modeling, drug screening, and personalized medicine. Notable examples include iPSC-derived organoids, which have been effectively employed to model enteric infections, and ESC-derived organoids, which have provided critical insights into fetal intestinal development. Patient-derived organoids have emerged as powerful tools for investigating personalized therapeutics and regenerative interventions for conditions such as inflammatory bowel disease (IBD), cystic fibrosis, and colorectal cancer. Preclinical studies involving transplantation of human intestinal organoids into murine models have shown promising outcomes, including functional integration, epithelial restoration, and immune system interactions. Despite these advancements, several challenges persist, particularly in achieving reproducibility, scalability, and maturation of organoids, which hinder their widespread clinical translation. Addressing these limitations requires the establishment of standardized protocols for organoid generation, culture, storage, and analysis to ensure reproducibility and comparability of findings across studies. Nevertheless, intestinal organoids hold immense promise for transforming our understanding of gastrointestinal pathophysiology, enhancing drug development pipelines, and advancing personalized medicine. By bridging the gap between preclinical research and clinical applications, these organoids represent a paradigm shift in the exploration of novel therapeutic strategies and the investigation of gut-associated diseases.
Collapse
Affiliation(s)
- Dinesh Kumar
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India.
| | - Sonia Gupta
- Swami Devi Dyal Group of Professional Institute, Panchkula, India
| | - Vrinda Gupta
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| | - Rajni Tanwar
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| | - Anchal Chandel
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| |
Collapse
|
2
|
Andreatta F, Hendriks D, Artegiani B. Human Organoids as an Emerging Tool for Genome Screenings. Annu Rev Biomed Eng 2025; 27:157-183. [PMID: 40310889 DOI: 10.1146/annurev-bioeng-103023-122327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Over the last decade, a plethora of organoid models have been generated to recapitulate aspects of human development, disease, tissue homeostasis, and repair. Organoids representing multiple tissues have emerged and are typically categorized based on their origin. Tissue-derived organoids are established directly from tissue-resident stem/progenitor cells of either adult or fetal origin. Starting from pluripotent stem cells (PSCs), PSC-derived organoids instead recapitulate the developmental trajectory of a given organ. Gene editing technologies, particularly the CRISPR-Cas toolbox, have greatly facilitated gene manipulation experiments with considerable ease and scalability, revolutionizing organoid-based human biology research. Here, we review the recent adaptation of CRISPR-based screenings in organoids. We examine the strategies adopted to perform CRISPR screenings in organoids, discuss different screening scopes and readouts, and highlight organoid-specific challenges. We then discuss individual organoid-based genome screening studies that have uncovered novel genes involved in a variety of biological processes. We close by providing an outlook on how widespread adaptation of CRISPR screenings across the organoid field may be achieved, to ultimately leverage our understanding of human biology.
Collapse
Affiliation(s)
| | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; ,
| | | |
Collapse
|
3
|
Wu X, Hu Y, Sheng S, Yang H, Li Z, Han Q, Zhang Q, Su J. DNA-based hydrogels for bone regeneration: A promising tool for bone organoids. Mater Today Bio 2025; 31:101502. [PMID: 39911372 PMCID: PMC11795821 DOI: 10.1016/j.mtbio.2025.101502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 02/07/2025] Open
Abstract
DNA-based hydrogels stand out for bone regeneration due to their exceptional biocompatibility and programmability. These hydrogels facilitate the formation of spatial bone structures through bulk hydrogel fabricating, microsphere formatting, and 3D printing. Furthermore, the bone microenvironment can be finely tuned by leveraging the degradation products, nanostructure, targeting, and delivery capabilities inherent to DNA-based materials. In this review, we underscore the advantages of DNA-based hydrogels, detailing their composition, gelation techniques, and structure optimization. We then delineate three critical elements in the promotion of bone regeneration using DNA-based hydrogels: (i) osteogenesis driven by phosphate ions, plasmids, and oligodeoxynucleotides (ODNs) that enhance mineralization and promote gene and protein expression; (ii) vascularization facilitated by tetrahedral DNA nanostructures (TDNs) and aptamers, which boosts gene expression and targeted release; (iii) immunomodulation achieved through loaded factors, TDNs, and bound ions that stimulate macrophage polarization and exhibit antibacterial properties. With these advantages and properties, these DNA-based hydrogels can be used to construct bone organoids, providing an innovative tool for disease modeling and therapeutic applications in bone tissue engineering. Finally, we discuss the current challenges and future prospects, emphasizing the potential impacts and applications in regenerative medicine.
Collapse
Affiliation(s)
- Xiang Wu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Materials Science and Engineering, Shanghai University, Shanghai, 200444, China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Huijian Yang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Clinical Laboratory, Shanghai Zhongye Hospital, Shanghai, 201941, China
| | - Zuhao Li
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Qinglin Han
- Department of Orthopedics, The Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Qin Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Sanming Institute of Translational Medicine, Sanming, 365004, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
4
|
Atalay-Sahar E, Yildiz-Ozturk E, Ozgur S, Aral A, Dayanc E, Goksel T, Meuwissen R, Yesil-Celiktas O, Goksel O. Novel Approach Methodologies in Modeling Complex Bioaerosol Exposure in Asthma and Allergic Rhinitis Under Climate Change. Expert Rev Mol Med 2025; 27:e13. [PMID: 40070355 PMCID: PMC11964095 DOI: 10.1017/erm.2025.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/28/2025] [Accepted: 03/06/2025] [Indexed: 03/26/2025]
Abstract
The undeniable impact of climate change and air pollution on respiratory health has led to increasing cases of asthma, allergic rhinitis and other chronic non-communicable immune-mediated upper and lower airway diseases. Natural bioaerosols, such as pollen and fungi, are essential atmospheric components undergoing significant structural and functional changes due to industrial pollution and atmospheric warming. Pollutants like particulate matter(PMx), polycyclic aromatic hydrocarbons(PAHs), nitrogen dioxide(NO2), sulfur dioxide(SO2) and carbon monoxide(CO) modify the surface and biological properties of atmospheric bioaerosols such as pollen and fungi, enhancing their allergenic potentials. As a result, sensitized individuals face heightened risks of asthma exacerbation, and these alterations likely contribute to the rise in frequency and severity of allergic diseases. NAMs, such as precision-cut lung slices(PCLS), air-liquid interface(ALI) cultures and lung-on-a-chip models, along with the integration of data from these innovative models with computational models, provide better insights into how environmental factors influence asthma and allergic diseases compared to traditional models. These systems simulate the interaction between pollutants and the respiratory system with higher precision, helping to better understand the health implications of bioaerosol exposure. Additionally, NAMs improve preclinical study outcomes by offering higher throughput, reduced costs and greater reproducibility, enhancing the translation of data into clinical applications. This review critically evaluates the potential of NAMs in researching airway diseases, with a focus on allergy and asthma. It highlights their advantages in studying the increasingly complex structures of bioaerosols under conditions of environmental pollution and climate change, while also addressing the existing gaps, challenges and limitations of these models.
Collapse
Affiliation(s)
- Esra Atalay-Sahar
- Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir, Türkiye
| | - Ece Yildiz-Ozturk
- Department of Food Processing, Food Technology Programme, Yasar University, Izmir, Türkiye
| | - Su Ozgur
- Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir, Türkiye
- Regional Hub for Cancer Registration in Northern Africa, Central and Western Asia, WHO/IARC GICR, Izmir, Türkiye
| | - Arzu Aral
- Department of Immunology, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Emre Dayanc
- Basic Medical Sciences, Faculty of Medicine, İzmir University of Economics, Izmir, Türkiye
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA
| | - Tuncay Goksel
- Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir, Türkiye
- Department of Pulmonary Medicine, Division of Immunology and Allergy, Faculty of Medicine, Ege University, Izmir, Türkiye
| | - Ralph Meuwissen
- Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir, Türkiye
| | - Ozlem Yesil-Celiktas
- Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir, Türkiye
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Türkiye
- ODTÜ MEMS Center[CMT2], Ankara, Türkiye
| | - Ozlem Goksel
- Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir, Türkiye
- Department of Pulmonary Medicine, Division of Immunology and Allergy, Faculty of Medicine, Ege University, Izmir, Türkiye
| |
Collapse
|
5
|
Kim J, Ślęczkowska M, Nobre B, Wieringa P. Study Models for Chlamydia trachomatis Infection of the Female Reproductive Tract. Microorganisms 2025; 13:553. [PMID: 40142446 PMCID: PMC11945960 DOI: 10.3390/microorganisms13030553] [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: 12/23/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
Chlamydia trachomatis (Ct) is a leading cause of sexually transmitted infections globally, often resulting in inflammatory disorders, ectopic pregnancies, and infertility. Studying Ct's pathogenesis remains challenging due to its unique life cycle and host-specific interactions, which require diverse experimental models. Animal studies using mouse, guinea pig, pig, and non-human primate models provide valuable insights into immune responses, hormonal influences, and disease progression. However, they face limitations in terms of translational relevance due to physiological differences, as well as ethical concerns. Complementing these, in vitro systems, ranging from simple monolayer to advanced three-dimensional models, exhibit improved physiological relevance by replicating the human tissue architecture. This includes the detailed investigation of epithelial barrier disruptions, epithelium-stroma interactions, and immune responses at a cellular level. Nonetheless, in vitro models fall short in mimicking the intricate tissue structures found in vivo and, therefore, cannot faithfully replicate the host-pathogen interactions or infection dynamics observed in living organisms. This review presents a comprehensive overview of the in vivo and in vitro models employed over the past few decades to investigate Ct and its pathogenesis, addressing their strengths and limitations. Furthermore, we explore emerging technologies, including organ-on-chip and in silico models, as promising tools to overcome the existing challenges and refine our understanding of Ct infections.
Collapse
Affiliation(s)
| | | | | | - Paul Wieringa
- Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.K.); (M.Ś.); (B.N.)
| |
Collapse
|
6
|
Kalla J, Pfneissl J, Mair T, Tran L, Egger G. A systematic review on the culture methods and applications of 3D tumoroids for cancer research and personalized medicine. Cell Oncol (Dordr) 2025; 48:1-26. [PMID: 38806997 PMCID: PMC11850459 DOI: 10.1007/s13402-024-00960-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2024] [Indexed: 05/30/2024] Open
Abstract
Cancer is a highly heterogeneous disease, and thus treatment responses vary greatly between patients. To improve therapy efficacy and outcome for cancer patients, more representative and patient-specific preclinical models are needed. Organoids and tumoroids are 3D cell culture models that typically retain the genetic and epigenetic characteristics, as well as the morphology, of their tissue of origin. Thus, they can be used to understand the underlying mechanisms of cancer initiation, progression, and metastasis in a more physiological setting. Additionally, co-culture methods of tumoroids and cancer-associated cells can help to understand the interplay between a tumor and its tumor microenvironment. In recent years, tumoroids have already helped to refine treatments and to identify new targets for cancer therapy. Advanced culturing systems such as chip-based fluidic devices and bioprinting methods in combination with tumoroids have been used for high-throughput applications for personalized medicine. Even though organoid and tumoroid models are complex in vitro systems, validation of results in vivo is still the common practice. Here, we describe how both animal- and human-derived tumoroids have helped to identify novel vulnerabilities for cancer treatment in recent years, and how they are currently used for precision medicine.
Collapse
Affiliation(s)
- Jessica Kalla
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Janette Pfneissl
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Theresia Mair
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Loan Tran
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Vienna, Austria.
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Li J, Li Y, Song G, Wang H, Zhang Q, Wang M, Zhao M, Wang B, Zhu H, Ranzhi L, Wang Q, Xiong Y. Revolutionizing cardiovascular research: Human organoids as a Beacon of hope for understanding and treating cardiovascular diseases. Mater Today Bio 2025; 30:101396. [PMID: 39802826 PMCID: PMC11719415 DOI: 10.1016/j.mtbio.2024.101396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 11/25/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Organoids, exhibiting the capability to undergo differentiation in specific in vitro growth environments, have garnered significant attention in recent years due to their capacity to recapitulate human organs with resemblant in vivo structures and physiological functions. This groundbreaking technology offers a unique opportunity to study human diseases and address the limitations of traditional animal models. Cardiovascular diseases (CVDs), a leading cause of mortality worldwide, have spurred an increasing number of researchers to explore the great potential of human cardiovascular organoids for cardiovascular research. This review initiates by elaborating on the development and manufacture of human cardiovascular organoids, including cardiac organoids and blood vessel organoids. Next, we provide a comprehensive overview of their applications in modeling various cardiovascular disorders. Furthermore, we shed light on the prospects of cardiovascular organoids in CVDs therapy, and unfold an in-depth discussion of the current challenges of human cardiovascular organoids in the development and application for understanding and treating CVDs.
Collapse
Affiliation(s)
- Jinli Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
- Department of Orthopaedics, Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Guangming Road, Shenmu, China
| | - Yang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Guangtao Song
- Department of Orthopaedics, Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Guangming Road, Shenmu, China
| | - Haiying Wang
- Department of Science and Education, Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Shenmu, China
| | - Qing Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Min Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Muxue Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Bei Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - HuiGuo Zhu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Liu Ranzhi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Qiang Wang
- Department of Orthopaedics, Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Guangming Road, Shenmu, China
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
| |
Collapse
|
8
|
Grigoreva TA, Kindt DN, Sagaidak AV, Novikova DS, Tribulovich VG. Cellular Systems for Colorectal Stem Cancer Cell Research. Cells 2025; 14:170. [PMID: 39936962 PMCID: PMC11817814 DOI: 10.3390/cells14030170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Oncological diseases consistently occupy leading positions among the most life-threatening diseases, including in highly developed countries. At the same time, the second most common cause of cancer death is colorectal cancer. The current level of research shows that the development of effective therapy, in this case, requires a new grade of understanding processes during the emergence and development of a tumor. In particular, the concept of cancer stem cells that ensure the survival of chemoresistant cells capable of giving rise to new tumors is becoming widespread. To provide adequate conditions that reproduce natural processes typical for tumor development, approaches based on increasingly complex cellular systems are being improved. This review discusses the main strategies that allow for the study of the properties of tumor cells with an emphasis on colorectal cancer stem cells. The features of working with tumor cells and the advantages and disadvantages of 2D and 3D culture systems are considered.
Collapse
Affiliation(s)
- Tatyana A. Grigoreva
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia (V.G.T.)
| | | | | | | | | |
Collapse
|
9
|
Diensthuber M, Stöver T. Organoids-the key to novel therapies for the inner ear? HNO 2024; 72:83-88. [PMID: 38775829 DOI: 10.1007/s00106-023-01367-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2023] [Indexed: 12/05/2024]
Abstract
The sensitivity and the complexity of the human inner ear in conjunction with the lack of regenerative capacity are the main reasons for hearing loss and tinnitus. Progress in the development of protective and regenerative therapies for the inner ear often failed in the past not least due to the fact that no suitable model systems for cell biological and pharmacological in vitro studies were available. A novel technology for creating "mini-organs", so-called organoids, could solve this problem and has now also reached inner ear research. It makes it possible to produce inner ear organoids from cochlear stem/progenitor cells, embryonic and induced pluripotent stem cells that mimic the structural characteristics and functional properties of the natural inner ear. This review focuses on the biological basis of these inner ear organoids, the current state of research and the promising prospects that are now opening up for basic and translational inner ear research.
Collapse
Affiliation(s)
- Marc Diensthuber
- University Hospital, Department of Otorhinolaryngology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt/M, Germany.
| | - Timo Stöver
- University Hospital, Department of Otorhinolaryngology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt/M, Germany
| |
Collapse
|
10
|
Al Monla R, Daien V, Michon F. Advanced bioengineering strategies broaden the therapeutic landscape for corneal failure. Front Bioeng Biotechnol 2024; 12:1480772. [PMID: 39605752 PMCID: PMC11598527 DOI: 10.3389/fbioe.2024.1480772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
The cornea acts as the eye foremost protective layer and is essential for its focusing power. Corneal blindness may arise from physical trauma or conditions like dystrophies, keratitis, keratoconus, or ulceration. While conventional treatments involve medical therapies and donor allografts-sometimes supplemented with keratoprostheses-these options are not suitable for all corneal defects. Consequently, the development of bioartificial corneal tissue has emerged as a critical research area, aiming to address the global shortage of human cornea donors. Bioengineered corneas hold considerable promise as substitutes, with the potential to replace either specific layers or the entire thickness of damaged corneas. This review first delves into the structural anatomy of the human cornea, identifying key attributes necessary for successful corneal tissue bioengineering. It then examines various corneal pathologies, current treatments, and their limitations. Finally, the review outlines the primary approaches in corneal tissue engineering, exploring cell-free, cell-based, and scaffold-based options as three emerging strategies to address corneal failure.
Collapse
Affiliation(s)
- Reem Al Monla
- Institute for Neurosciences of Montpellier, INSERM, University of Montpellier, Montpellier, France
| | - Vincent Daien
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
- Sydney Medical School, The Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Frederic Michon
- Institute for Neurosciences of Montpellier, INSERM, University of Montpellier, Montpellier, France
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
| |
Collapse
|
11
|
Tong L, Cui W, Zhang B, Fonseca P, Zhao Q, Zhang P, Xu B, Zhang Q, Li Z, Seashore-Ludlow B, Yang Y, Si L, Lundqvist A. Patient-derived organoids in precision cancer medicine. MED 2024; 5:1351-1377. [PMID: 39341206 DOI: 10.1016/j.medj.2024.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/11/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024]
Abstract
Organoids are three-dimensional (3D) cultures, normally derived from stem cells, that replicate the complex structure and function of human tissues. They offer a physiologically relevant model to address important questions in cancer research. The generation of patient-derived organoids (PDOs) from various human cancers allows for deeper insights into tumor heterogeneity and spatial organization. Additionally, interrogating non-tumor stromal cells increases the relevance in studying the tumor microenvironment, thereby enhancing the relevance of PDOs in personalized medicine. PDOs mark a significant advancement in cancer research and patient care, signifying a shift toward more innovative and patient-centric approaches. This review covers aspects of PDO cultures to address the modeling of the tumor microenvironment, including extracellular matrices, air-liquid interface and microfluidic cultures, and organ-on-chip. Specifically, the role of PDOs as preclinical models in gene editing, molecular profiling, drug testing, and biomarker discovery and their potential for guiding personalized treatment in clinical practice are discussed.
Collapse
Affiliation(s)
- Le Tong
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| | - Weiyingqi Cui
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Boya Zhang
- Organcare (Shenzhen) Biotechnology Company, Shenzhen, China
| | - Pedro Fonseca
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Qian Zhao
- Organcare (Shenzhen) Biotechnology Company, Shenzhen, China
| | - Ping Zhang
- Organcare (Shenzhen) Biotechnology Company, Shenzhen, China
| | - Beibei Xu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qisi Zhang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | | | - Ying Yang
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Zhejiang, China
| | - Longlong Si
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
12
|
Tatullo M, Cocco T, Ferretta A, Caroppo R, Marrelli B, Spagnuolo G, Paduano F. Unveiling the Neurodegenerative Alterations through Oral Stem Cells. J Dent Res 2024; 103:1100-1108. [PMID: 39275988 DOI: 10.1177/00220345241265661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative condition characterized by the progressive and selective loss of dopaminergic (DAergic) neurons in the midbrain. The replacement of neuromelanin (NM)-containing DAergic neurons in the substantia nigra and the enhancement of NM concentration could offer a promising and safe approach to treating PD symptoms. The objective of this study was to investigate and compare the potential of human periapical-cysts mesenchymal stem cells (hPCy-MSCs) and dental pulp stem cells (DPSCs) to differentiate into DAergic NM-producing neurons and to generate functional 3-dimensional (3D) midbrain-like organoids in vitro. We assessed the changes in morphology and behavior of neuron-like cells (NLCs) as well as the expression of molecular markers characterizing the DAergic neurons. Furthermore, we observed electrically active and functionally mature DAergic neurons by means of electrophysiological assays, NM dosage assays, and the quantification of dopamine release by high-performance liquid chromatography. Our results demonstrate for the first time that both hPCy-MSCs and DPSCs are capable of differentiating into NLCs, further confirmed by the increase in lactate levels in the medium of cells exposed to neurogenic conditions. Importantly, we have induced such NLCs to further differentiate into functional DAergic NM-producing neurons. Finally, 3D midbrain-like organoids have been produced from oral stem cells: they appear as neurosphere-like structures diffusely expressing the neural marker β-III tubulin and containing NM-like granules. Our findings open up a novel and fascinating opportunity to rethink oral stem cells, and the derived 3D disease models, as a strategic and reliable tool for unveiling the neurodegenerative alterations.
Collapse
Affiliation(s)
- M Tatullo
- Department of Translational Biomedicine and Neuroscience, University of Bari "Aldo Moro," Bari, Italy
- School of Dentistry, University of Dundee, Dundee, Scotland, UK
| | - T Cocco
- Department of Translational Biomedicine and Neuroscience, University of Bari "Aldo Moro," Bari, Italy
| | - A Ferretta
- Department of Translational Biomedicine and Neuroscience, University of Bari "Aldo Moro," Bari, Italy
| | - R Caroppo
- Department of Biosciences, Biotechnology, and Environment, University of Bari "Aldo Moro," Bari, Italy
| | - B Marrelli
- Stem Cells and Medical Genetics Units, Tecnologica Research Institute and Marrelli Health, Crotone, Italy
| | - G Spagnuolo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples "Federico II," Naples, Italy
| | - F Paduano
- Stem Cells and Medical Genetics Units, Tecnologica Research Institute and Marrelli Health, Crotone, Italy
| |
Collapse
|
13
|
Yao Q, Wang B, Yu J, Pan Q, Yu Y, Feng X, Chen W, Yang J, Gao C, Cao H. ROS-responsive nanoparticle delivery of obeticholic acid mitigate primary sclerosing cholangitis. J Control Release 2024; 374:112-126. [PMID: 39117112 DOI: 10.1016/j.jconrel.2024.08.006] [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: 04/12/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Primary sclerosing cholangitis (PSC) is a challenging cholestatic liver disease marked by progressive bile duct inflammation and fibrosis that has no FDA-approved therapy. Although obeticholic acid (OCA) has been sanctioned for PSC, its clinical utility in PSC is constrained by its potential hepatotoxicity. Here, we introduce a novel therapeutic construct consisting of OCA encapsulated within a reactive oxygen species (ROS)-responsive, biodegradable polymer, further cloaked with human placenta-derived mesenchymal stem cell (hP-MSC) membrane (MPPFTU@OCA). Using PSC patient-derived organoid models, we assessed its cellular uptake and cytotoxicity. Moreover, using a PSC mouse model induced by 3,5-diethoxycarbonyl-1,4-dihydro-collidine (DDC), we demonstrated that intravenous administration of MPPFTU@OCA not only improved cholestasis via the FXR-SHP pathway but also reduced macrophage infiltration and the accumulation of intracellular ROS, and alleviated mitochondria-induced apoptosis. Finally, we verified the ability of MPPFTU@OCA to inhibit mitochondrial ROS thereby alleviating apoptosis by measuring the mitochondrial adenosine triphosphate (ATP) concentration, ROS levels, and membrane potential (ΔΨm) using H2O2-stimulated PSC-derived organoids. These results illuminate the synergistic benefits of integrating an ROS-responsive biomimetic platform with OCA, offering a promising therapeutic avenue for PSC.
Collapse
Affiliation(s)
- Qigu Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Beiduo Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou City 310058, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China; Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, 79 Qingchun Rd, Hangzhou City 310003, China; National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China; National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Yingduo Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Xudong Feng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Wenyi Chen
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Jinfeng Yang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China; National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou City 310058, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China; Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, 79 Qingchun Rd, Hangzhou City 310003, China; National Clinical Research Center for Infectious Diseases, Hangzhou, China.
| |
Collapse
|
14
|
Lee BC. Challenges and innovations in hematopoietic stem cell transplantation: exploring bone marrow niches and new model systems. BMB Rep 2024; 57:352-362. [PMID: 38919014 PMCID: PMC11362137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/27/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) remains an indispensable therapeutic strategy for various hematological diseases. This review discusses the pivotal role of bone marrow (BM) niches in influencing the efficacy of HSCT and evaluates the current animal models, emphasizing their limitations and the need for alternative models. Traditional animal models, mainly murine xenograft, have provided significant insights, but due to species-specific differences, are often constrained from accurately mimicking human physiological responses. These limitations highlight the importance of developing alternative models that can more realistically replicate human hematopoiesis. Emerging models that include BM organoids and BM-on-a-chip microfluidic systems promise enhanced understanding of HSCT dynamics. These models aim to provide more accurate simulations of the human BM microenvironment, potentially leading to improved preclinical assessments and therapeutic outcomes. This review highlights the complexities of the BM niche, discusses the limitations of current models, and suggests directions for future research using advanced model systems. [BMB Reports 2024; 57(8): 352-362].
Collapse
Affiliation(s)
- Byung-Chul Lee
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
- Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| |
Collapse
|
15
|
Rejas C, Junger H. Cholangiocyte Organoids in Liver Transplantation; a Comprehensive Review. Transpl Int 2024; 37:12708. [PMID: 39100755 PMCID: PMC11294148 DOI: 10.3389/ti.2024.12708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 06/17/2024] [Indexed: 08/06/2024]
Abstract
Liver transplantation is the only curative option for many liver diseases that end up in liver failure, and cholangiopathy remains a challenging complication post-liver transplant, associated with significant morbidity and potential graft loss. The low availability of organs and high demand for transplantation motivate scientists to find novel interventions. Organoids, as three-dimensional cell cultures derived from adult cells or induced pluripotent cells, may help to address this problem. Different types of organoids have been described, from which cholangiocyte organoids offer a high level of versatility and plasticity for a deeper study of liver disease mechanisms. Cholangiocytes can be obtained from different segments of the biliary tree and have shown a remarkable capacity to adapt to new environments, presenting an effective system for studying cholangiopathies. Studies using cholangiocyte organoids show promising results for disease modeling, where organoids offer fundamental features to recapitulate the complexities of tissues in vitro and uncover fundamental pathological pathways to potentially reveal therapeutic strategies for personalized medicine. Organoids could hold the potential for regeneration of injured livers, representing tools of clinical impact in regenerative medicine when tissue damage is already present.
Collapse
Affiliation(s)
| | - H. Junger
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
16
|
Liberali P, Schier AF. The evolution of developmental biology through conceptual and technological revolutions. Cell 2024; 187:3461-3495. [PMID: 38906136 DOI: 10.1016/j.cell.2024.05.053] [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: 04/12/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024]
Abstract
Developmental biology-the study of the processes by which cells, tissues, and organisms develop and change over time-has entered a new golden age. After the molecular genetics revolution in the 80s and 90s and the diversification of the field in the early 21st century, we have entered a phase when powerful technologies provide new approaches and open unexplored avenues. Progress in the field has been accelerated by advances in genomics, imaging, engineering, and computational biology and by emerging model systems ranging from tardigrades to organoids. We summarize how revolutionary technologies have led to remarkable progress in understanding animal development. We describe how classic questions in gene regulation, pattern formation, morphogenesis, organogenesis, and stem cell biology are being revisited. We discuss the connections of development with evolution, self-organization, metabolism, time, and ecology. We speculate how developmental biology might evolve in an era of synthetic biology, artificial intelligence, and human engineering.
Collapse
Affiliation(s)
- Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland; University of Basel, Basel, Switzerland.
| | | |
Collapse
|
17
|
Branciforti F, Salvi M, D’Agostino F, Marzola F, Cornacchia S, De Titta MO, Mastronuzzi G, Meloni I, Moschetta M, Porciani N, Sciscenti F, Spertini A, Spilla A, Zagaria I, Deloria AJ, Deng S, Haindl R, Szakacs G, Csiszar A, Liu M, Drexler W, Molinari F, Meiburger KM. Segmentation and Multi-Timepoint Tracking of 3D Cancer Organoids from Optical Coherence Tomography Images Using Deep Neural Networks. Diagnostics (Basel) 2024; 14:1217. [PMID: 38928633 PMCID: PMC11203156 DOI: 10.3390/diagnostics14121217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Recent years have ushered in a transformative era in in vitro modeling with the advent of organoids, three-dimensional structures derived from stem cells or patient tumor cells. Still, fully harnessing the potential of organoids requires advanced imaging technologies and analytical tools to quantitatively monitor organoid growth. Optical coherence tomography (OCT) is a promising imaging modality for organoid analysis due to its high-resolution, label-free, non-destructive, and real-time 3D imaging capabilities, but accurately identifying and quantifying organoids in OCT images remain challenging due to various factors. Here, we propose an automatic deep learning-based pipeline with convolutional neural networks that synergistically includes optimized preprocessing steps, the implementation of a state-of-the-art deep learning model, and ad-hoc postprocessing methods, showcasing good generalizability and tracking capabilities over an extended period of 13 days. The proposed tracking algorithm thoroughly documents organoid evolution, utilizing reference volumes, a dual branch analysis, key attribute evaluation, and probability scoring for match identification. The proposed comprehensive approach enables the accurate tracking of organoid growth and morphological changes over time, advancing organoid analysis and serving as a solid foundation for future studies for drug screening and tumor drug sensitivity detection based on organoids.
Collapse
Affiliation(s)
- Francesco Branciforti
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Massimo Salvi
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Filippo D’Agostino
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Francesco Marzola
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Sara Cornacchia
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Maria Olimpia De Titta
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Girolamo Mastronuzzi
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Isotta Meloni
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Miriam Moschetta
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Niccolò Porciani
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Fabrizio Sciscenti
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Alessandro Spertini
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Andrea Spilla
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Ilenia Zagaria
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Abigail J. Deloria
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria; (A.J.D.); (S.D.); (R.H.); (M.L.); (W.D.)
| | - Shiyu Deng
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria; (A.J.D.); (S.D.); (R.H.); (M.L.); (W.D.)
| | - Richard Haindl
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria; (A.J.D.); (S.D.); (R.H.); (M.L.); (W.D.)
| | - Gergely Szakacs
- Center for Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (G.S.); (A.C.)
| | - Agnes Csiszar
- Center for Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (G.S.); (A.C.)
| | - Mengyang Liu
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria; (A.J.D.); (S.D.); (R.H.); (M.L.); (W.D.)
| | - Wolfgang Drexler
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria; (A.J.D.); (S.D.); (R.H.); (M.L.); (W.D.)
| | - Filippo Molinari
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| | - Kristen M. Meiburger
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.B.); (M.S.); (F.D.); (F.M.); (S.C.); (M.O.D.T.); (G.M.); (I.M.); (M.M.); (N.P.); (F.S.); (A.S.); (A.S.); (I.Z.); (F.M.)
| |
Collapse
|
18
|
Julaiti M, Guo H, Cui T, Nijiati N, Huang P, Hu B. Application of stem cells in the study of developmental and functional toxicity of endodermal-derived organs caused by nanoparticles. Toxicol In Vitro 2024; 98:105836. [PMID: 38702034 DOI: 10.1016/j.tiv.2024.105836] [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: 03/18/2024] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
Nanoparticles have unique properties that make them useful in biomedicine. However, their extensive use raises concerns about potential hazards to the body. Therefore, it is crucial to establish effective and robust toxicology models to evaluate the developmental and functional toxicity of nanoparticles on the body. This article discusses the use of stem cells to study the developmental and functional toxicity of organs of endodermal origin due to nanoparticles. The study discovered that various types of nanoparticles have varying effects on stem cells. The application of stem cell models can provide a possibility for studying the effects of nanoparticles on organ development and function, as they can more accurately reflect the toxic mechanisms of different types of nanoparticles. However, stem cell toxicology systems currently cannot fully reflect the effects of nanoparticles on entire organs. Therefore, the establishment of organoid models and other advanced assessment models is expected to address this issue.
Collapse
Affiliation(s)
- Mulati Julaiti
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China
| | - Haoqiang Guo
- Human anatomy, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China
| | - Tingting Cui
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China
| | - Nadire Nijiati
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China
| | - Pengfei Huang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China
| | - Bowen Hu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China.
| |
Collapse
|
19
|
Şişli HB, Şenkal Turhan S, Bulut Okumuş E, Böke ÖB, Erdoğmuş Ö, Kül B, Sümer E, Doğan A. Azoxymethane-induced carcinogenesis-like model of mouse intestine and mouse embryonic stem cell-derived intestinal organoids. Mol Biol Rep 2024; 51:704. [PMID: 38824233 DOI: 10.1007/s11033-024-09660-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: 03/15/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Tumor modeling using organoids holds potential in studies of cancer development, enlightening both the intracellular and extracellular molecular mechanisms behind different cancer types, biobanking, and drug screening. Intestinal organoids can be generated in vitro using a unique type of adult stem cells which are found at the base of crypts and are characterized by their high Lgr5 expression levels. METHODS AND RESULTS In this study, we successfully established intestinal cancer organoid models by using both the BALB/c derived and mouse embryonic stem cells (mESCs)-derived intestinal organoids. In both cases, carcinogenesis-like model was developed by using azoxymethane (AOM) treatment. Carcinogenesis-like model was verified by H&E staining, immunostaining, relative mRNA expression analysis, and LC/MS analysis. The morphologic analysis demonstrated that the number of generated organoids, the number of crypts, and the intensity of the organoids were significantly augmented in AOM-treated intestinal organoids compared to non-AOM-treated ones. Relative mRNA expression data revealed that there was a significant increase in both Wnt signaling pathway-related genes and pluripotency transcription factors in the AOM-induced intestinal organoids. CONCLUSION We successfully developed simple carcinogenesis-like models using mESC-based and Lgr5 + stem cell-based intestinal organoids. Intestinal organoid based carcinogenesi models might be used for personalized cancer therapy in the future.
Collapse
Affiliation(s)
- Hatice Burcu Şişli
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, Istanbul, Turkey
| | - Selinay Şenkal Turhan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, Istanbul, Turkey
| | - Ezgi Bulut Okumuş
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, Istanbul, Turkey
| | - Özüm Begüm Böke
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, Istanbul, Turkey
| | - Özüm Erdoğmuş
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, Istanbul, Turkey
| | - Berke Kül
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, Istanbul, Turkey
| | - Engin Sümer
- Faculty of Medicine, Experimental Research Center, Yeditepe University, Istanbul, Turkey
| | - Ayşegül Doğan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
20
|
Maharjan S, Ma C, Singh B, Kang H, Orive G, Yao J, Shrike Zhang Y. Advanced 3D imaging and organoid bioprinting for biomedical research and therapeutic applications. Adv Drug Deliv Rev 2024; 208:115237. [PMID: 38447931 PMCID: PMC11031334 DOI: 10.1016/j.addr.2024.115237] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/15/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Organoid cultures offer a valuable platform for studying organ-level biology, allowing for a closer mimicry of human physiology compared to traditional two-dimensional cell culture systems or non-primate animal models. While many organoid cultures use cell aggregates or decellularized extracellular matrices as scaffolds, they often lack precise biochemical and biophysical microenvironments. In contrast, three-dimensional (3D) bioprinting allows precise placement of organoids or spheroids, providing enhanced spatial control and facilitating the direct fusion for the formation of large-scale functional tissues in vitro. In addition, 3D bioprinting enables fine tuning of biochemical and biophysical cues to support organoid development and maturation. With advances in the organoid technology and its potential applications across diverse research fields such as cell biology, developmental biology, disease pathology, precision medicine, drug toxicology, and tissue engineering, organoid imaging has become a crucial aspect of physiological and pathological studies. This review highlights the recent advancements in imaging technologies that have significantly contributed to organoid research. Additionally, we discuss various bioprinting techniques, emphasizing their applications in organoid bioprinting. Integrating 3D imaging tools into a bioprinting platform allows real-time visualization while facilitating quality control, optimization, and comprehensive bioprinting assessment. Similarly, combining imaging technologies with organoid bioprinting can provide valuable insights into tissue formation, maturation, functions, and therapeutic responses. This approach not only improves the reproducibility of physiologically relevant tissues but also enhances understanding of complex biological processes. Thus, careful selection of bioprinting modalities, coupled with appropriate imaging techniques, holds the potential to create a versatile platform capable of addressing existing challenges and harnessing opportunities in these rapidly evolving fields.
Collapse
Affiliation(s)
- Sushila Maharjan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Chenshuo Ma
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Bibhor Singh
- Winthrop L. Chenery Upper Elementary School, Belmont, MA 02478, USA
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea; College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, 01007, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore 169856, Singapore
| | - Junjie Yao
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
| |
Collapse
|
21
|
Choe MS, Yeo HC, Kim JS, Lee J, Lee HJ, Kim HR, Baek KM, Jung NY, Choi M, Lee MY. Simple modeling of familial Alzheimer's disease using human pluripotent stem cell-derived cerebral organoid technology. Stem Cell Res Ther 2024; 15:118. [PMID: 38659053 PMCID: PMC11040922 DOI: 10.1186/s13287-024-03732-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 04/12/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Cerebral organoids (COs) are the most advanced in vitro models that resemble the human brain. The use of COs as a model for Alzheimer's disease (AD), as well as other brain diseases, has recently gained attention. This study aimed to develop a human AD CO model using normal human pluripotent stem cells (hPSCs) that recapitulates the pathological phenotypes of AD and to determine the usefulness of this model for drug screening. METHODS We established AD hPSC lines from normal hPSCs by introducing genes that harbor familial AD mutations, and the COs were generated using these hPSC lines. The pathological features of AD, including extensive amyloid-β (Aβ) accumulation, tauopathy, and neurodegeneration, were analyzed using enzyme-linked immunosorbent assay, Amylo-Glo staining, thioflavin-S staining, immunohistochemistry, Bielschowsky's staining, and western blot analysis. RESULTS The AD COs exhibited extensive Aβ accumulation. The levels of paired helical filament tau and neurofibrillary tangle-like silver deposits were highly increased in the AD COs. The number of cells immunoreactive for cleaved caspase-3 was significantly increased in the AD COs. In addition, treatment of AD COs with BACE1 inhibitor IV, a β-secretase inhibitor, and compound E, a γ-secretase inhibitor, significantly attenuated the AD pathological features. CONCLUSION Our model effectively recapitulates AD pathology. Hence, it is a valuable platform for understanding the mechanisms underlying AD pathogenesis and can be used to test the efficacy of anti-AD drugs.
Collapse
Affiliation(s)
- Mu Seog Choe
- Department of Molecular Physiology, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, 41566, Daegu, Republic of Korea
| | - Han Cheol Yeo
- Department of Molecular Physiology, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, 41566, Daegu, Republic of Korea
| | - Joong Sun Kim
- Department of Veterinary Anatomy, College of Veterinary Medicine, Chonnam National University, 61186, Gwangju, Republic of Korea
| | - Jean Lee
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 03080, Seoul, Republic of Korea
| | - Hae Jun Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), 01812, Seoul, Republic of Korea
| | - Hyung-Ryong Kim
- Department of Pharmacology, College of Dentistry, Jeonbuk National University, 54896, Jeonju, Republic of Korea
| | - Kyung Min Baek
- Department of Cardiovascular and Neurologic Disease, College of Oriental Medicine, Daegu Haany University, 42158, Daegu, Republic of Korea
| | - Na-Yeon Jung
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, 50612, Yangsan, Republic of Korea
| | - Murim Choi
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 03080, Seoul, Republic of Korea.
| | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Vessel-Organ Interaction Research Center (VOICE, MRC), Kyungpook National University, 41566, Daegu, Republic of Korea.
| |
Collapse
|
22
|
Hou J, Chen YG, Xiong JW. Unveiling new horizons in heart research: the promise of multi-chamber cardiac organoids. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:10. [PMID: 38649624 PMCID: PMC11035500 DOI: 10.1186/s13619-024-00193-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
Human cardiac and other organoids have recently emerged as a groundbreaking tool for advancing our understanding the developmental biology of human organs. A recent paper from Sasha Mendjan's laboratory published in the journal Cell on December 7, 2023, reported the generation of multi-chamber cardioids from human pluripotent stem cells, a transformative technology in the field of cardiology. In this short highlight paper, we summarize their findings. Their cardioids remarkably recapitulate the complexity of the human embryonic heart, including tissue architecture, cellular diversity, and functionality providing an excellent in vitro model for investigation of human heart development, disease modeling, precision medicine, and regenerative medicine. Thus, generating cardioids is an important step forward for understanding human heart development and developing potential therapies for heart diseases.
Collapse
Affiliation(s)
- Junjie Hou
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Ye-Guang Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jing-Wei Xiong
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871, China.
| |
Collapse
|
23
|
Jabri A, Khan J, Taftafa B, Alsharif M, Mhannayeh A, Chinnappan R, Alzhrani A, Kazmi S, Mir MS, Alsaud AW, Yaqinuddin A, Assiri AM, AlKattan K, Vashist YK, Broering DC, Mir TA. Bioengineered Organoids Offer New Possibilities for Liver Cancer Studies: A Review of Key Milestones and Challenges. Bioengineering (Basel) 2024; 11:346. [PMID: 38671768 PMCID: PMC11048289 DOI: 10.3390/bioengineering11040346] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Hepatic cancer is widely regarded as the leading cause of cancer-related mortality worldwide. Despite recent advances in treatment options, the prognosis of liver cancer remains poor. Therefore, there is an urgent need to develop more representative in vitro models of liver cancer for pathophysiology and drug screening studies. Fortunately, an exciting new development for generating liver models in recent years has been the advent of organoid technology. Organoid models hold huge potential as an in vitro research tool because they can recapitulate the spatial architecture of primary liver cancers and maintain the molecular and functional variations of the native tissue counterparts during long-term culture in vitro. This review provides a comprehensive overview and discussion of the establishment and application of liver organoid models in vitro. Bioengineering strategies used to construct organoid models are also discussed. In addition, the clinical potential and other relevant applications of liver organoid models in different functional states are explored. In the end, this review discusses current limitations and future prospects to encourage further development.
Collapse
Affiliation(s)
- Abdullah Jabri
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Jibran Khan
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Bader Taftafa
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Mohamed Alsharif
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Abdulaziz Mhannayeh
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Raja Chinnappan
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Alaa Alzhrani
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21423, Saudi Arabia
| | - Shadab Kazmi
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
- Pathology and laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mohammad Shabab Mir
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh 147301, Punjab, India;
| | - Aljohara Waleed Alsaud
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Abdullah M. Assiri
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Khaled AlKattan
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Yogesh K. Vashist
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Dieter C. Broering
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Tanveer Ahmad Mir
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
24
|
Lefferts JW, Kroes S, Smith MB, Niemöller PJ, Nieuwenhuijze NDA, Sonneveld van Kooten HN, van der Ent CK, Beekman JM, van Beuningen SFB. OrgaSegment: deep-learning based organoid segmentation to quantify CFTR dependent fluid secretion. Commun Biol 2024; 7:319. [PMID: 38480810 PMCID: PMC10937908 DOI: 10.1038/s42003-024-05966-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 02/23/2024] [Indexed: 03/17/2024] Open
Abstract
Epithelial ion and fluid transport studies in patient-derived organoids (PDOs) are increasingly being used for preclinical studies, drug development and precision medicine applications. Epithelial fluid transport properties in PDOs can be measured through visual changes in organoid (lumen) size. Such organoid phenotypes have been highly instrumental for the studying of diseases, including cystic fibrosis (CF), which is characterized by genetic mutations of the CF transmembrane conductance regulator (CFTR) ion channel. Here we present OrgaSegment, a MASK-RCNN based deep-learning segmentation model allowing for the segmentation of individual intestinal PDO structures from bright-field images. OrgaSegment recognizes spherical structures in addition to the oddly-shaped organoids that are a hallmark of CF organoids and can be used in organoid swelling assays, including the new drug-induced swelling assay that we show here. OrgaSegment enabled easy quantification of organoid swelling and could discriminate between organoids with different CFTR mutations, as well as measure responses to CFTR modulating drugs. The easy-to-apply label-free segmentation tool can help to study CFTR-based fluid secretion and possibly other epithelial ion transport mechanisms in organoids.
Collapse
Affiliation(s)
- Juliet W Lefferts
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Utrecht, University Medical Center, Utrecht University, 3584 CT, Utrecht, The Netherlands
| | - Suzanne Kroes
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Utrecht, University Medical Center, Utrecht University, 3584 CT, Utrecht, The Netherlands
| | - Matthew B Smith
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Utrecht, University Medical Center, Utrecht University, 3584 CT, Utrecht, The Netherlands
- Centre for Living Technologies, Alliance TU/e, WUR, UU, UMC Utrecht, 3584 CB, Utrecht, The Netherlands
| | - Paul J Niemöller
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Utrecht, University Medical Center, Utrecht University, 3584 CT, Utrecht, The Netherlands
| | - Natascha D A Nieuwenhuijze
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Utrecht, University Medical Center, Utrecht University, 3584 CT, Utrecht, The Netherlands
- Centre for Living Technologies, Alliance TU/e, WUR, UU, UMC Utrecht, 3584 CB, Utrecht, The Netherlands
| | - Heleen N Sonneveld van Kooten
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Utrecht, University Medical Center, Utrecht University, 3584 CT, Utrecht, The Netherlands
- Centre for Living Technologies, Alliance TU/e, WUR, UU, UMC Utrecht, 3584 CB, Utrecht, The Netherlands
| | - Cornelis K van der Ent
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, 3584 EA, Utrecht, The Netherlands
| | - Jeffrey M Beekman
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, 3584 EA, Utrecht, The Netherlands.
- Regenerative Medicine Utrecht, University Medical Center, Utrecht University, 3584 CT, Utrecht, The Netherlands.
- Centre for Living Technologies, Alliance TU/e, WUR, UU, UMC Utrecht, 3584 CB, Utrecht, The Netherlands.
| | - Sam F B van Beuningen
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, 3584 EA, Utrecht, The Netherlands.
- Regenerative Medicine Utrecht, University Medical Center, Utrecht University, 3584 CT, Utrecht, The Netherlands.
- Centre for Living Technologies, Alliance TU/e, WUR, UU, UMC Utrecht, 3584 CB, Utrecht, The Netherlands.
| |
Collapse
|
25
|
Leyva-Díaz E, Wilson ES, López-Bendito G. Development has the answer: Unraveling psychiatric disorders via thalamocortical organoids. Cell Stem Cell 2024; 31:283-284. [PMID: 38458174 DOI: 10.1016/j.stem.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 03/10/2024]
Abstract
Dissecting the role of the thalamus in neuropsychiatric disorders requires new models to analyze complex genetic interactions. In this issue of Cell Stem Cell, Shin et al. use patient-derived thalamocortical organoids to investigate 22q11.2 microdeletion impact on thalamic development, revealing significant transcriptional dysregulation linked to psychiatric disorders.
Collapse
Affiliation(s)
- Eduardo Leyva-Díaz
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), San Juan de Alicante, Alicante, Spain
| | - Emily S Wilson
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), San Juan de Alicante, Alicante, Spain
| | - Guillermina López-Bendito
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), San Juan de Alicante, Alicante, Spain.
| |
Collapse
|
26
|
Caipa Garcia AL, Kucab JE, Al-Serori H, Beck RSS, Bellamri M, Turesky RJ, Groopman JD, Francies HE, Garnett MJ, Huch M, Drost J, Zilbauer M, Arlt VM, Phillips DH. Tissue Organoid Cultures Metabolize Dietary Carcinogens Proficiently and Are Effective Models for DNA Adduct Formation. Chem Res Toxicol 2024; 37:234-247. [PMID: 38232180 PMCID: PMC10880098 DOI: 10.1021/acs.chemrestox.3c00255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/19/2024]
Abstract
Human tissue three-dimensional (3D) organoid cultures have the potential to reproduce in vitro the physiological properties and cellular architecture of the organs from which they are derived. The ability of organoid cultures derived from human stomach, liver, kidney, and colon to metabolically activate three dietary carcinogens, aflatoxin B1 (AFB1), aristolochic acid I (AAI), and 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), was investigated. In each case, the response of a target tissue (liver for AFB1; kidney for AAI; colon for PhIP) was compared with that of a nontarget tissue (gastric). After treatment cell viabilities were measured, DNA damage response (DDR) was determined by Western blotting for p-p53, p21, p-CHK2, and γ-H2AX, and DNA adduct formation was quantified by mass spectrometry. Induction of the key xenobiotic-metabolizing enzymes (XMEs) CYP1A1, CYP1A2, CYP3A4, and NQO1 was assessed by qRT-PCR. We found that organoids from different tissues can activate AAI, AFB1, and PhIP. In some cases, this metabolic potential varied between tissues and between different cultures of the same tissue. Similarly, variations in the levels of expression of XMEs were observed. At comparable levels of cytotoxicity, organoids derived from tissues that are considered targets for these carcinogens had higher levels of adduct formation than a nontarget tissue.
Collapse
Affiliation(s)
- Angela L. Caipa Garcia
- Department
of Analytical, Environmental and Forensic Sciences, School of Cancer
& Pharmaceutical Sciences, King’s
College London, London SE1 9NH, U.K.
| | - Jill E. Kucab
- Department
of Analytical, Environmental and Forensic Sciences, School of Cancer
& Pharmaceutical Sciences, King’s
College London, London SE1 9NH, U.K.
| | - Halh Al-Serori
- Department
of Analytical, Environmental and Forensic Sciences, School of Cancer
& Pharmaceutical Sciences, King’s
College London, London SE1 9NH, U.K.
| | - Rebekah S. S. Beck
- Department
of Analytical, Environmental and Forensic Sciences, School of Cancer
& Pharmaceutical Sciences, King’s
College London, London SE1 9NH, U.K.
| | - Madjda Bellamri
- Department
of Medicinal Chemistry, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Robert J. Turesky
- Department
of Medicinal Chemistry, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - John D. Groopman
- Department
of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, United States
| | | | | | - Meritxell Huch
- Max
Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Jarno Drost
- Princess
Máxima Center for Pediatric Oncology, Oncode Institute, 3584
CS Utrecht, The Netherlands
| | - Matthias Zilbauer
- Department
of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, U.K.
| | - Volker M. Arlt
- Department
of Analytical, Environmental and Forensic Sciences, School of Cancer
& Pharmaceutical Sciences, King’s
College London, London SE1 9NH, U.K.
| | - David H. Phillips
- Department
of Analytical, Environmental and Forensic Sciences, School of Cancer
& Pharmaceutical Sciences, King’s
College London, London SE1 9NH, U.K.
| |
Collapse
|
27
|
Pianigiani G, Roccio M. Inner Ear Organoids: Strengths and Limitations. J Assoc Res Otolaryngol 2024; 25:5-11. [PMID: 38334886 PMCID: PMC10907556 DOI: 10.1007/s10162-024-00929-2] [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: 11/27/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Inner ear organoids derived from differentiation of human pluripotent stem cells have recently gained momentum as tools to study inner ear development and developmental defects. An additional exciting aspect about this technology is represented by its translational potential, specifically, the use of organoids to validate therapeutics for hearing and balance restoration on human/patient-specific cells. This latter aspect will be briefly discussed here including opportunities and current limitations.
Collapse
Affiliation(s)
- Giulia Pianigiani
- Institute for Maternal and Child Health - I.R.C.C.S. "Burlo Garofolo", Trieste, Italy
| | - Marta Roccio
- Inner Ear Stem Cell Lab, Department Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich and University of Zurich, USZ Campus WAGI18, Wagistrasse 18, 8952, Schlieren, Switzerland.
| |
Collapse
|
28
|
Rispoli P, Scandiuzzi Piovesan T, Decorti G, Stocco G, Lucafò M. iPSCs as a groundbreaking tool for the study of adverse drug reactions: A new avenue for personalized therapy. WIREs Mech Dis 2024; 16:e1630. [PMID: 37770042 DOI: 10.1002/wsbm.1630] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/10/2023] [Accepted: 09/07/2023] [Indexed: 10/03/2023]
Abstract
Induced pluripotent stem cells (iPSCs), obtained by reprogramming different somatic cell types, represent a promising tool for the study of drug toxicities, especially in the context of personalized medicine. Indeed, these cells retain the same genetic heritage of the donor, allowing the development of personalized models. In addition, they represent a useful tool for the study of adverse drug reactions (ADRs) in special populations, such as pediatric patients, which are often poorly represented in clinical trials due to ethical issues. Particularly, iPSCs can be differentiated into any tissue of the human body, following several protocols which use different stimuli to induce specific differentiation processes. Differentiated cells also maintain the genetic heritage of the donor, and therefore are suitable for personalized pharmacological studies; moreover, iPSC-derived differentiated cells are a valuable tool for the investigation of the mechanisms underlying the physiological differentiation processes. iPSCs-derived organoids represent another important tool for the study of ADRs. Precisely, organoids are in vitro 3D models which better represent the native organ, both from a structural and a functional point of view. Moreover, in the same way as iPSC-derived 2D models, iPSC-derived organoids are appropriate personalized models since they retain the genetic heritage of the donor. In comparison to other in vitro models, iPSC-derived organoids present advantages in terms of versatility, patient-specificity, and ethical issues. This review aims to provide an updated report of the employment of iPSCs, and 2D and 3D models derived from these, for the study of ADRs. This article is categorized under: Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- Paola Rispoli
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | | | - Giuliana Decorti
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
| | - Gabriele Stocco
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
| | - Marianna Lucafò
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
29
|
Haider S, Knöfler M, Latos PA. Trophoblast Organoids as a Novel Tool to Study Human Placental Development and Function. Methods Mol Biol 2024; 2728:195-222. [PMID: 38019403 DOI: 10.1007/978-1-0716-3495-0_17] [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] [Indexed: 11/30/2023]
Abstract
The human placenta provides the site of exchange between the maternal and fetal bloodstreams, acts as an endocrine organ, and has immunological functions. The majority of pregnancy disorders including preeclampsia and fetal growth restriction have their roots in pathological placentation. Yet, the underlying molecular causes of these complications remain largely unknown, not least due to the lack of reliable in vitro models. Recent establishment of 2D human trophoblast stem cells and 3D trophoblast organoids has been a major advancement that opened new avenues for trophoblast research. Here we provide a protocol detailing isolation of cytotrophoblast from the first trimester human placenta, establishment of trophoblast organoids, their culture and differentiation conditions. Overall, we describe an in vitro system that offers an excellent model to study the molecular basis of placental development and disease.
Collapse
Affiliation(s)
- Sandra Haider
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Placental Development Group, Medical University of Vienna, Vienna, Austria.
| | - Martin Knöfler
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Placental Development Group, Medical University of Vienna, Vienna, Austria
| | - Paulina A Latos
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
30
|
Dos Reis RS, Selvam S, Wagner MCE, Ayyavoo V. Modeling HIV-1 Infection in CNS via Infected Monocytes Using Immunocompetent Brain Organoids. Methods Mol Biol 2024; 2807:261-270. [PMID: 38743234 DOI: 10.1007/978-1-0716-3862-0_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The development of 3D-organoid models has revolutionized the way diseases are studied. Recently, our brain organoid model has been shown to recapitulate in in vitro the human brain cytoarchitecture originally encountered in HIV-1 neuropathogenesis, allowing downstream applications. Infected monocytes, macrophages, and microglia are critically important immune cells for infection and dissemination of HIV-1 throughout brain during acute and chronic phase of the disease. Once in the brain parenchyma, long-lived infected monocytes/macrophages along with resident microglia contribute to the establishment of CNS latency in people with HIV (PWH). Hence, it is important to better understand how HIV-1 enters and establishes infection and latency in CNS to further develop cure strategies. Here we detailed an accessible protocol to incorporate monocytes (infected and/or labeled) as a model of transmigration of peripheral monocytes into brain organoids that can be applied to characterize HIV-1 neuroinvasion and virus dissemination.
Collapse
Affiliation(s)
- Roberta S Dos Reis
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sathish Selvam
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marc C E Wagner
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Velpandi Ayyavoo
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
31
|
Mishra I, Gupta K, Mishra R, Chaudhary K, Sharma V. An Exploration of Organoid Technology: Present Advancements, Applications, and Obstacles. Curr Pharm Biotechnol 2024; 25:1000-1020. [PMID: 37807405 DOI: 10.2174/0113892010273024230925075231] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/19/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Organoids are in vitro models that exhibit a three-dimensional structure and effectively replicate the structural and physiological features of human organs. The capacity to research complex biological processes and disorders in a controlled setting is laid out by these miniature organ-like structures. OBJECTIVES This work examines the potential applications of organoid technology, as well as the challenges and future directions associated with its implementation. It aims to emphasize the pivotal role of organoids in disease modeling, drug discovery, developmental biology, precision medicine, and fundamental research. METHODS The manuscript was put together by conducting a comprehensive literature review, which involved an in-depth evaluation of globally renowned scientific research databases. RESULTS The field of organoids has generated significant attention due to its potential applications in tissue development and disease modelling, as well as its implications for personalised medicine, drug screening, and cell-based therapies. The utilisation of organoids has proven to be effective in the examination of various conditions, encompassing genetic disorders, cancer, neurodevelopmental disorders, and infectious diseases. CONCLUSION The exploration of the wider uses of organoids is still in its early phases. Research shall be conducted to integrate 3D organoid systems as alternatives for current models, potentially improving both fundamental and clinical studies in the future.
Collapse
Affiliation(s)
- Isha Mishra
- Department of Pharmacy, Galgotias College of Pharmacy, Greater Noida, Uttar Pradesh, 201310, India
| | - Komal Gupta
- Department of Pharmacy, Galgotias College of Pharmacy, Greater Noida, Uttar Pradesh, 201310, India
| | - Raghav Mishra
- Department of Pharmacy, GLA University, Mathura, 281406, Uttar Pradesh, India
| | - Kajal Chaudhary
- Department of Pharmacy, GLA University, Mathura, 281406, Uttar Pradesh, India
| | - Vikram Sharma
- Department of Pharmacy, Galgotias College of Pharmacy, Greater Noida, Uttar Pradesh, 201310, India
| |
Collapse
|
32
|
Joo H, Min S, Cho SW. Advanced lung organoids for respiratory system and pulmonary disease modeling. J Tissue Eng 2024; 15:20417314241232502. [PMID: 38406820 PMCID: PMC10894554 DOI: 10.1177/20417314241232502] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
Amidst the recent coronavirus disease 2019 (COVID-19) pandemic, respiratory system research has made remarkable progress, particularly focusing on infectious diseases. Lung organoid, a miniaturized structure recapitulating lung tissue, has gained global attention because of its advantages over other conventional models such as two-dimensional (2D) cell models and animal models. Nevertheless, lung organoids still face limitations concerning heterogeneity, complexity, and maturity compared to the native lung tissue. To address these limitations, researchers have employed co-culture methods with various cell types including endothelial cells, mesenchymal cells, and immune cells, and incorporated bioengineering platforms such as air-liquid interfaces, microfluidic chips, and functional hydrogels. These advancements have facilitated applications of lung organoids to studies of pulmonary diseases, providing insights into disease mechanisms and potential treatments. This review introduces recent progress in the production methods of lung organoids, strategies for improving maturity, functionality, and complexity of organoids, and their application in disease modeling, including respiratory infection and pulmonary fibrosis.
Collapse
Affiliation(s)
- Hyebin Joo
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
| |
Collapse
|
33
|
Dortaj H, Azarpira N, Pakbaz S. Insight to Biofabrication of Liver Microtissues for Disease Modeling: Challenges and Opportunities. Curr Stem Cell Res Ther 2024; 19:1303-1311. [PMID: 37846577 DOI: 10.2174/011574888x257744231009071810] [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: 04/13/2023] [Revised: 08/26/2023] [Accepted: 09/13/2023] [Indexed: 10/18/2023]
Abstract
In the last decade, liver diseases with high mortality rates have become one of the most important health problems in the world. Organ transplantation is currently considered the most effective treatment for compensatory liver failure. An increasing number of patients and shortage of donors has led to the attention of reconstructive medicine methods researchers. The biggest challenge in the development of drugs effective in chronic liver disease is the lack of a suitable preclinical model that can mimic the microenvironment of liver problems. Organoid technology is a rapidly evolving field that enables researchers to reconstruct, evaluate, and manipulate intricate biological processes in vitro. These systems provide a biomimetic model for studying the intercellular interactions necessary for proper organ function and architecture in vivo. Liver organoids, formed by the self-assembly of hepatocytes, are microtissues and can exhibit specific liver characteristics for a long time in vitro. Hepatic organoids are identified as an impressive tool for evaluating potential cures and modeling liver diseases. Modeling various liver diseases, including tumors, fibrosis, non-alcoholic fatty liver, etc., allows the study of the effects of various drugs on these diseases in personalized medicine. Here, we summarize the literature relating to the hepatic stem cell microenvironment and the formation of liver Organoids.
Collapse
Affiliation(s)
- Hengameh Dortaj
- Department of Tissue Engineering and Applied Cell Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Pakbaz
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
34
|
Ryu S, Weber C, Chu PH, Ernest B, Jovanovic VM, Deng T, Slamecka J, Hong H, Jethmalani Y, Baskir HM, Inman J, Braisted J, Hirst MB, Simeonov A, Voss TC, Tristan CA, Singeç I. Stress-free cell aggregation by using the CEPT cocktail enhances embryoid body and organoid fitness. Biofabrication 2023; 16:015016. [PMID: 37972398 DOI: 10.1088/1758-5090/ad0d13] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/16/2023] [Indexed: 11/19/2023]
Abstract
Embryoid bodies (EBs) and self-organizing organoids derived from human pluripotent stem cells (hPSCs) recapitulate tissue development in a dish and hold great promise for disease modeling and drug development. However, current protocols are hampered by cellular stress and apoptosis during cell aggregation, resulting in variability and impaired cell differentiation. Here, we demonstrate that EBs and various organoid models (e.g., brain, gut, kidney) can be optimized by using the small molecule cocktail named CEPT (chroman 1, emricasan, polyamines, trans-ISRIB), a polypharmacological approach that ensures cytoprotection and cell survival. Application of CEPT for just 24 h during cell aggregation has long-lasting consequences affecting morphogenesis, gene expression, cellular differentiation, and organoid function. Various qualification methods confirmed that CEPT treatment enhanced experimental reproducibility and consistently improved EB and organoid fitness as compared to the widely used ROCK inhibitor Y-27632. Collectively, we discovered that stress-free cell aggregation and superior cell survival in the presence of CEPT are critical quality control determinants that establish a robust foundation for bioengineering complex tissue and organ models.
Collapse
Affiliation(s)
- Seungmi Ryu
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - Claire Weber
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - Pei-Hsuan Chu
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - Ben Ernest
- Rancho Biosciences, 16955 Via Del Campo, #200, San Diego, CA 92127, United States of America
| | - Vukasin M Jovanovic
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - Tao Deng
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - Jaroslav Slamecka
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - Hyenjong Hong
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - Yogita Jethmalani
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - Hannah M Baskir
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - Jason Inman
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - John Braisted
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - Marissa B Hirst
- Rancho Biosciences, 16955 Via Del Campo, #200, San Diego, CA 92127, United States of America
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - Ty C Voss
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - Carlos A Tristan
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| | - Ilyas Singeç
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, United States of America
| |
Collapse
|
35
|
Li H, Chen Z, Chen N, Fan Y, Xu Y, Xu X. Applications of lung cancer organoids in precision medicine: from bench to bedside. Cell Commun Signal 2023; 21:350. [PMID: 38057851 PMCID: PMC10698950 DOI: 10.1186/s12964-023-01332-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/25/2023] [Indexed: 12/08/2023] Open
Abstract
As the leading cause of cancer-related mortality, lung cancer continues to pose a menacing threat to human health worldwide. Lung cancer treatment options primarily rely on chemoradiotherapy, surgery, targeted therapy, or immunotherapy. Despite significant progress in research and treatment, the 5-year survival rate for lung cancer patients is only 10-20%. There is an urgent need to develop more reliable preclinical models and valid therapeutic approaches. Patient-derived organoids with highly reduced tumour heterogeneity have emerged as a promising model for high-throughput drug screening to guide treatment of lung cancer patients. Organoid technology offers a novel platform for disease modelling, biobanking and drug development. The expected benefit of organoids is for cancer patients as the subsequent precision medicine technology. Over the past few years, numerous basic and clinical studies have been conducted on lung cancer organoids, highlighting the significant contributions of this technique. This review comprehensively examines the current state-of-the-art technologies and applications relevant to the formation of lung cancer organoids, as well as the potential of organoids in precision medicine and drug testing. Video Abstract.
Collapse
Affiliation(s)
- Huihui Li
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zexin Chen
- Guangdong Research Center of Organoid Engineering and Technology, Guangzhou, 510535, Guangdong, China
| | - Ning Chen
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Department of Oncology, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yun Fan
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Yaping Xu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Xiaoling Xu
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| |
Collapse
|
36
|
von Coburg E, Dunst S. The adverse outcome pathway for breast cancer: a knowledge management framework bridging biomedicine and toxicology. Discov Oncol 2023; 14:223. [PMID: 38051394 DOI: 10.1007/s12672-023-00840-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/26/2023] [Indexed: 12/07/2023] Open
Abstract
Breast cancer is the most common cancer worldwide, with an estimated 2.3 million new cases diagnosed every year. Effective measures for cancer prevention and cancer therapy require a detailed understanding of the individual key disease mechanisms involved and their interactions at the molecular, cellular, tissue, organ, and organism level. In that regard, the rapid progress of biomedical and toxicological research in recent years now allows the pursuit of new approaches based on non-animal methods that provide greater mechanistic insight than traditional animal models and therefore facilitate the development of Adverse Outcome Pathways (AOPs) for human diseases. We performed a systematic review of the current state of published knowledge with regard to breast cancer to identify relevant key mechanisms for inclusion into breast cancer AOPs, i.e. decreased cell stiffness and decreased cell adhesion, and to concurrently map non-animal methods addressing these key events. We conclude that the broader sharing of expertise and methods between biomedical research and toxicology enabled by the AOP knowledge management framework can help to coordinate global research efforts and accelerate the transition to advanced non-animal methods, which, when combined into powerful method batteries, closely mimic human physiology and disease states without the need for animal testing.
Collapse
Affiliation(s)
- Elena von Coburg
- German Centre for the Protection of Laboratory Animals (Bf3R), Department Experimental Toxicology and ZEBET, German Federal Institute for Risk Assessment, Berlin, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Sebastian Dunst
- German Centre for the Protection of Laboratory Animals (Bf3R), Department Experimental Toxicology and ZEBET, German Federal Institute for Risk Assessment, Berlin, Germany.
| |
Collapse
|
37
|
Joo L, Jeong HY, Bae DH, Jee JH, Choi WH, Kim HY, Kim S, Yang DH, Gee HY, Jeon S, Roh YG, Yoo J. Prostaglandin F2α analogue, bimatoprost ameliorates colistin-induced nephrotoxicity. Biomed Pharmacother 2023; 168:115446. [PMID: 37918255 DOI: 10.1016/j.biopha.2023.115446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 11/04/2023] Open
Abstract
Colistin (polymyxin E) is an antibiotic that is effective against multidrug-resistant gram-negative bacteria. However, the high incidence of nephrotoxicity caused by colistin limits its clinical use. To identify compounds that might ameliorate colistin-induced nephrotoxicity, we obtained 1707 compounds from the Korea Chemical Bank and used a high-content screening (HCS) imaging-based assay. In this way, we found that bimatoprost (one of prostaglandin F2α analogue) ameliorated colistin-induced nephrotoxicity. To further assess the effects of bimatoprost on colistin-induced nephrotoxicity, we used in vitro and in vivo models. In cultured human proximal tubular cells (HK-2), colistin induced dose-dependent cytotoxicity. The number of terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL)-positive cells, indicative of apoptosis, was higher in colistin-treated cells, but this effect of colistin was ameliorated by cotreatment with bimatoprost. The generation of reactive oxygen species, assessed using 2,7-dichlorodihydrofluorescein diacetate, was less marked in cells treated with both colistin and bimatoprost than in those treated with colistin alone. Female C57BL/6 mice (n = 10 per group) that were intraperitoneally injected with colistin (10 mg/kg/12 hr) for 14 days showed high blood urea nitrogen and serum creatinine concentrations that were reduced by the coadministration of bimatoprost (0.5 mg/kg/12 hr). In addition, kidney injury molecule-1 (KIM1) and Neutrophil gelatinase-associated lipocalin (NGAL) expression also reduced by bimatoprost administration. Further investigation in tubuloid and kidney organoids also showed that bimatoprost attenuated the nephrotoxicity by colistin, showing dose-dependent reducing effect of KIM1 expression. In this study, we have identified bimatoprost, prostaglandin F2α analogue as a drug that ameliorates colistin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Lina Joo
- Department of Microbiology, CHA University School of Medicine, Seongnam, the Republic of Korea; CHA Organoid Research Center, CHA University, Seongnam, the Republic of Korea
| | - Hye Yun Jeong
- Division of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, the Republic of Korea
| | - Dong Hyuck Bae
- Department of Microbiology, CHA University School of Medicine, Seongnam, the Republic of Korea; CHA Organoid Research Center, CHA University, Seongnam, the Republic of Korea
| | - Joo Hyun Jee
- Department of Microbiology, CHA University School of Medicine, Seongnam, the Republic of Korea; CHA Organoid Research Center, CHA University, Seongnam, the Republic of Korea
| | - Woo Hee Choi
- Department of Microbiology, CHA University School of Medicine, Seongnam, the Republic of Korea; CHA Organoid Research Center, CHA University, Seongnam, the Republic of Korea; R&D Institute, ORGANOIDSCIENCES LTD., Seongnam, the Republic of Korea
| | - Hye-Youn Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, the Republic of Korea
| | - Sejoong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonnggi-do 13620, the Republic of Korea; Department of Internal Medicine, Seoul National University College of Medicine Seoul, 03080, the Republic of Korea
| | - Dong-Ho Yang
- Division of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, the Republic of Korea
| | - Heon Yung Gee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, the Republic of Korea
| | - SeongGyeong Jeon
- Department of Microbiology, CHA University School of Medicine, Seongnam, the Republic of Korea; CHA Organoid Research Center, CHA University, Seongnam, the Republic of Korea
| | - Yun-Gil Roh
- Program in Health Policy, Chung-Buk National University, Republic of Korea
| | - Jongman Yoo
- Department of Microbiology, CHA University School of Medicine, Seongnam, the Republic of Korea; CHA Organoid Research Center, CHA University, Seongnam, the Republic of Korea; R&D Institute, ORGANOIDSCIENCES LTD., Seongnam, the Republic of Korea.
| |
Collapse
|
38
|
Abdel Fattah AR, Grebenyuk S, de Rooij LPMH, Salmon I, Ranga A. Neuroepithelial organoid patterning is mediated by a neighborhood watch mechanism. Cell Rep 2023; 42:113334. [PMID: 38511989 DOI: 10.1016/j.celrep.2023.113334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/29/2023] [Accepted: 10/10/2023] [Indexed: 03/22/2024] Open
Abstract
During epithelial tissue patterning, morphogens operate across multiple length scales to instruct cell identities. However, how cell fate changes are coordinated over these scales to establish spatial organization remains poorly understood. Here, we use human neural tube organoids as models of epithelial patterning and develop an in silico approach to define conditions permissive to patterning. By systematically varying morphogen position, diffusivity, and fate-inducing concentration levels, we show that cells follow a "neighborhood watch" (NW) mechanism that is deterministically dictated by initial morphogen source positions, reflecting scale-invariant in vitro phenotypes. We define how the frequency and local bias of morphogen sources stabilize pattern orientation. The model predicts enhanced patterning through floor plate inhibition, and receptor-ligand interaction analysis of single-cell RNA sequencing (scRNA-seq) data identifies wingless-related integration site (WNT) and bone morphogenic protein (BMP) as inhibition modulators, which we validate in vitro. These results suggest that developing neuroepithelia employ NW-based mechanisms to organize morphogen sources, define cellular identity, and establish patterns.
Collapse
Affiliation(s)
- Abdel Rahman Abdel Fattah
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Sergei Grebenyuk
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Laura P M H de Rooij
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Idris Salmon
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Adrian Ranga
- Laboratory of Bioengineering and Morphogenesis, Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.
| |
Collapse
|
39
|
Gómez-Álvarez M, Agustina-Hernández M, Francés-Herrero E, Rodríguez-Eguren A, Bueno-Fernandez C, Cervelló I. Addressing Key Questions in Organoid Models: Who, Where, How, and Why? Int J Mol Sci 2023; 24:16014. [PMID: 37958996 PMCID: PMC10650475 DOI: 10.3390/ijms242116014] [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: 09/29/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Organoids are three-dimensional cellular structures designed to recreate the biological characteristics of the body's native tissues and organs in vitro. There has been a recent surge in studies utilizing organoids due to their distinct advantages over traditional two-dimensional in vitro approaches. However, there is no consensus on how to define organoids. This literature review aims to clarify the concept of organoids and address the four fundamental questions pertaining to organoid models: (i) What constitutes organoids?-The cellular material. (ii) Where do organoids grow?-The extracellular scaffold. (iii) How are organoids maintained in vitro?-Via the culture media. (iv) Why are organoids suitable in vitro models?-They represent reproducible, stable, and scalable models for biological applications. Finally, this review provides an update on the organoid models employed within the female reproductive tract, underscoring their relevance in both basic biology and clinical applications.
Collapse
Affiliation(s)
- María Gómez-Álvarez
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
| | - Marcos Agustina-Hernández
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
| | - Emilio Francés-Herrero
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
- Department of Pediatrics, Obstetrics and Gynecology, Universitat de València, 46010 Valencia, Spain
| | - Adolfo Rodríguez-Eguren
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
| | - Clara Bueno-Fernandez
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
- Department of Pediatrics, Obstetrics and Gynecology, Universitat de València, 46010 Valencia, Spain
| | - Irene Cervelló
- Instituto de Investigación Sanitaria La Fe (IIS La Fe), IVI Foundation, IVIRMA Global Research Alliance, 46026 Valencia, Spain; (M.G.-Á.); (M.A.-H.); (E.F.-H.); (A.R.-E.); (C.B.-F.)
| |
Collapse
|
40
|
Diensthuber M, Stöver T. [Organoids-the key to novel therapies for the inner ear? German version]. HNO 2023; 71:702-707. [PMID: 37845538 DOI: 10.1007/s00106-023-01366-y] [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] [Accepted: 08/21/2023] [Indexed: 10/18/2023]
Abstract
The sensitivity and the complexity of the human inner ear in conjunction with the lack of regenerative capacity are the main reasons for hearing loss and tinnitus. Progress in the development of protective and regenerative therapies for the inner ear often failed in the past not least due to the fact that no suitable model systems for cell biological and pharmacological in vitro studies were available. A novel technology for creating "mini-organs", so-called organoids, could solve this problem and has now also reached inner ear research. It makes it possible to produce inner ear organoids from cochlear stem/progenitor cells, embryonic and induced pluripotent stem cells that mimic the structural characteristics and functional properties of the natural inner ear. This review focuses on the biological basis of these inner ear organoids, the current state of research and the promising prospects that are now opening up for basic and translational inner ear research.
Collapse
Affiliation(s)
- Marc Diensthuber
- Klinik für Hals-Nasen-Ohrenheilkunde, Universitätsklinikum Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt/M., Deutschland.
| | - Timo Stöver
- Klinik für Hals-Nasen-Ohrenheilkunde, Universitätsklinikum Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt/M., Deutschland
| |
Collapse
|
41
|
Milián L, Molina P, Oliver-Ferrándiz M, Fernández-Sellers C, Monzó A, Sánchez-Sánchez R, Braza-Boils A, Mata M, Zorio E. Cadaveric Adipose-Derived Stem Cells for Regenerative Medicine and Research. Int J Mol Sci 2023; 24:15696. [PMID: 37958680 PMCID: PMC10647636 DOI: 10.3390/ijms242115696] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Advances in regenerative medicine have enabled the search for new solutions to current health problems in so far unexplored fields. Thus, we focused on cadaveric subcutaneous fat as a promising source of adipose-derived stem cells (ADSCs) that have potential to differentiate into different cell lines. With this aim, we isolated and characterized ADSCs from cadaveric samples with a postmortem interval ranging from 30 to 55 h and evaluated their ability to differentiate into chondrocytes or osteocytes. A commercial ADSC line was used as reference. Morphological and protein expression analyses were used to confirm the final stage of differentiation. Eight out of fourteen samples from patients were suitable to complete the whole protocol. Cadaveric ADSCs exhibited features of stem cells based upon several markers: CD29 (84.49 ± 14.07%), CD105 (94.38 ± 2.09%), and CD44 (99.77 ± 0.32%). The multiparametric assessment of differentiation confirmed the generation of stable lines of chondrocytes and osteocytes. In conclusion, we provide evidence supporting the feasibility of obtaining viable postmortem human subcutaneous fat ADSCs with potential application in tissue engineering and research fields.
Collapse
Affiliation(s)
- Lara Milián
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain; (L.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Pilar Molina
- Department of Pathology, Instituto de Medicina Legal y Ciencias Forenses, 46010 Valencia, Spain; (P.M.); (C.F.-S.); (A.M.)
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (R.S.-S.); (A.B.-B.); (E.Z.)
| | - María Oliver-Ferrándiz
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain; (L.M.)
| | - Carlos Fernández-Sellers
- Department of Pathology, Instituto de Medicina Legal y Ciencias Forenses, 46010 Valencia, Spain; (P.M.); (C.F.-S.); (A.M.)
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (R.S.-S.); (A.B.-B.); (E.Z.)
| | - Ana Monzó
- Department of Pathology, Instituto de Medicina Legal y Ciencias Forenses, 46010 Valencia, Spain; (P.M.); (C.F.-S.); (A.M.)
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (R.S.-S.); (A.B.-B.); (E.Z.)
| | - Rafael Sánchez-Sánchez
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (R.S.-S.); (A.B.-B.); (E.Z.)
| | - Aitana Braza-Boils
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (R.S.-S.); (A.B.-B.); (E.Z.)
- CIBERCV, Center for Biomedical Network Research on Cardiovascular Diseases, 28015 Madrid, Spain
| | - Manuel Mata
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain; (L.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Esther Zorio
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (R.S.-S.); (A.B.-B.); (E.Z.)
- CIBERCV, Center for Biomedical Network Research on Cardiovascular Diseases, 28015 Madrid, Spain
- Inherited Cardiac Diseases Unit, Cardiology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Department of Medicine, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain
| |
Collapse
|
42
|
Barroso M, Monaghan MG, Niesner R, Dmitriev RI. Probing organoid metabolism using fluorescence lifetime imaging microscopy (FLIM): The next frontier of drug discovery and disease understanding. Adv Drug Deliv Rev 2023; 201:115081. [PMID: 37647987 PMCID: PMC10543546 DOI: 10.1016/j.addr.2023.115081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/20/2023] [Accepted: 08/24/2023] [Indexed: 09/01/2023]
Abstract
Organoid models have been used to address important questions in developmental and cancer biology, tissue repair, advanced modelling of disease and therapies, among other bioengineering applications. Such 3D microenvironmental models can investigate the regulation of cell metabolism, and provide key insights into the mechanisms at the basis of cell growth, differentiation, communication, interactions with the environment and cell death. Their accessibility and complexity, based on 3D spatial and temporal heterogeneity, make organoids suitable for the application of novel, dynamic imaging microscopy methods, such as fluorescence lifetime imaging microscopy (FLIM) and related decay time-assessing readouts. Several biomarkers and assays have been proposed to study cell metabolism by FLIM in various organoid models. Herein, we present an expert-opinion discussion on the principles of FLIM and PLIM, instrumentation and data collection and analysis protocols, and general and emerging biosensor-based approaches, to highlight the pioneering work being performed in this field.
Collapse
Affiliation(s)
- Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Michael G Monaghan
- Department of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin 02, Ireland
| | - Raluca Niesner
- Dynamic and Functional In Vivo Imaging, Freie Universität Berlin and Biophysical Analytics, German Rheumatism Research Center, Berlin, Germany
| | - Ruslan I Dmitriev
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, 9000 Ghent, Belgium; Ghent Light Microscopy Core, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
43
|
Debruyne AC, Okkelman IA, Dmitriev RI. Balance between the cell viability and death in 3D. Semin Cell Dev Biol 2023; 144:55-66. [PMID: 36117019 DOI: 10.1016/j.semcdb.2022.09.005] [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: 04/19/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 11/25/2022]
Abstract
Cell death is a phenomenon, frequently perceived as an absolute event for cell, tissue and the organ. However, the rising popularity and complexity of such 3D multicellular 'tissue building blocks' as heterocellular spheroids, organoids, and 'assembloids' prompts to revise the definition and quantification of cell viability and death. It raises several questions on the overall viability of all the cells within 3D volume and on choosing the appropriate, continuous, and non-destructive viability assay enabling for a single-cell analysis. In this review, we look at cell viability and cell death modalities with attention to the intrinsic features of such 3D models as spheroids, organoids, and bioprints. Furthermore, we look at emerging and promising methodologies, which can help define and understand the balance between cell viability and death in dynamic and complex 3D environments. We conclude that the recent innovations in biofabrication, biosensor probe development, and fluorescence microscopy can help answer these questions.
Collapse
Affiliation(s)
- Angela C Debruyne
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent 9000, Belgium
| | - Irina A Okkelman
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent 9000, Belgium
| | - Ruslan I Dmitriev
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent 9000, Belgium.
| |
Collapse
|
44
|
Muniyandi P, O’Hern C, Popa MA, Aguirre A. Biotechnological advances and applications of human pluripotent stem cell-derived heart models. Front Bioeng Biotechnol 2023; 11:1214431. [PMID: 37560538 PMCID: PMC10407810 DOI: 10.3389/fbioe.2023.1214431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023] Open
Abstract
In recent years, significant biotechnological advancements have been made in engineering human cardiac tissues and organ-like models. This field of research is crucial for both basic and translational research due to cardiovascular disease being the leading cause of death in the developed world. Additionally, drug-associated cardiotoxicity poses a major challenge for drug development in the pharmaceutical and biotechnological industries. Progress in three-dimensional cell culture and microfluidic devices has enabled the generation of human cardiac models that faithfully recapitulate key aspects of human physiology. In this review, we will discuss 3D pluripotent stem cell (PSC)-models of the human heart, such as engineered heart tissues and organoids, and their applications in disease modeling and drug screening.
Collapse
Affiliation(s)
- Priyadharshni Muniyandi
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
| | - Colin O’Hern
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
| | - Mirel Adrian Popa
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
- Institute of Cellular Biology and Pathology Nicolae Simionescu, Bucharest, Romania
| | - Aitor Aguirre
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
45
|
Shevade K, Peddada S, Mader K, Przybyla L. Functional genomics in stem cell models: considerations and applications. Front Cell Dev Biol 2023; 11:1236553. [PMID: 37554308 PMCID: PMC10404852 DOI: 10.3389/fcell.2023.1236553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/13/2023] [Indexed: 08/10/2023] Open
Abstract
Protocols to differentiate human pluripotent stem cells have advanced in terms of cell type specificity and tissue-level complexity over the past 2 decades, which has facilitated human disease modeling in the most relevant cell types. The ability to generate induced PSCs (iPSCs) from patients further enables the study of disease mutations in an appropriate cellular context to reveal the mechanisms that underlie disease etiology and progression. As iPSC-derived disease models have improved in robustness and scale, they have also been adopted more widely for use in drug screens to discover new therapies and therapeutic targets. Advancement in genome editing technologies, in particular the discovery of CRISPR-Cas9, has further allowed for rapid development of iPSCs containing disease-causing mutations. CRISPR-Cas9 technologies have now evolved beyond creating single gene edits, aided by the fusion of inhibitory (CRISPRi) or activation (CRISPRa) domains to a catalytically dead Cas9 protein, enabling inhibition or activation of endogenous gene loci. These tools have been used in CRISPR knockout, CRISPRi, or CRISPRa screens to identify genetic modifiers that synergize or antagonize with disease mutations in a systematic and unbiased manner, resulting in identification of disease mechanisms and discovery of new therapeutic targets to accelerate drug discovery research. However, many technical challenges remain when applying large-scale functional genomics approaches to differentiated PSC populations. Here we review current technologies in the field of iPSC disease modeling and CRISPR-based functional genomics screens and practical considerations for implementation across a range of modalities, applications, and disease areas, as well as explore CRISPR screens that have been performed in iPSC models to-date and the insights and therapies these screens have produced.
Collapse
Affiliation(s)
- Kaivalya Shevade
- Laboratory for Genomics Research, San Francisco, CA, United States
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, United States
| | - Sailaja Peddada
- Laboratory for Genomics Research, San Francisco, CA, United States
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, United States
| | - Karl Mader
- Laboratory for Genomics Research, San Francisco, CA, United States
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, United States
| | - Laralynne Przybyla
- Laboratory for Genomics Research, San Francisco, CA, United States
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
46
|
Perez-Luz S, Matamala N, Gomez-Mariano G, Janciauskiene S, Martínez-Delgado B. NAFLD and AATD Are Two Diseases with Unbalanced Lipid Metabolism: Similarities and Differences. Biomedicines 2023; 11:1961. [PMID: 37509601 PMCID: PMC10377048 DOI: 10.3390/biomedicines11071961] [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: 05/26/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a type of steatosis commonly associated with obesity, dyslipidemia, hypertension, and diabetes. Other diseases such as inherited alpha-1 antitrypsin deficiency (AATD) have also been related to the development of liver steatosis. The primary reasons leading to hepatic lipid deposits can be genetic and epigenetic, and the outcomes range from benign steatosis to liver failure, as well as to extrahepatic diseases. Progressive hepatocellular damage and dysregulated systemic immune responses can affect extrahepatic organs, specifically the heart and lungs. In this review, we discuss the similarities and differences between the molecular pathways of NAFLD and AATD, and the putative value of hepatic organoids as novel models to investigate the physio pathological mechanisms of liver steatosis.
Collapse
Affiliation(s)
- Sara Perez-Luz
- Molecular Genetics Unit, Institute of Rare Diseases Research (IIER), Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain
| | - Nerea Matamala
- Molecular Genetics Unit, Institute of Rare Diseases Research (IIER), Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain
| | - Gema Gomez-Mariano
- Molecular Genetics Unit, Institute of Rare Diseases Research (IIER), Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain
| | - Sabina Janciauskiene
- Department of Respiratory Medicine and Infectious Diseases, Biomedical Research in Endstage and Obstructive Lung Disease Hannover BREATH, Member of the German Center for Lung Research DZL, Hannover Medical School, 30625 Hannover, Germany
| | - Beatriz Martínez-Delgado
- Molecular Genetics Unit, Institute of Rare Diseases Research (IIER), Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain
- CIBER de Enfermedades Raras, Instituto de Salud Carlos III, CIBERER U758, 28029 Madrid, Spain
| |
Collapse
|
47
|
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
|
48
|
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: 13] [Impact Index Per Article: 6.5] [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
|
49
|
Chohra I, Giri S, Malgrange B. Generation of a Well-Characterized Homozygous Chromodomain-Helicase-DNA-Binding Protein 4 G1003D Mutant hESC Line Using CRISPR/eCas9 (ULIEGEe001-A-1). Int J Mol Sci 2023; 24:10543. [PMID: 37445725 DOI: 10.3390/ijms241310543] [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: 05/26/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
The chromatin remodeler Chromodomain-helicase-DNA-binding protein 4 (CHD4) is crucial for the development of multiple organ systems. Functional mutations of CHD4 have recently been described in a developmental disorder, namely Siffrim-Hitz-Weiss syndrome (SIHIWES). Herein, we have generated a homozygous CHD4G1003D hESC line (WAe025-A-1) using CRISPR/eCas9-based gene editing in the WA-25 hESC line. The edited hESC line maintains normal karyotype, pluripotency, and ability to differentiate into three germ layers. This cell line will be a valuable resource for studying the functional role of CHD4 during the development and disease modeling of SIHIWES in vitro.
Collapse
Affiliation(s)
- Ilyas Chohra
- Developmental Neurobiology Unit, GIGA-Stem Cells, Av Hippocrate, 15 B-4000 Liege, Belgium
| | - Subhajit Giri
- Developmental Neurobiology Unit, GIGA-Stem Cells, Av Hippocrate, 15 B-4000 Liege, Belgium
| | - Brigitte Malgrange
- Developmental Neurobiology Unit, GIGA-Stem Cells, Av Hippocrate, 15 B-4000 Liege, Belgium
| |
Collapse
|
50
|
Krawczyk E, Kitlińska J. Preclinical Models of Neuroblastoma-Current Status and Perspectives. Cancers (Basel) 2023; 15:3314. [PMID: 37444423 PMCID: PMC10340830 DOI: 10.3390/cancers15133314] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Preclinical in vitro and in vivo models remain indispensable tools in cancer research. These classic models, including two- and three-dimensional cell culture techniques and animal models, are crucial for basic and translational studies. However, each model has its own limitations and typically does not fully recapitulate the course of the human disease. Therefore, there is an urgent need for the development of novel, advanced systems that can allow for efficient evaluation of the mechanisms underlying cancer development and progression, more accurately reflect the disease pathophysiology and complexity, and effectively inform therapeutic decisions for patients. Preclinical models are especially important for rare cancers, such as neuroblastoma, where the availability of patient-derived specimens that could be used for potential therapy evaluation and screening is limited. Neuroblastoma modeling is further complicated by the disease heterogeneity. In this review, we present the current status of preclinical models for neuroblastoma research, discuss their development and characteristics emphasizing strengths and limitations, and describe the necessity of the development of novel, more advanced and clinically relevant approaches.
Collapse
Affiliation(s)
- Ewa Krawczyk
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Joanna Kitlińska
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|