1
|
Madrid MF, Mendoza EN, Padilla AL, Choquenaira-Quispe C, de Jesus Guimarães C, de Melo Pereira JV, Barros-Nepomuceno FWA, Lopes Dos Santos I, Pessoa C, de Moraes Filho MO, Rocha DD, Ferreira PMP. In vitro models to evaluate multidrug resistance in cancer cells: Biochemical and morphological techniques and pharmacological strategies. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2025; 28:1-27. [PMID: 39363148 DOI: 10.1080/10937404.2024.2407452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
The overexpression of ATP-binding cassette (ABC) transporters contributes to the failure of chemotherapies and symbolizes a great challenge in oncology, associated with the adaptation of tumor cells to anticancer drugs such that these transporters become less effective, a mechanism known as multidrug resistance (MDR). The aim of this review is to present the most widely used methodologies for induction and comprehension of in vitro models for detection of multidrug-resistant (MDR) modulators or inhibitors, including biochemical and morphological techniques for chemosensitivity studies. The overexpression of MDR proteins, predominantly, the subfamily glycoprotein-1 (P-gp or ABCB1) multidrug resistance, multidrug resistance-associated protein 1 (MRP1 or ABCCC1), multidrug resistance-associated protein 2 (MRP2 or ABCC2) and cancer resistance protein (ABCG2), in chemotherapy-exposed cancer lines have been established/investigated by several techniques. Amongst these techniques, the most used are (i) colorimetric/fluorescent indirect bioassays, (ii) rhodamine and efflux analysis, (iii) release of 3,30-diethyloxacarbocyanine iodide by fluorescence microscopy and flow cytometry to measure P-gp function and other ABC transporters, (iv) exclusion of calcein-acetoxymethylester, (v) ATPase assays to distinguish types of interaction with ABC transporters, (vi) morphology to detail phenotypic characteristics in transformed cells, (vii) molecular testing of resistance-related proteins (RT-qPCR) and (viii) 2D and 3D models, (ix) organoids, and (x) microfluidic technology. Then, in vitro models for detecting chemotherapy MDR cells to assess innovative therapies to modulate or inhibit tumor cell growth and overcome clinical resistance. It is noteworthy that different therapies including anti-miRNAs, antibody-drug conjugates (to natural products), and epigenetic modifications were also considered as promising alternatives, since currently no anti-MDR therapies are able to improve patient quality of life. Therefore, there is also urgency for new clinical markers of resistance to more reliably reflect in vivo effectiveness of novel antitumor drugs.
Collapse
Affiliation(s)
- Maria Fernanda Madrid
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Eleicy Nathaly Mendoza
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Ana Lizeth Padilla
- Pharmaceutical Sciences, Faculty of Pharmacy, Dentistry, and Nursing, Federal University of Ceará, Fortaleza, Brazil
| | - Celia Choquenaira-Quispe
- Pharmaceutical Sciences, Faculty of Pharmacy, Dentistry, and Nursing, Federal University of Ceará, Fortaleza, Brazil
- Catholic University of Santa María, Arequipa, Perú
| | - Celina de Jesus Guimarães
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - João Victor de Melo Pereira
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | | | - Ingredy Lopes Dos Santos
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| | - Claudia Pessoa
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Manoel Odorico de Moraes Filho
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Danilo Damasceno Rocha
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| |
Collapse
|
2
|
Abdulla A, Yan H, Chen S, Wu L, Chen XS, Zhang Y, Zhang M, Zhuang TY, Ahmad KZ, Lin J, Ding X, Jiang L. A multichannel microfluidic device for revealing the neurotoxic effects of Bisphenol S on cerebral organoids under low-dose constant exposure. Biosens Bioelectron 2025; 267:116754. [PMID: 39332252 DOI: 10.1016/j.bios.2024.116754] [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: 06/11/2024] [Revised: 08/20/2024] [Accepted: 09/04/2024] [Indexed: 09/29/2024]
Abstract
Bisphenol S is a widely used plasticizer in manufacturing daily supplies, while little was known about its adverse effect on human health, especially on fetal brain development. Due to the complexity and subtlety of the brain, it remains challenging to reveal the hazardous effects of environmental pollution on human fetal brain development. Taking advantage of stem cell application, cerebral organoids generated from stem cells are becoming powerful tools for understanding brain development and drug toxicity testing models. Here, we developed a microfluidic chip for cerebral organoid culturing to reveal the neurotoxicity of low-dose constant BPS exposure on cerebral organoids. The organoids in our microfluidic system could be continuously cultured for 34 days and expressed all the essential properties of the cerebral organoids. Exposure to BPS was initiated from day 20 for concessive two weeks. The neurotoxic effects were evaluated by immunofluorescence staining and proteomics, and verified by quantitative real-time PCR. Our results indicated BPS exposure would inhibit neuron differentiation, hinder the Wnt signaling pathway, and cause alteration of signaling molecule expressions in brain regionalization. Even exposure to a low dose of BPS constantly might cause neurotoxicity during fetal brain development. Altogether, the multichannel microfluidic chip offers a general platform technique to reveal the effects of different hazardous chemicals on cerebral organoids.
Collapse
Affiliation(s)
- Aynur Abdulla
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200092, China; State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Haoni Yan
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Shujin Chen
- Ministry of Education, Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Leqi Wu
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xu-Sen Chen
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yizhi Zhang
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Manlin Zhang
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Tsz Yui Zhuang
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Khan Zara Ahmad
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institute, Sweden
| | - Jinjin Lin
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China; School of Psychology, Shaanxi Normal University, Xi' an 710062, Shaanxi Province, China
| | - Xianting Ding
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200092, China; State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200092, China.
| |
Collapse
|
3
|
Jiao J, Xu D, Kong Y, Cao Y, Wang L, Hong Y, Li L, Gao C, Liu J, Zhang G, Zhou J, Dai J, Lu Z, Liu Y, Wang Y, Zhang Z. circFKBP8(5S,6)-encoded protein as a novel endogenous regulator in major depressive disorder by inhibiting glucocorticoid receptor nucleus translocation. Sci Bull (Beijing) 2024; 69:3826-3831. [PMID: 38945750 DOI: 10.1016/j.scib.2024.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/28/2024] [Accepted: 06/04/2024] [Indexed: 07/02/2024]
Affiliation(s)
- Jiao Jiao
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210009, China
| | - Dandan Xu
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210009, China; Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yan Kong
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Yujia Cao
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210009, China
| | - Liyuan Wang
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yuan Hong
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ling Li
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210009, China
| | - Chenyu Gao
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210009, China; Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jing Liu
- Department of Anesthesiology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen 518027, China; Shenzhen Key Laboratory for Molecular Biology of Neural Development, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Gaojia Zhang
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210009, China
| | - Jiangning Zhou
- CAS Key Laboratory of Brain Function and Diseases, Life Science School, University of Science and Technology of China, Hefei 230026, China
| | - Ji Dai
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518055, China
| | - Zhonghua Lu
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518055, China
| | - Yan Liu
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yutian Wang
- Faculty of Life and Health Sciences, Shenzhen Institutite of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Medicine and DM Center for Brain Health, University of British Columbia, Vancouver V6T 1Z4, Canada.
| | - Zhijun Zhang
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210009, China; Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
4
|
Jain S, Bhatt J, Gupta S, Bhatia DD. Nanotechnology at the crossroads of stem cell medicine. Biomater Sci 2024; 13:161-178. [PMID: 39584588 DOI: 10.1039/d4bm01257g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Nanotechnology in stem cell medicine is an interdisciplinary field which has gained a lot of interest recently. This domain addresses key challenges associated with stem cell medicine such as cell isolation, targeted delivery, and tracking. Nanotechnology-based approaches, including magnetic cell sorting, fluorescent tagging, and drug or biomolecule conjugation for delivery, have enhanced precision in stem cell isolation and guided cell migration, increasing the therapeutic potential. Recent studies have focused on using nanomaterials and scaffolds to drive stem cell differentiation by activating specific molecular pathways, achieved through embedding biomolecules within the scaffold or through the scaffold's material composition and structure alone. These innovations hold promise in therapeutic applications across various diseases, including cancer stem cell targeting, neurodegenerative disorders, pre-eclampsia, cardiovascular conditions, and organoid development. This review examines recent advancements in the field, explores potential applications like biosensors and nanochips, and highlights the challenges and research gaps.
Collapse
Affiliation(s)
- Sweny Jain
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Palaj, Gujarat, 382355, India.
| | - Jay Bhatt
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Palaj, Gujarat, 382355, India.
| | - Sharad Gupta
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Palaj, Gujarat, 382355, India.
| | - Dhiraj Devidas Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Palaj, Gujarat, 382355, India.
| |
Collapse
|
5
|
Xu L, Zhang Y, Chen X, Hong Y, Zhang X, Hu H, Han X, Zou X, Xu M, Zhu W, Liu Y. Human Brain Organoids Model Abnormal Prenatal Neural Development Induced by Thermal Stimulation. Cell Prolif 2024:e13777. [PMID: 39668124 DOI: 10.1111/cpr.13777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/17/2024] [Accepted: 11/05/2024] [Indexed: 12/14/2024] Open
Abstract
The developing human foetal brain is sensitive to thermal stimulation during pregnancy. However, the mechanisms by which heat exposure affects human foetal brain development remain unclear, largely due to the lack of appropriate research models for studying thermal stimulation. To address this, we have developed a periodic heating model based on brain organoids derived from human pluripotent stem cells. The model recapitulated neurodevelopmental disruptions under prenatal heat exposure at the early stages, providing a paradigm for studying the altered neurodevelopment under environmental stimulation. Our study found that periodic heat exposure led to decreased size and impaired neural tube development in the brain organoids. Bulk RNA-seq analysis revealed that the abnormal WNT signalling pathway and the reduction of G2/M progenitor cells might be involved in heat stimulation. Further investigation revealed increased neural differentiation and decreased proliferation under heat stimulation, indicating that periodic heat exposure might lead to abnormal brain development by altering key developmental processes. Hence, our model of periodically heating brain organoids provides a platform for modelling the effects of maternal fever on foetal brain development and could be extended to applications in neurodevelopmental disorders intervention.
Collapse
Affiliation(s)
- Lei Xu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Department of Neurology, Affiliated Zhongda Hospital, Southeast University, Nanjing, China
- Institute of Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Yufan Zhang
- Institute of Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Xingyi Chen
- Institute of Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Yuan Hong
- Institute of Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Xu Zhang
- Institute of Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Hao Hu
- Institute of Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Xiao Han
- Institute of Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Xiao Zou
- Institute of Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Min Xu
- Institute of Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Wanying Zhu
- Institute of Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Yan Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Department of Neurology, Affiliated Zhongda Hospital, Southeast University, Nanjing, China
- Institute of Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
6
|
Wang X, Tian Y, Chen H, Hou H, Hu Q. Airway organoids: 3D toxicology evaluation models in vitro of heated tobacco products for health risk. Toxicol In Vitro 2024; 104:105995. [PMID: 39667640 DOI: 10.1016/j.tiv.2024.105995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/14/2024]
Abstract
Cigarette smoking poses significant health risks, particularly to the airway, which consists predominantly of basal, club, and ciliated cells that are highly susceptible to damage from exogenous stimuli. Traditional in vitro toxicology relies on 2D cell cultures, which lack the structural complexity and functional relevance of airway architecture. As a novel category of tobacco products, the health implications of heated tobacco products (HTPs) remain largely unknown. To address this, 3D airway organoids were developed as a more physiologically relevant in vitro model for evaluating the toxicity of HTPs. Airway organoids derived from mouse lungs were induced to differentiate into various airway cell types and exposed to HTP aerosols. The exposure impaired organoid growth, reduced cell viability, and altered the proportions of secretory, basal, and ciliated cells, effectively replicating the complex cellular damage observed in vivo. Additionally, typical adverse outcomes, such as oxidative stress, inflammation, and genetic toxicity, were induced, paralleling findings from conventional 2D models. These results established the airway organoids as a viable alternative to animal testing for toxicological studies and offer critical insights into the respiratory health risks associated with HTPs.
Collapse
Affiliation(s)
- Xianglong Wang
- China National Tobacco Quality Supervision and Test Center, Zhengzhou 450001, China; Beijing Life Science Academy, Beijing 102209, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou 450001, China
| | - Yushan Tian
- China National Tobacco Quality Supervision and Test Center, Zhengzhou 450001, China; Beijing Life Science Academy, Beijing 102209, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou 450001, China
| | - Huan Chen
- China National Tobacco Quality Supervision and Test Center, Zhengzhou 450001, China; Beijing Life Science Academy, Beijing 102209, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou 450001, China.
| | - Hongwei Hou
- China National Tobacco Quality Supervision and Test Center, Zhengzhou 450001, China; Beijing Life Science Academy, Beijing 102209, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou 450001, China.
| | - Qingyuan Hu
- China National Tobacco Quality Supervision and Test Center, Zhengzhou 450001, China; Beijing Life Science Academy, Beijing 102209, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou 450001, China
| |
Collapse
|
7
|
Kuzinska MZ, Lin SYY, Klämbt V, Bufler P, Rezvani M. Ciliopathy organoid models: a comprehensive review. Am J Physiol Cell Physiol 2024; 327:C1604-C1625. [PMID: 39495251 DOI: 10.1152/ajpcell.00343.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/25/2024] [Accepted: 10/14/2024] [Indexed: 11/05/2024]
Abstract
Cilia are membrane-bound organelles found on the surface of most mammalian cell types and play numerous roles in human physiology and development, including osmo- and mechanosensation, as well as signal transduction. Ciliopathies are a large group of, usually rare, genetic disorders resulting from abnormal ciliary structure or ciliary dysfunction that have a high collective prevalence. Autosomal dominant or recessive polycystic kidney disease (ADPKD/ARPKD), Bardet-Biedl-Syndrome, and primary ciliary dyskinesia (PCD) are the most frequent etiologies. Rodent and zebrafish models have improved the understanding of ciliopathy pathophysiology. Yet, the limitations of these genetically modified animal strains include the inability to fully replicate the phenotypic heterogeneity found in humans, including variable multiorgan involvement. Organoids, self-assembled three-dimensional cell-based models derived from human induced pluripotent stem cells (iPSCs) or primary tissues, can recapitulate certain aspects of the development, architecture, and function of the target organ "in the dish." The potential of organoids to model patient-specific genotype-phenotype correlations has increased their popularity in ciliopathy research and led to the first preclinical organoid-based ciliopathy drug screens. This review comprehensively summarizes and evaluates current ciliopathy organoid models, focusing on kidney, airway, liver, and retinal organoids, as well as the specific methodologies used for their cultivation and for interrogating ciliary dysfunction.
Collapse
Affiliation(s)
- Matylda Zofia Kuzinska
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin Berlin-Campus Virchow Klinikum, Berlin, Germany
- Berlin School for Regenerative Therapies (BSRT), Berlin, Germany
| | - Sally Yuan-Yin Lin
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin Berlin-Campus Virchow Klinikum, Berlin, Germany
| | - Verena Klämbt
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin Berlin-Campus Virchow Klinikum, Berlin, Germany
- BIH Charité Clinician Scientist Program, BIH Biomedical Innovation Academy, Berlin Institute of Health at Charité-Universitätsmedizin, Berlin, Germany
| | - Philip Bufler
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin Berlin-Campus Virchow Klinikum, Berlin, Germany
- German Center for Child and Adolescent Health (DZKJ), Partner Site Berlin, Berlin, Germany
| | - Milad Rezvani
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin Berlin-Campus Virchow Klinikum, Berlin, Germany
- BIH Charité Clinician Scientist Program, BIH Biomedical Innovation Academy, Berlin Institute of Health at Charité-Universitätsmedizin, Berlin, Germany
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
- Berlin Institute of Health, Center for Regenerative Therapies (BCRT), Berlin, Germany
| |
Collapse
|
8
|
Xiao QX, Geng MJ, Wang QL, Fang CL, Zhang JH, Liu Q, Xiong LL. Unraveling the effects of prenatal anesthesia on neurodevelopment: A review of current evidence and future directions. Neurotoxicology 2024; 105:96-110. [PMID: 39276873 DOI: 10.1016/j.neuro.2024.09.003] [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: 05/26/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Human brain development is a complex, multi-stage, and sensitive process, especially during the fetal stage. Animal studies over the last two decades have highlighted the potential risks of anesthetics to the developing brain, impacting its structure and function. This has raised concerns regarding the safety of anesthesia during pregnancy and its influence on fetal brain development, garnering significant attention from the anesthesiology community. Although preclinical studies predominantly indicate the neurotoxic effects of prenatal anesthesia, these findings cannot be directly extrapolated to humans due to interspecies variations. Clinical research, constrained by ethical and technical hurdles in accessing human prenatal brain tissues, often yields conflicting results compared to preclinical data. The emergence of brain organoids as a cutting-edge research tool shows promise in modeling human brain development. When integrated with single-cell sequencing, these organoids offer insights into potential neurotoxic mechanisms triggered by prenatal anesthesia. Despite several retrospective and cohort studies exploring the clinical impact of anesthesia on brain development, many findings remain inconclusive. As such, this review synthesizes preclinical and clinical evidence on the effects of prenatal anesthesia on fetal brain development and suggests areas for future research advancement.
Collapse
Affiliation(s)
- Qiu-Xia Xiao
- Department of Anesthesiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, China
| | - Min-Jian Geng
- The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Qiu-Lin Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, China
| | - Chang-Le Fang
- Department of Anesthesiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, China
| | - Jing-Han Zhang
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Qi Liu
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Liu-Lin Xiong
- Department of Anesthesiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, China.
| |
Collapse
|
9
|
Jalali H, Rahimian S, Shahsavarian N, Norouzi R, Ahmadiyeh Z, Najafi H, Golchin H. The organoid modeling approach to understanding the mechanisms underlying neurodegeneration: A comprehensive review. Life Sci 2024; 358:123198. [PMID: 39486620 DOI: 10.1016/j.lfs.2024.123198] [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: 07/31/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
Neurodegenerative diseases (NDs) are severe disorders of the nervous system, and their causes are still not completely understood. Modeling the complex pathological mechanisms underlying NDs has long posed a significant challenge, as traditional in vitro and animal models often fail to accurately recapitulate the disease phenotypes observed in humans; however, the rise of organoid technology has opened new approaches for developing innovative disease models that can better capture the nuances of the human nervous system. Organoid platforms hold promise for contributing to the design of future clinical trials and advancing our understanding of these devastating neurological conditions and accelerate the discovery of effective, personalized therapies. This comprehensive review discusses the recent advancements in neural organoid technology and explores the potential of patient-derived organoids for modeling NDs conditions and presents findings related to the mechanisms of their development or progress.
Collapse
Affiliation(s)
- Hanieh Jalali
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Sana Rahimian
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Nasim Shahsavarian
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Rozhan Norouzi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Zahra Ahmadiyeh
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Hossein Najafi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Hasti Golchin
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
10
|
Cordeiro S, Oliveira BB, Valente R, Ferreira D, Luz A, Baptista PV, Fernandes AR. Breaking the mold: 3D cell cultures reshaping the future of cancer research. Front Cell Dev Biol 2024; 12:1507388. [PMID: 39659521 PMCID: PMC11628512 DOI: 10.3389/fcell.2024.1507388] [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: 10/07/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Despite extensive efforts to unravel tumor behavior and develop anticancer therapies, most treatments fail when advanced to clinical trials. The main challenge in cancer research has been the absence of predictive cancer models, accurately mimicking the tumoral processes and response to treatments. The tumor microenvironment (TME) shows several human-specific physical and chemical properties, which cannot be fully recapitulated by the conventional 2D cell cultures or the in vivo animal models. These limitations have driven the development of novel in vitro cancer models, that get one step closer to the typical features of in vivo systems while showing better species relevance. This review introduces the main considerations required for developing and exploiting tumor spheroids and organoids as cancer models. We also detailed their applications in drug screening and personalized medicine. Further, we show the transition of these models into novel microfluidic platforms, for improved control over physiological parameters and high-throughput screening. 3D culture models have provided key insights into tumor biology, more closely resembling the in vivo TME and tumor characteristics, while enabling the development of more reliable and precise anticancer therapies.
Collapse
Affiliation(s)
- Sandra Cordeiro
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Beatriz B. Oliveira
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Ruben Valente
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Daniela Ferreira
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - André Luz
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Pedro V. Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Alexandra R. Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
11
|
Wang J, Jiang N, Liu F, Wang C, Zhou W. Uncovering the intricacies of O-GlcNAc modification in cognitive impairment: New insights from regulation to therapeutic targeting. Pharmacol Ther 2024; 266:108761. [PMID: 39603350 DOI: 10.1016/j.pharmthera.2024.108761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
O-linked β-N-acetylglucosamine (O-GlcNAc) represents a post-translational modification that occurs on serine or threonine residues on various proteins. This conserved modification interacts with vital cellular pathways. Although O-GlcNAc is widely distributed throughout the body, it is particularly enriched in the brain, where most proteins are O-GlcNAcylated. Recent studies have established a causal link between O-GlcNAc regulation in the brain and alterations in neurophysiological function. Alterations in O-GlcNAc levels in the brain are associated with the pathogenesis of several neurogenic diseases that can lead to cognitive impairment. Remarkably, manipulation of O-GlcNAc levels demonstrated a protective effect on cognitive function. Although the precise molecular mechanism of O-GlcNAc modification in the nervous system remains elusive, its regulation is fundamental to multiple neural and cognitive functions, fluctuating levels during normal and pathological cognitive processes. In this review, we highlight the significant functional importance of O-GlcNAc modification in pathological cognitive impairments and the potential application of O-GlcNAc as a promising target for the intervention or amelioration of cognitive impairments.
Collapse
Affiliation(s)
- Jianhui Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Medicines, Beijing 100850, China
| | - Ning Jiang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Medicines, Beijing 100850, China
| | - Feng Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Medicines, Beijing 100850, China
| | - Chenran Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Medicines, Beijing 100850, China
| | - Wenxia Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Medicines, Beijing 100850, China.
| |
Collapse
|
12
|
Zhang T, Yang S, Ge Y, Yin L, Pu Y, Gu Z, Chen Z, Liang G. Unveiling the Heart's Hidden Enemy: Dynamic Insights into Polystyrene Nanoplastic-Induced Cardiotoxicity Based on Cardiac Organoid-on-a-Chip. ACS NANO 2024; 18:31569-31585. [PMID: 39482939 DOI: 10.1021/acsnano.4c13262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Exposure to micro- and nanoplastics (MNPs) has been implicated in potential cardiotoxicity. However, in vitro models based on cardiomyocyte cell lines lack crucial cardiac characteristics, while interspecies differences in animal models compromise the reliability of the conclusions. In addition, current research has predominantly focused on single-time point exposures to MNPs, neglecting comparative analyses of cardiac injury across early and late stages. Moreover, there remains a large gap in understanding the susceptibility to MNPs under pathological conditions. To address these limitations, this study integrated cardiac organoids (COs) and organ-on-a-chip (OoC) technology to develop the cardiac organoid-on-a-chip (COoC), which was validated for cardiotoxicity evaluation through multiple dimensions. Based on COoC, we conducted a dynamic observation of the cardiac damage caused by short- and long-term exposure to polystyrene nanoplastics (PS-NPs). Oxidative stress, inflammation, disruption of calcium ion homeostasis, and mitochondrial dysfunction were confirmed as the potential mechanisms of PS-NP-induced cardiotoxicity and the crucial events in the early stages, while cardiac fibrosis emerged as a prominent feature in late stages. Notably, low-dose exposure exacerbated myocardial infarction symptoms under pathological states, despite no significant cardiotoxicity shown in healthy models. In conclusion, these findings further deepened our understanding of PS-NP-induced cardiotoxic effects and introduced a promising in vitro platform for assessing cardiotoxicity.
Collapse
Affiliation(s)
- Tianyi Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
- Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou 215163, China
| | - Sheng Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
- Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou 215163, China
| | - Yiling Ge
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
- Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou 215163, China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
13
|
Bhattacharya R, Bose D, Kaur T, Patel R, Renuka O, Rodriguez RV. Model Organoids: Integrated Frameworks for the Next Frontier of Healthcare Advancements. Stem Cell Rev Rep 2024:10.1007/s12015-024-10814-3. [PMID: 39527389 DOI: 10.1007/s12015-024-10814-3] [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: 10/24/2024] [Indexed: 11/16/2024]
Abstract
The morphogenetic events leading to tissue formation can be recapitulated using organoids, which allows studying new diseases and modelling personalized medicines. In this review, culture systems comparable to human organs are presented, these organoids are created from pluripotent stem cells or adult stem cells. The efficient and reproducible models of human tissues are discussed for biobanking, precision medicine and basic research. Mechanisms used by these model systems with an overview of models from human cells are also covered. As human physiology is different from animals, culture conditions and tissue limits often become challenging. Organoids offer novel approaches for such cases with rapid screening, transplantation studies and in immunotherapy. Discrepancies with large datasets can be handled with an integrated framework of artificial intelligence or AI and machine learning. An attempt has been made to show the improved effectiveness, simplified iterations, along with image analysis that are possible from this synergy. AI-assisted organoids have the potential to transform healthcare by improving disease understanding and accelerating clinical decision-making through personalized and precision medicine.
Collapse
Affiliation(s)
- Riya Bhattacharya
- AI-Research Centre, School of Business, Woxsen University, Hyderabad, Telangana, India
- Centre of Excellence for Health Technology, Ecosystems, & Biodiversity, Woxsen University, Hyderabad, Telangana, India
| | - Debajyoti Bose
- AI-Research Centre, School of Business, Woxsen University, Hyderabad, Telangana, India.
- Centre of Excellence for Health Technology, Ecosystems, & Biodiversity, Woxsen University, Hyderabad, Telangana, India.
| | - Tanveen Kaur
- Roy J. Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University of Science and Technology, Ames, IA, USA
| | - Rushik Patel
- AI-Research Centre, School of Business, Woxsen University, Hyderabad, Telangana, India
- School of Technology, Woxsen University, Hyderabad, Telangana, India
| | - Oladri Renuka
- AI-Research Centre, School of Business, Woxsen University, Hyderabad, Telangana, India
- School of Technology, Woxsen University, Hyderabad, Telangana, India
| | - Raul V Rodriguez
- AI-Research Centre, School of Business, Woxsen University, Hyderabad, Telangana, India.
- Centre of Excellence for Health Technology, Ecosystems, & Biodiversity, Woxsen University, Hyderabad, Telangana, India.
- School of Business, Woxsen University, Hyderabad, Telangana, India.
| |
Collapse
|
14
|
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: 0] [Impact Index Per Article: 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
|
15
|
Li K, Gu L, Cai H, Lu HC, Mackie K, Guo F. Human brain organoids for understanding substance use disorders. Drug Metab Pharmacokinet 2024:101036. [PMID: 39567282 DOI: 10.1016/j.dmpk.2024.101036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Substance use disorders (SUDs) are complex mental health conditions involving a problematic pattern of substance use. Challenges remain in understanding its neural mechanisms, which are likely to lead to improved SUD treatments. Human brain organoids, brain-like 3D in vitro cultures derived from human stem cells, show unique potential in recapitulating the response of a developing human brain to substances. Here, we review the recent progress in understanding SUD using human brain organoid models focusing on neurodevelopmental perspectives. We first summarize the background of SUD in humans. Moreover, we introduce the development of various human brain organoid models and then discuss current progress and findings underlying the abuse of substances like nicotine, alcohol, and other addictive drugs using organoid models. Furthermore, we review efforts to develop organ chips and microphysiological systems to engineer better human brain organoids for advancing SUD studies. Lastly, we conclude by elaborating on the current challenges and future directions of SUD studies using human brain organoids.
Collapse
Affiliation(s)
- Kangle Li
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Longjun Gu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Hui-Chen Lu
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, USA
| | - Ken Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, USA
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA.
| |
Collapse
|
16
|
Hsu CC, Wang G, Li CF, Zhang X, Cai Z, Chen T, Pan BS, Manne RK, Deep G, Gu H, Wang Y, Peng D, Penugurti V, Zhou X, Xu Z, Chen Z, Chen M, Armstrong AJ, Huang J, Li HY, Lin HK. IMPA1-derived inositol maintains stemness in castration-resistant prostate cancer via IMPDH2 activation. J Exp Med 2024; 221:e20231832. [PMID: 39470689 PMCID: PMC11528126 DOI: 10.1084/jem.20231832] [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: 10/08/2023] [Revised: 07/09/2024] [Accepted: 08/19/2024] [Indexed: 10/30/2024] Open
Abstract
Acquisition of prostate cancer stem cells (PCSCs) manifested during androgen ablation therapy (ABT) contributes to castration-resistant prostate cancer (CRPC). However, little is known about the specific metabolites critically orchestrating this process. Here, we show that IMPA1-derived inositol enriched in PCSCs is a key metabolite crucially maintaining PCSCs for CRPC progression and ABT resistance. Notably, conditional Impa1 knockout in the prostate abrogates the pool and properties of PCSCs to orchestrate CRPC progression and prolong the survival of TRAMP mice. IMPA1-derived inositol serves as a cofactor that directly binds to and activates IMPDH2, which synthesizes guanylate nucleotides for maintaining PCSCs with ARlow/- features leading to CRPC progression and ABT resistance. IMPA1/inositol/IMPDH2 axis is upregulated in human prostate cancer, and its overexpression predicts poor survival outcomes. Genetically and pharmacologically targeting the IMPA1/inositol/IMPDH2 axis abrogates CRPC and overcomes ABT resistance in various CRPC xenografts, patient-derived xenograft (PDX) tumor models, and TRAMP mouse models. Our study identifies IMPDH2 as an inositol sensor whose activation by inositol represents a key mechanism for maintaining PCSCs for CRPC and ABT resistance.
Collapse
Affiliation(s)
- Che-Chia Hsu
- Department of Pathology, Duke University Medical Center, Duke University School of Medicine, Durham, NC, USA
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Guihua Wang
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Chien-Feng Li
- Department of Pathology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Xian Zhang
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Zhen Cai
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Tingjin Chen
- Department of Pathology, Duke University Medical Center, Duke University School of Medicine, Durham, NC, USA
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Bo-Syong Pan
- Department of Pathology, Duke University Medical Center, Duke University School of Medicine, Durham, NC, USA
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Rajesh Kumar Manne
- Department of Pathology, Duke University Medical Center, Duke University School of Medicine, Durham, NC, USA
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Haiwei Gu
- Cellular Biology and Pharmacology Department, Center for Translational Science, The Herbert Wertheim College of Medicine, Florida International University, Port St. Lucie, FL, USA
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer Research Institute, Vancouver, Canada
| | - Danni Peng
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Vasudevarao Penugurti
- Department of Pathology, Duke University Medical Center, Duke University School of Medicine, Durham, NC, USA
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Xiaobo Zhou
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhigang Xu
- Chongqing Engineering Laboratory of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Zhongzhu Chen
- Chongqing Engineering Laboratory of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Ming Chen
- Department of Pathology, Duke University Medical Center, Duke University School of Medicine, Durham, NC, USA
| | - Andrew J. Armstrong
- Duke Cancer Institute Center, Duke University School of Medicine, Durham, NC, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University Medical Center, Duke University School of Medicine, Durham, NC, USA
| | - Hong-Yu Li
- Division of Pharmaceutical Science, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Hui-Kuan Lin
- Department of Pathology, Duke University Medical Center, Duke University School of Medicine, Durham, NC, USA
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| |
Collapse
|
17
|
Li M, Yuan Y, Hou Z, Hao S, Jin L, Wang B. Human brain organoid: trends, evolution, and remaining challenges. Neural Regen Res 2024; 19:2387-2399. [PMID: 38526275 PMCID: PMC11090441 DOI: 10.4103/1673-5374.390972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/26/2023] [Accepted: 10/28/2023] [Indexed: 03/26/2024] Open
Abstract
Advanced brain organoids provide promising platforms for deciphering the cellular and molecular processes of human neural development and diseases. Although various studies and reviews have described developments and advancements in brain organoids, few studies have comprehensively summarized and analyzed the global trends in this area of neuroscience. To identify and further facilitate the development of cerebral organoids, we utilized bibliometrics and visualization methods to analyze the global trends and evolution of brain organoids in the last 10 years. First, annual publications, countries/regions, organizations, journals, authors, co-citations, and keywords relating to brain organoids were identified. The hotspots in this field were also systematically identified. Subsequently, current applications for brain organoids in neuroscience, including human neural development, neural disorders, infectious diseases, regenerative medicine, drug discovery, and toxicity assessment studies, are comprehensively discussed. Towards that end, several considerations regarding the current challenges in brain organoid research and future strategies to advance neuroscience will be presented to further promote their application in neurological research.
Collapse
Affiliation(s)
- Minghui Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuhan Yuan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Zongkun Hou
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Liang Jin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
18
|
Gonzalez-Horta EE, Burgueno JF, Leiva MJ, Villavicencio C, Kawaguchi FI, Hazime H, Reyes F, Manrique-Suárez V, Parra NC, Abreu MT, Toledo JR. Oxidized Low-Density Lipoprotein Induces Reactive Oxygen Species-Dependent Proliferation of Intestinal Epithelial Cells. Pharmaceuticals (Basel) 2024; 17:1466. [PMID: 39598378 PMCID: PMC11597178 DOI: 10.3390/ph17111466] [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: 09/25/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Oxidized low-density lipoprotein (ox-LDL) is a proinflammatory particle associated with various diseases and affects cell proliferation and viability in multiple cell types. However, its impact on intestinal epithelial cells remains underexplored. This study investigates the effect of ox-LDL on colonic epithelial cell proliferation and viability, as well as the underlying mechanisms involved. Methods: The expression levels of ox-LDL receptors in human colonoids were analyzed at baseline and in response to proinflammatory signals by qRT-PCR. The effect of ox-LDL on organoid proliferation was analyzed using morphometric measurements, viability assays, and the incorporation of a thymidine analog into DNA. The generation of reactive oxygen species (ROS) was determined by Amplex Red assays. Additionally, ox-LDL-induced ROS-dependent organoid proliferation was studied by exposing colonoids to an antioxidant or ROS inhibitors. Results: Colonic epithelial cells express ox-LDL receptors. Ox-LDL significantly induces the proliferation of colonic epithelial cells, which are dependent on ROS generation. Notably, ROS scavengers and NADPH inhibitors reduced ox-LDL-induced proliferation, highlighting the crucial role of oxidative stress in this process. Conclusions: This study demonstrates for the first time that ox-LDL stimulates CEC proliferation mediated by ROS production and validates that the colonic organoid model enables the analysis of potential pharmacological strategies for intestinal diseases characterized by oxidative stress and inflammation.
Collapse
Affiliation(s)
- Eddy E. Gonzalez-Horta
- Biotechnology and Biopharmaceutical Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción 4030000, Chile; (E.E.G.-H.); (M.J.L.); (C.V.); (F.R.); (V.M.-S.); (N.C.P.)
- Division of Gastroenterology, Department of Medicine, University of Miami–Miller School of Medicine, Miami, FL 33136, USA; (J.F.B.); (H.H.); (M.T.A.)
| | - Juan F. Burgueno
- Division of Gastroenterology, Department of Medicine, University of Miami–Miller School of Medicine, Miami, FL 33136, USA; (J.F.B.); (H.H.); (M.T.A.)
| | - María J. Leiva
- Biotechnology and Biopharmaceutical Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción 4030000, Chile; (E.E.G.-H.); (M.J.L.); (C.V.); (F.R.); (V.M.-S.); (N.C.P.)
| | - Carla Villavicencio
- Biotechnology and Biopharmaceutical Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción 4030000, Chile; (E.E.G.-H.); (M.J.L.); (C.V.); (F.R.); (V.M.-S.); (N.C.P.)
| | | | - Hajar Hazime
- Division of Gastroenterology, Department of Medicine, University of Miami–Miller School of Medicine, Miami, FL 33136, USA; (J.F.B.); (H.H.); (M.T.A.)
| | - Fátima Reyes
- Biotechnology and Biopharmaceutical Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción 4030000, Chile; (E.E.G.-H.); (M.J.L.); (C.V.); (F.R.); (V.M.-S.); (N.C.P.)
| | - Viana Manrique-Suárez
- Biotechnology and Biopharmaceutical Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción 4030000, Chile; (E.E.G.-H.); (M.J.L.); (C.V.); (F.R.); (V.M.-S.); (N.C.P.)
| | - Natalie C. Parra
- Biotechnology and Biopharmaceutical Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción 4030000, Chile; (E.E.G.-H.); (M.J.L.); (C.V.); (F.R.); (V.M.-S.); (N.C.P.)
| | - Maria T. Abreu
- Division of Gastroenterology, Department of Medicine, University of Miami–Miller School of Medicine, Miami, FL 33136, USA; (J.F.B.); (H.H.); (M.T.A.)
| | - Jorge R. Toledo
- Biotechnology and Biopharmaceutical Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción 4030000, Chile; (E.E.G.-H.); (M.J.L.); (C.V.); (F.R.); (V.M.-S.); (N.C.P.)
| |
Collapse
|
19
|
Chen X, Shen J, Jiang X, Pan M, Chang S, Li J, Wang L, Miao M, Feng X, Zhang L, Shu G, Liu W, Xu F, Zhang W, Ding Z, Zong H, Liu W, Li D, Chen B, Shao M, Fei G, Zha X, Fan X. Characterization of dipyridamole as a novel ferroptosis inhibitor and its therapeutic potential in acute respiratory distress syndrome management. Theranostics 2024; 14:6947-6968. [PMID: 39629132 PMCID: PMC11610143 DOI: 10.7150/thno.102318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/10/2024] [Indexed: 12/06/2024] Open
Abstract
Rationale: Ferroptosis in lung epithelium and endothelium contributes to the pathogenesis of acute respiratory distress syndrome (ARDS), a critical and often fatal condition marked by acute inflammation and elevated pulmonary vascular permeability. Despite this, there are currently no FDA-approved therapeutics specifically targeting ferroptosis for ARDS management. Methods: A screening of 259 FDA-approved drugs was conducted to identify an effective ferroptosis inhibitor in pulmonary epithelial and endothelial cells. The anti-ferroptotic and therapeutic efficacy of this screened drug was rigorously evaluated using two distinct ARDS mouse models (LPS-induced acute lung injury and CLP-induced sepsis) and human airway organoids (hAOs). The regulatory mechanism of this drug on ferroptosis inhibition was investigated via RNA-sequencing, qRT-PCR, western blotting, IF, luciferase reporter assay, chromatin immunoprecipitation assay, limited proteolysis-mass spectrometry assay, cellular thermal shift assay, and drug affinity responsive target stability assay. Furthermore, a proof-of-concept clinical trial was conducted, wherein ARDS patients were administered with the drug as adjunctive therapy. Results: Dipyridamole (DIPY) was identified as a potent inhibitor of ferroptosis in pulmonary epithelial and endothelial cells. DIPY effectively mitigated ferroptosis and pulmonary damage in both mouse models and hAOs, primarily by downregulating heme oxygenase 1 (HMOX1). The transcription factor cAMP responsive element binding protein 1 (CREB1) was identified as a key transactivator of HMOX1, which DIPY effectively downregulated. Mechanistically, DIPY binds to and activates superoxide dismutase 1 (SOD1), which in turn inhibits the CREB1/HMOX1 pathway, thereby suppressing ferroptosis. Notably, the clinical trial further corroborated the therapeutic potential of DIPY in ARDS patients, demonstrating improved outcomes with DIPY adjunctive therapy. Conclusions: These findings provide compelling evidence that DIPY inhibits ferroptosis in pulmonary epithelial and endothelial cells by modulating the SOD1/CREB1/HMOX1 signaling axis and suggest DIPY as a promising therapeutic strategy for ARDS treatment.
Collapse
Affiliation(s)
- Xu Chen
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Jiapan Shen
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Xueqin Jiang
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, China
| | - Min Pan
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Shuang Chang
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Juanjuan Li
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Lei Wang
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Manli Miao
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Xiaoxia Feng
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Ling Zhang
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Guoqing Shu
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Wenjian Liu
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fangzhou Xu
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Wentao Zhang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Zhao Ding
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Huaiyuan Zong
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Weiwei Liu
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Dapeng Li
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Biao Chen
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China
| | - Min Shao
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Guanghe Fei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, China
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Xiaoyun Fan
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, China
| |
Collapse
|
20
|
Airola C, Pallozzi M, Cesari E, Cerrito L, Stella L, Sette C, Giuliante F, Gasbarrini A, Ponziani FR. Hepatocellular-Carcinoma-Derived Organoids: Innovation in Cancer Research. Cells 2024; 13:1726. [PMID: 39451244 PMCID: PMC11505656 DOI: 10.3390/cells13201726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Hepatocellular carcinomas (HCCs) are highly heterogeneous malignancies. They are characterized by a peculiar tumor microenvironment and dense vascularization. The importance of signaling between immune cells, endothelial cells, and tumor cells leads to the difficult recapitulation of a reliable in vitro HCC model using the conventional two-dimensional cell cultures. The advent of three-dimensional organoid tumor technology has revolutionized our understanding of the pathogenesis and progression of several malignancies by faithfully replicating the original cancer genomic, epigenomic, and microenvironmental landscape. Organoids more closely mimic the in vivo environment and cell interactions, replicating factors such as the spatial organization of cell surface receptors and gene expression, and will probably become an important tool in the choice of therapies and the evaluation of tumor response to treatments. This review aimed to describe the ongoing and potential applications of organoids as an in vitro model for the study of HCC development, its interaction with the host's immunity, the analysis of drug sensitivity tests, and the current limits in this field.
Collapse
Affiliation(s)
- Carlo Airola
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
| | - Maria Pallozzi
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
| | - Eleonora Cesari
- GSTeP Organoids Research Core Facility, Fondazione Policlinico A. Gemelli, 00168 Rome, Italy; (E.C.); (C.S.)
| | - Lucia Cerrito
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
| | - Leonardo Stella
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
| | - Claudio Sette
- GSTeP Organoids Research Core Facility, Fondazione Policlinico A. Gemelli, 00168 Rome, Italy; (E.C.); (C.S.)
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Felice Giuliante
- Department of Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
21
|
Man Y, Liu Y, Chen Q, Zhang Z, Li M, Xu L, Tan Y, Liu Z. Organoids-On-a-Chip for Personalized Precision Medicine. Adv Healthc Mater 2024:e2401843. [PMID: 39397335 DOI: 10.1002/adhm.202401843] [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: 06/01/2024] [Revised: 08/25/2024] [Indexed: 10/15/2024]
Abstract
The development of personalized precision medicine has become a pivotal focus in modern healthcare. Organoids-on-a-Chip (OoCs), a groundbreaking fusion of organoid culture and microfluidic chip technology, has emerged as a promising approach to advancing patient-specific treatment strategies. In this review, the diverse applications of OoCs are explored, particularly their pivotal role in personalized precision medicine, and their potential as a cutting-edge technology is highlighted. By utilizing patient-derived organoids, OoCs offer a pathway to optimize treatments, create precise disease models, investigate disease mechanisms, conduct drug screenings, and individualize therapeutic strategies. The emphasis is on the significance of this technological fusion in revolutionizing healthcare and improving patient outcomes. Furthermore, the transformative potential of personalized precision medicine, future prospects, and ongoing advancements in the field, with a focus on genomic medicine, multi-omics integration, and ethical frameworks are discussed. The convergence of these innovations can empower patients, redefine treatment approaches, and shape the future of healthcare.
Collapse
Affiliation(s)
- Yunqi Man
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Zhirou Zhang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| |
Collapse
|
22
|
Fujimori K, Yamanaka S, Shimada K, Matsui K, Kawagoe S, Kuroda T, Ikeda A, Inoue M, Kobayashi E, Yokoo T. Generation of human-pig chimeric renal organoids using iPSC technology. Commun Biol 2024; 7:1278. [PMID: 39375428 PMCID: PMC11458617 DOI: 10.1038/s42003-024-06986-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: 04/22/2024] [Accepted: 09/28/2024] [Indexed: 10/09/2024] Open
Abstract
Porcine organs and human induced pluripotent stem cell (iPSC)-derived organoids as alternative organs for human transplantation have garnered attention, but both face technical challenges. Interspecies chimeric organ production using human iPSCs shows promise in overcoming these challenges. Our group successfully generated chimeric renal organoids using human iPSC-derived nephron progenitor cells (NPCs) and fetal mouse kidneys. However, the current technology is limited to rodents. Therefore, this study focused on producing human-pig chimeric renal organoids, as pigs are the most promising species for xenotransplantation. Modification of existing culture systems enables continuous renal development in both species, resulting in the successful creation of human-pig chimeric renal organoids. Moreover, this method can be applied to generate humanized xenogeneic kidneys for future clinical applications. This study provides evidence that optimizing culture conditions enables the early-stage kidney development beyond species barriers, thus laying the foundation for accelerating research on humanized xenogeneic kidney fabrication for clinical purposes.
Collapse
Affiliation(s)
| | - Shuichiro Yamanaka
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan.
| | | | - Kenji Matsui
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Shiho Kawagoe
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | | | | | | | - Eiji Kobayashi
- Department of Kidney Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
23
|
Rao J, Song C, Hao Y, Chen Z, Feng S, Xu S, Wu X, Xuan Z, Fan Y, Li W, Li J, Ren Y, Li J, Cheng F, Gu Z. Leveraging Patient-Derived Organoids for Personalized Liver Cancer Treatment. Int J Biol Sci 2024; 20:5363-5374. [PMID: 39430248 PMCID: PMC11488587 DOI: 10.7150/ijbs.96317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 09/15/2024] [Indexed: 10/22/2024] Open
Abstract
Primary liver cancer (PLC) is a primary cause of cancer-related death worldwide, and novel treatments are needed due to the limited options available for treatment and tumor heterogeneity. 66 surgically removed PLC samples were cultured using the self-developed 2:2 method, and the final success rate for organoid culture was 40.9%. Organoid performance has been evaluated using comprehensive molecular measurements, such as whole-exome and RNA sequencing, as well as anticancer drug testing. Multiple organoids and their corresponding tumor tissues contained several of the same mutations, with all pairs sharing conventional TP53 mutations. Regarding copy number variations and gene expression, significant correlations were observed between the organoids and their corresponding parental tumor tissues. Comparisons at the molecular level provided us with an assessment of organoid-to-tumor concordance, which, in combination with drug sensitivity testing provided direct guidance for treatment selection. Finally, we were able to determine an appropriate pharmacological regimen for a patient with ICC, demonstrating the clinical practicality in tailoring patient-specific drug regimens. Our study provides an organoid culture technology that can cultivate models that retain most of the molecular characteristics of tumors and can be used for drug sensitivity testing, demonstrating the broad potential application of organoid technology in precision medicine for liver cancer treatment.
Collapse
Affiliation(s)
- Jianhua Rao
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Chao Song
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Yangyang Hao
- Key Laboratory of DGHD, MOE, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Zaozao Chen
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China
- Jiangsu Avatarget Co, Suzhou, China
- Institute of Medical Devices (Suzhou), Southeast University, Nanjing, China
| | - Sidu Feng
- Key Laboratory of DGHD, MOE, School of Life Science and Technology, Southeast University, Nanjing, China
| | | | - Xiaoyue Wu
- Jiangsu Institute for Health and Sport (JIHS), Nanjing, China
| | - Zhengfeng Xuan
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Ye Fan
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Wenzhu Li
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Junda Li
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Yong Ren
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co, Nanjing, China
| | - Jian Li
- Key Laboratory of DGHD, MOE, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Feng Cheng
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University; Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Zhongze Gu
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
- Jiangsu Avatarget Co, Suzhou, China
- Jiangsu Institute for Health and Sport (JIHS), Nanjing, China
- Institute of Medical Devices (Suzhou), Southeast University, Nanjing, China
| |
Collapse
|
24
|
Zhang D, Jin W, Cui Y, He Z. Establishment and Characterization of Testis Organoids with Proliferation and Differentiation of Spermatogonial Stem Cells into Spermatocytes and Spermatids. Cells 2024; 13:1642. [PMID: 39404405 PMCID: PMC11476282 DOI: 10.3390/cells13191642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Organoids play pivotal roles in uncovering the molecular mechanisms underlying organogenesis, intercellular communication, and high-throughput drug screening. Testicular organoids are essential for exploring the genetic and epigenetic regulation of spermatogenesis in vivo and the treatment of male infertility. However, the formation of testicular organoids with full spermatogenesis has not yet been achieved. In this study, neonatal mouse testicular cells were isolated by two-step enzymatic digestion, and they were combined with Matrigel and transplanted subcutaneously into nude mice. Histological examination (H&E) staining and immunohistochemistry revealed that cell grafts assembled to form seminiferous tubules that contained spermatogonial stem cells (SSCs) and Sertoli cells, as illustrated by the co-expression of PLZF (a hallmark for SSCs) and SOX9 (a marker for Sertoli cells) as well as the co-expression of UCHL1 (a hallmark for SSCs) and SOX9, after 8 weeks of transplantation. At 10 weeks of transplantation, SSCs could proliferate and differentiate into spermatocytes as evidenced by the expression of PCNA, Ki67, c-Kit, SYCP3, γ-HA2X, and MLH1. Notably, testicular organoids were seen, and spermatids were observed within the lumen of testicular organoids after 16 weeks of transplantation, as shown by the presence of TNP1 and ACROSIN (hallmarks for spermatids). Collectively, these results implicate that we successfully established testicular organoids with spermatogenesis in vivo. This study thus provides an excellent platform for unveiling the mechanisms underlying mammalian spermatogenesis, and it might offer valuable male gametes for treating male infertility.
Collapse
Affiliation(s)
- Dong Zhang
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China; (D.Z.); (W.J.); (Y.C.)
- Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha 410008, China
- Manufacture-Based Learning & Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Wencong Jin
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China; (D.Z.); (W.J.); (Y.C.)
- Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha 410008, China
- Manufacture-Based Learning & Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Yinghong Cui
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China; (D.Z.); (W.J.); (Y.C.)
- Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha 410008, China
- Manufacture-Based Learning & Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Zuping He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China; (D.Z.); (W.J.); (Y.C.)
- Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha 410008, China
- Manufacture-Based Learning & Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| |
Collapse
|
25
|
Zhang H, Fu L, Leiliang X, Qu C, Wu W, Wen R, Huang N, He Q, Cheng Q, Liu G, Cheng Y. Beyond the Gut: The intratumoral microbiome's influence on tumorigenesis and treatment response. Cancer Commun (Lond) 2024; 44:1130-1167. [PMID: 39087354 PMCID: PMC11483591 DOI: 10.1002/cac2.12597] [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: 01/18/2024] [Revised: 06/25/2024] [Accepted: 07/13/2024] [Indexed: 08/02/2024] Open
Abstract
The intratumoral microbiome (TM) refers to the microorganisms in the tumor tissues, including bacteria, fungi, viruses, and so on, and is distinct from the gut microbiome and circulating microbiota. TM is strongly associated with tumorigenesis, progression, metastasis, and response to therapy. This paper highlights the current status of TM. Tract sources, adjacent normal tissue, circulatory system, and concomitant tumor co-metastasis are the main origin of TM. The advanced techniques in TM analysis are comprehensively summarized. Besides, TM is involved in tumor progression through several mechanisms, including DNA damage, activation of oncogenic signaling pathways (phosphoinositide 3-kinase [PI3K], signal transducer and activator of transcription [STAT], WNT/β-catenin, and extracellular regulated protein kinases [ERK]), influence of cytokines and induce inflammatory responses, and interaction with the tumor microenvironment (anti-tumor immunity, pro-tumor immunity, and microbial-derived metabolites). Moreover, promising directions of TM in tumor therapy include immunotherapy, chemotherapy, radiotherapy, the application of probiotics/prebiotics/synbiotics, fecal microbiome transplantation, engineered microbiota, phage therapy, and oncolytic virus therapy. The inherent challenges of clinical application are also summarized. This review provides a comprehensive landscape for analyzing TM, especially the TM-related mechanisms and TM-based treatment in cancer.
Collapse
Affiliation(s)
- Hao Zhang
- Department of NeurosurgeryThe Second Affiliated HospitalChongqing Medical UniversityChongqingP. R. China
| | - Li Fu
- Department of NeurosurgeryThe Second Affiliated HospitalChongqing Medical UniversityChongqingP. R. China
- Department of GastroenterologyThe Second Affiliated HospitalChongqing Medical UniversityChongqingP. R. China
| | - Xinwen Leiliang
- Department of NeurosurgeryThe Second Affiliated HospitalChongqing Medical UniversityChongqingP. R. China
| | - Chunrun Qu
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanP. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanP. R. China
| | - Wantao Wu
- Department of OncologyXiangya HospitalCentral South UniversityChangshaHunanP. R. China
| | - Rong Wen
- Department of NeurosurgeryThe Second Affiliated HospitalChongqing Medical UniversityChongqingP. R. China
| | - Ning Huang
- Department of NeurosurgeryThe Second Affiliated HospitalChongqing Medical UniversityChongqingP. R. China
| | - Qiuguang He
- Department of NeurosurgeryThe Second Affiliated HospitalChongqing Medical UniversityChongqingP. R. China
| | - Quan Cheng
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanP. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanP. R. China
| | - Guodong Liu
- Department of NeurosurgeryThe Second Affiliated HospitalChongqing Medical UniversityChongqingP. R. China
| | - Yuan Cheng
- Department of NeurosurgeryThe Second Affiliated HospitalChongqing Medical UniversityChongqingP. R. China
| |
Collapse
|
26
|
Shang L, Wang S, Mao Y. Recent advances in plant-derived polysaccharide scaffolds in tissue engineering: A review. Int J Biol Macromol 2024; 277:133830. [PMID: 39002914 DOI: 10.1016/j.ijbiomac.2024.133830] [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: 03/03/2024] [Revised: 06/13/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
As a natural three-dimensional biopolymer, decellularized plant-derived scaffolds usually comprise various polysaccharides, mostly cellulose, pectin, and hemicellulose. They are characterized by natural biocompatibility and porous structures. The emergence of decellularized purified polysaccharide scaffolds provides an attractive method to overcome the challenges associated with nutrient delivery and biocompatibility, as they serve as optimal non-immune environments for stem cell adhesion and proliferation. To date, limited corresponding literature is available to systemically summarize the development and potential of these scaffolds in tissue engineering. Therefore, the current review summarized the biomimetic properties of plant-derived polysaccharide scaffolds and the latest progress in tissue engineering applications. This review first discusses the advantages of decellularized plant-derived polysaccharide scaffolds by briefly introducing their features and current limitations in clinical applications. Subsequently, the latest progress in emerging applications of regenerative biomaterials is reviewed, followed by a discussion of the studies on the interactions of biomaterials with cells and tissues. Finally, challenges in obtaining reliable scaffolds and possible future directions are discussed.
Collapse
Affiliation(s)
- Lijun Shang
- School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Shan Wang
- School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Yingji Mao
- School of Life Sciences, Bengbu Medical University, Bengbu, China.
| |
Collapse
|
27
|
Yao Q, Cheng S, Pan Q, Yu J, Cao G, Li L, Cao H. Organoids: development and applications in disease models, drug discovery, precision medicine, and regenerative medicine. MedComm (Beijing) 2024; 5:e735. [PMID: 39309690 PMCID: PMC11416091 DOI: 10.1002/mco2.735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Organoids are miniature, highly accurate representations of organs that capture the structure and unique functions of specific organs. Although the field of organoids has experienced exponential growth, driven by advances in artificial intelligence, gene editing, and bioinstrumentation, a comprehensive and accurate overview of organoid applications remains necessary. This review offers a detailed exploration of the historical origins and characteristics of various organoid types, their applications-including disease modeling, drug toxicity and efficacy assessments, precision medicine, and regenerative medicine-as well as the current challenges and future directions of organoid research. Organoids have proven instrumental in elucidating genetic cell fate in hereditary diseases, infectious diseases, metabolic disorders, and malignancies, as well as in the study of processes such as embryonic development, molecular mechanisms, and host-microbe interactions. Furthermore, the integration of organoid technology with artificial intelligence and microfluidics has significantly advanced large-scale, rapid, and cost-effective drug toxicity and efficacy assessments, thereby propelling progress in precision medicine. Finally, with the advent of high-performance materials, three-dimensional printing technology, and gene editing, organoids are also gaining prominence in the field of regenerative medicine. Our insights and predictions aim to provide valuable guidance to current researchers and to support the continued advancement of this rapidly developing field.
Collapse
Affiliation(s)
- Qigu Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Sheng Cheng
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Guoqiang Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lanjuan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic‐Chemical and Aging‐Related InjuriesHangzhouChina
| |
Collapse
|
28
|
Du X, Jia H, Chang Y, Zhao Y, Song J. Progress of organoid platform in cardiovascular research. Bioact Mater 2024; 40:88-103. [PMID: 38962658 PMCID: PMC11220467 DOI: 10.1016/j.bioactmat.2024.05.043] [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: 03/04/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 07/05/2024] Open
Abstract
Cardiovascular disease is a significant cause of death in humans. Various models are necessary for the study of cardiovascular diseases, but once cellular and animal models have some defects, such as insufficient fidelity. As a new technology, organoid has certain advantages and has been used in many applications in the study of cardiovascular diseases. This article aims to summarize the application of organoid platforms in cardiovascular diseases, including organoid construction schemes, modeling, and application of cardiovascular organoids. Advances in cardiovascular organoid research have provided many models for different cardiovascular diseases in a variety of areas, including myocardium, blood vessels, and valves. Physiological and pathological models of different diseases, drug research models, and methods for evaluating and promoting the maturation of different kinds of organ tissues are provided for various cardiovascular diseases, including cardiomyopathy, myocardial infarction, and atherosclerosis. This article provides a comprehensive overview of the latest research progress in cardiovascular organ tissues, including construction protocols for cardiovascular organoid tissues and their evaluation system, different types of disease models, and applications of cardiovascular organoid models in various studies. The problems and possible solutions in organoid development are summarized.
Collapse
Affiliation(s)
- Xingchao Du
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yuan Chang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yiqi Zhao
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| |
Collapse
|
29
|
Yan Y, Yang Z, Chen L. High-quality models for assessing the effects of environmental pollutants on the nervous system: 3D brain organoids. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116876. [PMID: 39146594 DOI: 10.1016/j.ecoenv.2024.116876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
The prevalence of environmental problems and the increasing risk of human exposure to environmental pollutants have become a global concern. The increasing environmental pollution is one of the main reasons for the rising incidence of most neurological-related diseases in recent years. However, the ethical constraints of direct human research and the racial limitations of animal models have slowed the progress of research in this area. The purpose of this study is to review the neurotoxicity of different environmental pollutants on the brain using brain organoids as a new model and to conclude that brain organoids may play a key role in assessing the mechanisms by which environmental pollutants affect neurogenesis and cause neurological pathogenesis. To accurately determine the negative effects of environmental pollutants on the nervous system, self-organizing brain organoids that are highly similar to the developing brain have become a new model system for studying the effects of environmental pollutants on human brain development and disease. This study uses brain organoids as a model to summarize the neurotoxicity of different environmental pollutants on the nervous system, including structural changes in brain organoids, inhibition of neuronal differentiation and migration, impairment of mitochondrial function, damage to cellular cilia, and influence on signaling pathways. In conclusion, exposure to environmental pollutants may cause different neurotoxicity to the nervous system. Therefore, it is crucial to understand how to use brain organoids to ameliorate neurological disorders caused by environmental pollution.
Collapse
Affiliation(s)
- Yu Yan
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Ziye Yang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Liqun Chen
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
30
|
Gao Y, Yang Z, Bajpai AK, Wang W, Zhang L, Xia Z. Resveratrol enhances the antiliver cancer effect of cisplatin by targeting the cell membrane protein PLA2. Front Oncol 2024; 14:1453164. [PMID: 39381045 PMCID: PMC11458693 DOI: 10.3389/fonc.2024.1453164] [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: 06/22/2024] [Accepted: 08/30/2024] [Indexed: 10/10/2024] Open
Abstract
Background In this study, we aimed to explore the mechanism by which resveratrol promotes cisplatin-induced death of HepG2 cells and to provide a potential strategy for resveratrol in the treatment of cancer. Methods HepG2 cells were exposed to a range of drug concentrations for 24 h: resveratrol (2.5 μg/mL [10.95 μM], 5 μg/mL [21.91 μM], 10 μg/mL [43.81 μM], 20 μg/mL [87.62 μM], 40 μg/mL [175.25 μM], and 80 μg/mL [350.50 μM]), cisplatin (0.625 μg/mL [2.08 μM], 1.25 μg/mL [4.17 μM], 2.5 μg/mL [8.33 μM], 4.5 μg/mL [15.00 μM], and 10 μg/mL [33.33 μM]), 24 μg/mL (105.15 μM) resveratrol + 9 μg/mL (30.00 μM) cisplatin, and 12 μg/mL (52.57 μM) resveratrol + 4.5 μg/mL (15.00 μM) cisplatin. The interaction of two drugs was evaluated by coefficient of drug interaction (CDI), which was based on the Pharmacological Additivity model. The MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was used to detect the effect of different concentrations of drugs on cell viability, while transcriptome sequencing was used to identify pathways associated with higher gene enrichment. Synchrotron radiation FTIR microspectroscopy experiments and data analysis were conducted to obtain detailed spectral information. The second-derivative spectra were calculated using the Savitzky-Golay algorithm. Single-cell infrared spectral absorption matrices were constructed to analyze the spectral characteristics of individual cells. The Euclidean distance between cells was calculated to assess their spectral similarity. The cell-to-cell Euclidean distance was computed to evaluate the spatial relationships between cells. The target protein of resveratrol was verified by performing a Western blot analysis. Results After 24 h of treatment with resveratrol, HepG2 cell growth was inhibited in a dose-dependent manner. Resveratrol promotes cisplatin-induced HepG2 cell death through membrane-related pathways. It also significantly changes the membrane components of HepG2 cells. Additionally, resveratrol changes the morphology of the HepG2 cell membrane by decreasing the expression of PLA2G2. Conclusion Resveratrol changes the morphology of the HepG2 cell membrane by decreasing the expression of PLA2G2 and promotes cisplatin-induced HepG2 cell death. The combination of cisplatin and resveratrol can play a synergistic therapeutic effect on HepG2 cells.
Collapse
Affiliation(s)
- Yu Gao
- Department of Pharmacy, Binzhou Medical University, Yantai, China
| | - Zhanyi Yang
- Department of Pharmacy, Binzhou Medical University, Yantai, China
| | - Akhilesh Kumar Bajpai
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Wenben Wang
- Department of Pharmacy, Binzhou Medical University, Yantai, China
| | - Liyuan Zhang
- Department of Pharmacy, Binzhou Medical University, Yantai, China
| | - Zhenhong Xia
- Department of Pharmacy, Binzhou Medical University, Yantai, China
- Key Laboratory of Ion Beam Bioengineering, Hefei Institute of Physical Sciences, Chinese Academy of Sciences, Hefei, China
| |
Collapse
|
31
|
Zhou LF, Liao HY, Han Y, Zhao Y. The use of organoids in creating immune microenvironments and treating gynecological tumors. J Transl Med 2024; 22:856. [PMID: 39313812 PMCID: PMC11421176 DOI: 10.1186/s12967-024-05649-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Owing to patient-derived tumor tissues and cells, significant advances have been made in personalized cancer treatment and precision medicine, with cancer stem cell-derived three-dimensional tumor organoids serving as crucial in vitro models that accurately replicate the structural, phenotypic, and genetic characteristics of tumors. However, despite their extensive use in drug testing, genome editing, and transplantation for facilitating personalized treatment approaches in clinical practice, the inadequate capacity of these organoids to effectively model immune cells and stromal components within the tumor microenvironment limits their potential. Additionally, effective clinical immunotherapy has led the tumor immune microenvironment to garner considerable attention, increasing the demand for simulating patient-specific tumor-immune interactions. Consequently, co-culture techniques integrating tumor organoids with immune cells and tumor microenvironment constituents have been developed to expand the possibilities for personalized drug response investigations, with recent advancements enhancing the understanding of the strengths, limitations, and applicability of the co-culture approach. Herein, the recent advancements in the field of tumor organoids have been comprehensively reviewed, specifically highlighting the tumor organoid co-culture-related developments with various immune cell models and their implications for clinical research. Furthermore, this review delineates the current state of research and application of organoid models regarding the therapeutic approaches and related challenges for gynecological tumors. This study may provide a theoretical basis for further research on the use of patient-derived organoids in tumor immunity, drug development, and precision medicine.
Collapse
Affiliation(s)
- Ling-Feng Zhou
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, No.63 Duobao Road, Liwan District, Guangzhou City, Guangdong Province, P.R. China
| | - Hui-Yan Liao
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, No.63 Duobao Road, Liwan District, Guangzhou City, Guangdong Province, P.R. China
| | - Yang Han
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, No.63 Duobao Road, Liwan District, Guangzhou City, Guangdong Province, P.R. China
| | - Yang Zhao
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, No.63 Duobao Road, Liwan District, Guangzhou City, Guangdong Province, P.R. China.
| |
Collapse
|
32
|
Wu HF, Hamilton C, Porritt H, Winbo A, Zeltner N. Modelling neurocardiac physiology and diseases using human pluripotent stem cells: current progress and future prospects. J Physiol 2024. [PMID: 39235952 DOI: 10.1113/jp286416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 08/07/2024] [Indexed: 09/07/2024] Open
Abstract
Throughout our lifetime the heart executes cycles of contraction and relaxation to meet the body's ever-changing metabolic needs. This vital function is continuously regulated by the autonomic nervous system. Cardiovascular dysfunction and autonomic dysregulation are also closely associated; however, the degrees of cause and effect are not always readily discernible. Thus, to better understand cardiovascular disorders, it is crucial to develop model systems that can be used to study the neurocardiac interaction in healthy and diseased states. Human pluripotent stem cell (hiPSC) technology offers a unique human-based modelling system that allows for studies of disease effects on the cells of the heart and autonomic neurons as well as of their interaction. In this review, we summarize current understanding of the embryonic development of the autonomic, cardiac and neurocardiac systems, their regulation, as well as recent progress of in vitro modelling systems based on hiPSCs. We further discuss the advantages and limitations of hiPSC-based models in neurocardiac research.
Collapse
Affiliation(s)
- Hsueh-Fu Wu
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Charlotte Hamilton
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Harrison Porritt
- Department of Physiology, The University of Auckland, Auckland, New Zealand
- Department of Chemical and Materials Engineering, Faculty of Engineering, The University of Auckland, Auckland, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, New Zealand
| | - Annika Winbo
- Department of Physiology, The University of Auckland, Auckland, New Zealand
- Manaaki Manawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Nadja Zeltner
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
33
|
Gopallawa I, Gupta C, Jawa R, Cyril A, Jawa V, Chirmule N, Gujar V. Applications of Organoids in Advancing Drug Discovery and Development. J Pharm Sci 2024; 113:2659-2667. [PMID: 39002723 DOI: 10.1016/j.xphs.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 07/15/2024]
Abstract
Organoids are small, self-organizing three-dimensional cell cultures that are derived from stem cells or primary organs. These cultures replicate the complexity of an organ, which cannot be achieved by single-cell culture systems. Organoids can be used in testing of new drugs instead of animals. Development and validation of organoids is thus important to reduce the reliance on animals for drug testing. In this review, we have discussed the developmental and regulatory aspects of organoids and highlighted their importance in drug development. We have first summarized different types of culture-based organoid systems such as submerged Matrigel, micro-fluidic 3D cultures, inducible pluripotent stem cells, and air-liquid interface cultures. These systems help us understand the intricate interplay between cells and their surrounding milieu for identifying functions of target receptors, soluble factors, and spatial interactions. Further, we have discussed the advances in humanized severe-combined immunodeficiency mouse models and their applications in the pharmacology of immune-oncology. Since regulatory aspects are important in using organoids for drug development, we have summarized FDA and EMA regulations on organoid research to support pre-clinical studies. Finally, we have included some unique studies highlighting the use of organoids in studying infectious diseases, cancer, and fundamental biology. These studies also exemplify the latest technological advances in organoid development resulting in improved efficiency. Overall, this review comprehensively summarizes the applications of organoids in early drug development during discovery and pre-clinical studies.
Collapse
Affiliation(s)
- Indiwari Gopallawa
- Clinical Pharmacology & Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | | | - Rayan Jawa
- University of Pennsylvania, Philadelphia, PA, USA
| | - Arya Cyril
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Vibha Jawa
- Bristol Myers Squibb, Lawrenceville, NY, USA.
| | | | - Vikramsingh Gujar
- Anatomy and Cell Biology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| |
Collapse
|
34
|
Zhou L, Luo D, Lu W, Han J, Zhao M, Li X, Shen T, Jin Z, Zeng J, Wen Y. Gastrointestinal tract organoids as novel tools in drug discovery. Front Pharmacol 2024; 15:1463114. [PMID: 39281285 PMCID: PMC11394194 DOI: 10.3389/fphar.2024.1463114] [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: 07/11/2024] [Accepted: 08/20/2024] [Indexed: 09/18/2024] Open
Abstract
Organoids, characterized by their high physiological attributes, effectively preserve the genetic characteristics, physiological structure, and function of the simulated organs. Since the inception of small intestine organoids, other organoids for organs including the liver, lungs, stomach, and pancreas have subsequently been developed. However, a comprehensive summary and discussion of research findings on gastrointestinal tract (GIT) organoids as disease models and drug screening platforms is currently lacking. Herein, in this review, we address diseases related to GIT organoid simulation and highlight the notable advancements that have been made in drug screening and pharmacokinetics, as well as in disease research and treatment using GIT organoids. Organoids of GIT diseases, including inflammatory bowel disease, irritable bowel syndrome, necrotizing enterocolitis, and Helicobacter pylori infection, have been successfully constructed. These models have facilitated the study of the mechanisms and effects of various drugs, such as metformin, Schisandrin C, and prednisolone, in these diseases. Furthermore, GIT organoids have been used to investigate viruses that elicit GIT reactions, including Norovirus, SARS-CoV-2, and rotavirus. Previous studies by using GIT organoids have shown that dasabuvir, gemcitabine, and imatinib possess the capability to inhibit viral replication. Notably, GIT organoids can mimic GIT responses to therapeutic drugs at the onset of disease. The GIT toxicities of compounds like gefitinib, doxorubicin, and sunset yellow have also been evaluated. Additionally, these organoids are instrumental for the study of immune regulation, post-radiation intestinal epithelial repair, treatment for cystic fibrosis and diabetes, the development of novel drug delivery systems, and research into the GIT microbiome. The recent use of conditioned media as a culture method for replacing recombinant hepatocyte growth factor has significantly reduced the cost associated with human GIT organoid culture. This advancement paves the way for large-scale culture and compound screening of GIT organoids. Despite the ongoing challenges in GIT organoid development (e.g., their inability to exist in pairs, limited cell types, and singular drug exposure mode), these organoids hold considerable potential for drug screening. The use of GIT organoids in this context holds great promises to enhance the precision of medical treatments for patients living with GIT diseases.
Collapse
Affiliation(s)
- Li Zhou
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Luo
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Lu
- Department of Elderly Care Center, Chengdu Pidu District Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Jun Han
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Maoyuan Zhao
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xueyi Li
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Shen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhao Jin
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pediatrics, Guang'an Hospital of Traditional Chinese Medicine, Guang'an, China
| |
Collapse
|
35
|
Li X, Lu Y, Zhao J, Yu Y, Tian H, Zhu H, Li W, Xia Y, Chen L. Savolitinib conferred sensitivity in a patient with D1228H mutation-induced capmatinib-resistant MET exon 14 skipping mutated lung adenocarcinoma. J Cancer Res Clin Oncol 2024; 150:395. [PMID: 39180576 PMCID: PMC11344724 DOI: 10.1007/s00432-024-05920-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/11/2024] [Indexed: 08/26/2024]
Abstract
Traditionally, the D1228 E/G/H/N mutation has been thought to cause Type I MET-TKI resistance. We reported a 75-year-old female with non-small cell lung cancer harboring MET exon 14 skipping mutation, who developed acquired MET D1228H mutation induced by capmatinib treatment. Interestingly, the patient demonstrated marked response to second-line savolitinib treatment with the duration of response of 19 months and several additional metastatic lesions appeared. Pathological assessment of rebiopsy sample showed adenocarcinoma and targeted next-generation sequencing revealed the loss of MET D1228H mutation and presence of MET p.Y1230N mutation. In response, the treatment regimen was amended to include a daily administration of 60 mg of cabozantinib, which resulted in moderate size reduction of the tumours. The switch of resistance mutations indicated that different type Ib MET inhibitors may exhibit distinct mechanisms of resistance. We call for futher studies on resistance based on patient-derived pre-clinical models including patient-derived tumor-like cell clusters, patient-derived organoids, and patient-derived xenografts.
Collapse
Affiliation(s)
- Xiuzhen Li
- Department of Pathology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Yuefei Lu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Jie Zhao
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Yinghui Yu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Heshen Tian
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Hao Zhu
- Department of Respiratory and Critical Care Medicine, Wuyi First People's Hospital, Jinhua, 321200, Zhejiang, China
| | - Wen Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Yang Xia
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, 310009, Zhejiang, China.
| | - Laijuan Chen
- Department of Endoscopic Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
36
|
Zhao X, Li N, Zhang Z, Hong J, Zhang X, Hao Y, Wang J, Xie Q, Zhang Y, Li H, Liu M, Zhang P, Ren X, Wang X. Beyond hype: unveiling the Real challenges in clinical translation of 3D printed bone scaffolds and the fresh prospects of bioprinted organoids. J Nanobiotechnology 2024; 22:500. [PMID: 39169401 PMCID: PMC11337604 DOI: 10.1186/s12951-024-02759-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
Bone defects pose significant challenges in healthcare, with over 2 million bone repair surgeries performed globally each year. As a burgeoning force in the field of bone tissue engineering, 3D printing offers novel solutions to traditional bone transplantation procedures. However, current 3D-printed bone scaffolds still face three critical challenges in material selection, printing methods, cellular self-organization and co-culture, significantly impeding their clinical application. In this comprehensive review, we delve into the performance criteria that ideal bone scaffolds should possess, with a particular focus on the three core challenges faced by 3D printing technology during clinical translation. We summarize the latest advancements in non-traditional materials and advanced printing techniques, emphasizing the importance of integrating organ-like technologies with bioprinting. This combined approach enables more precise simulation of natural tissue structure and function. Our aim in writing this review is to propose effective strategies to address these challenges and promote the clinical translation of 3D-printed scaffolds for bone defect treatment.
Collapse
Affiliation(s)
- Xiangyu Zhao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Na Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Ziqi Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Jinjia Hong
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Xiaoxuan Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Yujia Hao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Jia Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Qingpeng Xie
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Yuan Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Huifei Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Meixian Liu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Pengfei Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Xiuyun Ren
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China.
| | - Xing Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China.
| |
Collapse
|
37
|
Liu W, Cheng G, Cui H, Tian Z, Li B, Han Y, Wu JX, Sun J, Zhao Y, Chen T, Yu G. Theoretical basis, state and challenges of living cell-based drug delivery systems. Theranostics 2024; 14:5152-5183. [PMID: 39267776 PMCID: PMC11388066 DOI: 10.7150/thno.99257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
The therapeutic efficacy of drugs is determined, to a certain extent, by the efficiency of drug delivery. The low efficiency of drug delivery systems (DDSs) is frequently associated with serious toxic side effects and can even prove fatal in certain cases. With the rapid development of technology, drug delivery has evolved from using traditional frameworks to using nano DDSs (NDDSs), endogenous biomaterials DDSs (EBDDSs), and living cell DDSs (LCDDSs). LCDDSs are receiving widespread attention from researchers at present owing to the unique advantages of living cells in targeted drug delivery, including their excellent biocompatibility properties, low immunogenicity, unique biological properties and functions, and role in the treatment of diseases. However, the theoretical basis and techniques involved in the application of LCDDSs have not been extensively summarized to date. Therefore, this review comprehensively summarizes the properties and applications of living cells, elaborates the various drug loading approaches and controlled drug release, and discusses the results of clinical trials. The review also discusses the current shortcomings and prospects for the future development of LCDDSs, which will serve as highly valuable insights for the development and clinical transformation of LCDDSs in the future.
Collapse
Affiliation(s)
- Wei Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Guowang Cheng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Hao Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Zhen Tian
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Bowen Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Yanhua Han
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jia-Xin Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jie Sun
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Yuyue Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Guangtao Yu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
38
|
Chen Z, Chen J, Lin D, Kang H, Luo Y, Wang X, Wang L, Liu D. Forming Single-Cell-Derived Colon Cancer Organoid Arrays on a Microfluidic Chip for High Throughput Tumor Heterogeneity Analysis. ACS Biomater Sci Eng 2024; 10:5265-5273. [PMID: 39087916 DOI: 10.1021/acsbiomaterials.4c00727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Single-cell-derived tumor organoids (STOs) possess a distinct genetic background, making them valuable tools for demonstrating tumor heterogeneity. In order to fulfill the high throughput demands of STO assays, we have developed a microfluidic chip containing 30 000 microwells, which is dedicated to a single cell culture approach for selective expansion and differential induction of cancer stem cells. The microwells are coated with a hydrophilic copolymer to eliminate cell adhesion, and the cell culture is supported by poly(ethylene glycol) (PEG) to establish a nonadhesive culture environment. By utilizing an input cell density of 7 × 103·mL-1, it is possible to construct a 4000 single cell culture system through stochastic cell occupation. We demonstrate that the addition of 15% PEG10000 in the cell culture medium effectively prevents cell loss while facilitating tumor stem cell expansion. As were demonstrated by HCT116, HT29, and SW480 colon cancer cells, the microfluidic approach achieved a STO formation rate of ∼20%, resulting in over 800 STOs generated from a single culture. Comprehensive analysis through histomorphology, immunohistochemistry, drug response evaluation, assessment of cell invasion, and biomarker detection reveals the heterogeneity among individual STOs. Specifically, the smaller STOs exhibited higher invasion and drug resistance capabilities compared with the larger ones. The developed microfluidic approach effectively facilitates STO formation and offers promising prospects for investigating tumor heterogeneity, as well as conducting personalized therapy-focused drug screening.
Collapse
Affiliation(s)
- Zihe Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Jueming Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Dongguo Lin
- Department of Laboratory Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
- Guangdong Engineering Technology Research Center of Microfluidic Chip Medical Diagnosis, Guangzhou 510180, China
| | - Hui Kang
- Department of Pathology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yanzhang Luo
- Department of Laboratory Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Xiaogang Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Lihui Wang
- Department of Pathology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Dayu Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
- Guangdong Engineering Technology Research Center of Microfluidic Chip Medical Diagnosis, Guangzhou 510180, China
| |
Collapse
|
39
|
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] [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
|
40
|
Lou Y, Ma J, Hu Y, Yao X, Liu Y, Wu M, Jia G, Chen Y, Chai R, Xia M, Li W. Integration of Functional Human Auditory Neural Circuits Based on a 3D Carbon Nanotube System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309617. [PMID: 38889308 PMCID: PMC11348147 DOI: 10.1002/advs.202309617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/27/2024] [Indexed: 06/20/2024]
Abstract
The physiological interactions between the peripheral and central auditory systems are crucial for auditory information transmission and perception, while reliable models for auditory neural circuits are currently lacking. To address this issue, mouse and human neural pathways are generated by utilizing a carbon nanotube nanofiber system. The super-aligned pattern of the scaffold renders the axons of the bipolar and multipolar neurons extending in a parallel direction. In addition, the electrical conductivity of the scaffold maintains the electrophysiological activity of the primary mouse auditory neurons. The mouse and human primary neurons from peripheral and central auditory units in the system are then co-cultured and showed that the two kinds of neurons form synaptic connections. Moreover, neural progenitor cells of the cochlea and auditory cortex are derived from human embryos to generate region-specific organoids and these organoids are assembled in the nanofiber-combined 3D system. Using optogenetic stimulation, calcium imaging, and electrophysiological recording, it is revealed that functional synaptic connections are formed between peripheral neurons and central neurons, as evidenced by calcium spiking and postsynaptic currents. The auditory circuit model will enable the study of the auditory neural pathway and advance the search for treatment strategies for disorders of neuronal connectivity in sensorineural hearing loss.
Collapse
Affiliation(s)
- Yiyun Lou
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Jiaoyao Ma
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Yangnan Hu
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjing210096China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Xiaoying Yao
- Obstetrics and Gynecology HospitalFudan UniversityShanghai200011China
| | - Yaoqian Liu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Mingxuan Wu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Gaogan Jia
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Yan Chen
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
- NHC Key Laboratory of Hearing MedicineFudan UniversityShanghai200031China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain ScienceFudan UniversityShanghai200032China
| | - Renjie Chai
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjing210096China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Mingyu Xia
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
- NHC Key Laboratory of Hearing MedicineFudan UniversityShanghai200031China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain ScienceFudan UniversityShanghai200032China
| | - Wenyan Li
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
- NHC Key Laboratory of Hearing MedicineFudan UniversityShanghai200031China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain ScienceFudan UniversityShanghai200032China
| |
Collapse
|
41
|
Wang J, Wu X, Zhao J, Ren H, Zhao Y. Developing Liver Microphysiological Systems for Biomedical Applications. Adv Healthc Mater 2024; 13:e2302217. [PMID: 37983733 DOI: 10.1002/adhm.202302217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/15/2023] [Indexed: 11/22/2023]
Abstract
Microphysiological systems (MPSs), also known as organ chips, are micro-units that integrate cells with diverse physical and biochemical environmental cues. In the field of liver MPSs, cellular components have advanced from simple planar cell cultures to more sophisticated 3D formations such as spheroids and organoids. Additionally, progress in microfluidic devices, bioprinting, engineering of matrix materials, and interdisciplinary technologies have significant promise for producing MPSs with biomimetic structures and functions. This review provides a comprehensive summary of biomimetic liver MPSs including their clinical applications and future developmental potential. First, the key components of liver MPSs, including the principal cell types and engineered structures utilized for cell cultivation, are briefly introduced. Subsequently, the biomedical applications of liver MPSs, including the creation of disease models, drug absorption, distribution, metabolism, excretion, and toxicity, are discussed. Finally, the challenges encountered by MPSs are summarized, and future research directions for their development are proposed.
Collapse
Affiliation(s)
- Jinglin Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xiangyi Wu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Junqi Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Haozhen Ren
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 518071, China
| |
Collapse
|
42
|
Liu H, Gan Z, Qin X, Wang Y, Qin J. Advances in Microfluidic Technologies in Organoid Research. Adv Healthc Mater 2024; 13:e2302686. [PMID: 38134345 DOI: 10.1002/adhm.202302686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/19/2023] [Indexed: 12/24/2023]
Abstract
Organoids have emerged as major technological breakthroughs and novel organ models that have revolutionized biomedical research by recapitulating the key structural and functional complexities of their in vivo counterparts. The combination of organoid systems and microfluidic technologies has opened new frontiers in organoid engineering and offers great opportunities to address the current challenges of existing organoid systems and broaden their biomedical applications. In this review, the key features of the existing organoids, including their origins, development, design principles, and limitations, are described. Then the recent progress in integrating organoids into microfluidic systems is highlighted, involving microarrays for high-throughput organoid manipulation, microreactors for organoid hydrogel scaffold fabrication, and microfluidic chips for functional organoid culture. The opportunities in the nascent combination of organoids and microfluidics that lie ahead to accelerate research in organ development, disease studies, drug screening, and regenerative medicine are also discussed. Finally, the challenges and future perspectives in the development of advanced microfluidic platforms and modified technologies for building organoids with higher fidelity and standardization are envisioned.
Collapse
Affiliation(s)
- Haitao Liu
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Zhongqiao Gan
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinyuan Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaqing Wang
- University of Science and Technology of China, Hefei, 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- University of Science and Technology of China, Hefei, 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
43
|
Lin J, Xia H, Yu J, Wang Y, Wang H, Xie D, Cheng C, Lu L, Bian T, Wu Y, Liu Q. circADAMTS6 via stabilizing CAMK2A is involved in smoking-induced emphysema through driving M2 macrophage polarization. ENVIRONMENT INTERNATIONAL 2024; 190:108832. [PMID: 38936066 DOI: 10.1016/j.envint.2024.108832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/08/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
Cigarette smoke (CS), an indoor environmental pollutant, is a prominent risk factor for emphysema, which is a pathological feature of chronic obstructive pulmonary disease (COPD). Emerging function of circRNAs in immune responses and disease progression shed new light to explore the pathogenesis of emphysema. In this research, we demonstrated, by single-cell RNA sequencing (scRNAseq), that the ratio of M2 macrophages were increased in lung tissues of humans and mice with smoking-related emphysema. Further, our data showed that circADAMTS6 was associated with cigarette smoke extract (CSE)-induced M2 macrophage polarization. Mechanistically, in macrophages, circADAMTS6 stabilized CAMK2A mRNA via forming a circADAMTS6/IGF2BP2/CAMK2A RNA-protein ternary complex to activate CREB, which drives M2 macrophage polarization and leads to emphysema. In addition, in macrophages of mouse lung tissues, downregulation of circADAMTS6 reversed M2 macrophage polarization, the proteinase/anti-proteinase imbalance, and the elastin degradation, which protecting against CS-induced emphysema. Moreover, for macrophages and in a model with co-cultured lung organoids, the target of circADAMTS6 restored the growth of lung organoids compared to CSE-treated macrophages. Our results also demonstrated that, for smokers and COPD smokers, elevation of circADAMTS6 negatively correlated with lung function. Overall, this study reveals a novel mechanism for circADAMTS6-driven M2 macrophage polarization in smoking-related emphysema and postulates that circADAMTS6 could serve as a diagnostic and therapeutic marker for smoking-related emphysema.
Collapse
Affiliation(s)
- Jiaheng Lin
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute for Advanced Study of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Haibo Xia
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute for Advanced Study of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; School of Public Health, Southeast University, Nanjing 210009, Jiangsu, People's Republic of China
| | - Jinyan Yu
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Department of Respiratory and Critical Care Medicine, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, Jiangsu, People's Republic of China
| | - Yue Wang
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute for Advanced Study of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Hailan Wang
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute for Advanced Study of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Daxiao Xie
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute for Advanced Study of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Cheng Cheng
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute for Advanced Study of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Lu Lu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute for Advanced Study of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Tao Bian
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Department of Respiratory and Critical Care Medicine, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, Jiangsu, People's Republic of China.
| | - Yan Wu
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Department of Respiratory and Critical Care Medicine, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, Jiangsu, People's Republic of China.
| | - Qizhan Liu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute for Advanced Study of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.
| |
Collapse
|
44
|
Urciuolo F, Imparato G, Netti PA. Engineering Cell Instructive Microenvironments for In Vitro Replication of Functional Barrier Organs. Adv Healthc Mater 2024; 13:e2400357. [PMID: 38695274 DOI: 10.1002/adhm.202400357] [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: 01/29/2024] [Revised: 04/02/2024] [Indexed: 05/14/2024]
Abstract
Multicellular organisms exhibit synergistic effects among their components, giving rise to emergent properties crucial for their genesis and overall functionality and survival. Morphogenesis involves and relies upon intricate and biunivocal interactions among cells and their environment, that is, the extracellular matrix (ECM). Cells secrete their own ECM, which in turn, regulates their morphogenetic program by controlling time and space presentation of matricellular signals. The ECM, once considered passive, is now recognized as an informative space where both biochemical and biophysical signals are tightly orchestrated. Replicating this sophisticated and highly interconnected informative media in a synthetic scaffold for tissue engineering is unattainable with current technology and this limits the capability to engineer functional human organs in vitro and in vivo. This review explores current limitations to in vitro organ morphogenesis, emphasizing the interplay of gene regulatory networks, mechanical factors, and tissue microenvironment cues. In vitro efforts to replicate biological processes for barrier organs such as the lung and intestine, are examined. The importance of maintaining cells within their native microenvironmental context is highlighted to accurately replicate organ-specific properties. The review underscores the necessity for microphysiological systems that faithfully reproduce cell-native interactions, for advancing the understanding of developmental disorders and disease progression.
Collapse
Affiliation(s)
- Francesco Urciuolo
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Piazzale Tecchio 80, Napoli, 80125, Italy
| | - Giorgia Imparato
- Centre for Advanced Biomaterials for Health Care (IIT@CRIB), Istituto Italiano di Tecnologia, L.go Barsanti e Matteucci, Napoli, 80125, Italy
| | - Paolo Antonio Netti
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Piazzale Tecchio 80, Napoli, 80125, Italy
- Centre for Advanced Biomaterials for Health Care (IIT@CRIB), Istituto Italiano di Tecnologia, L.go Barsanti e Matteucci, Napoli, 80125, Italy
| |
Collapse
|
45
|
Calabrese V, Osakabe N, Siracusa R, Modafferi S, Di Paola R, Cuzzocrea S, Jacob UM, Fritsch T, Abdelhameed AS, Rashan L, Wenzel U, Franceschi C, Calabrese EJ. Transgenerational hormesis in healthy aging and antiaging medicine from bench to clinics: Role of food components. Mech Ageing Dev 2024; 220:111960. [PMID: 38971236 DOI: 10.1016/j.mad.2024.111960] [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: 04/27/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024]
Abstract
Neurodegenerative diseases have multifactorial pathogenesis, mainly involving neuroinflammatory processes. Finding drugs able to treat these diseases, expecially because for most of these diseases there are no effective drugs, and the current drugs cause undesired side effects, represent a crucial point. Most in vivo and in vitro studies have been concentrated on various aspects related to neurons (e.g. neuroprotection), however, there has not been focus on the prevention of early stages involving glial cell activation and neuroinflammation. Recently, it has been demonstrated that nutritional phytochemicals including polyphenols, the main active constituents of the Mediterranean diet, maintain redox balance and neuroprotection through the activation of hormetic vitagene pathway. Recent lipidomics data from our laboratory indicate mushrooms as strong nutritional neuronutrients with strongly activity against neuroinflammation in Meniere' diseaseas, a model of cochleovestibular neural degeneration, as well as in animal model of traumatic brain injury, or rotenone induced parkinson's disease. Moreover, Hidrox®, an aqueous extract of olive containing hydroxytyrosol, and Boswellia, acting as Nrf2 activators, promote resilience by enhancing the redox potential, and thus, regulate through hormetic mechanisms, cellular stress response mechanisms., Thus, modulation of cellular stress pathways, in particular vitagenes system, may be an innovative approach for therapeutic intervention in neurodegenerative disorders.
Collapse
Affiliation(s)
- Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| | - Naomi Osakabe
- Department of Bioscience and Engineering, Shibaura Institute Technology, Tokyo, Japan.
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina 98166, Italy
| | - Sergio Modafferi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, Messina 98168, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina 98166, Italy
| | | | | | - Ali S Abdelhameed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Luay Rashan
- Biodiversity Unit, Dhofar University, Salalah, Oman
| | - Uwe Wenzel
- Institut für Ernährungswissenschaft, Justus Liebig Universitat Giessen, Germany
| | | | - Edward J Calabrese
- Department of Environmental Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
46
|
Quílez C, Jeon EY, Pappalardo A, Pathak P, Abaci HE. Efficient Generation of Skin Organoids from Pluripotent Cells via Defined Extracellular Matrix Cues and Morphogen Gradients in a Spindle-Shaped Microfluidic Device. Adv Healthc Mater 2024; 13:e2400405. [PMID: 38452278 PMCID: PMC11305970 DOI: 10.1002/adhm.202400405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/05/2024] [Indexed: 03/09/2024]
Abstract
Pluripotent stem cell-derived skin organoids (PSOs) emerge as a developmental skin model that is self-organized into multiple components, such as hair follicles. Despite their impressive complexity, PSOs are currently generated in the absence of 3D extracellular matrix (ECM) signals and have several major limitations, including an inverted anatomy (e.g., epidermis inside/dermis outside). In this work, a method is established to generate PSOs effectively in a chemically-defined 3D ECM environment. After examining various dermal ECM molecules, it is found that PSOs generated in collagen -type I (COLI) supplemented with laminin 511 (LAM511) exhibit increased growth compared to conventional free-floating conditions, but fail to induce complete skin differentiation due in part to necrosis. This problem is addressed by generating the PSOs in a 3D bioprinted spindle-shaped hydrogel device, which constrains organoid growth longitudinally. This culture system significantly reduces organoid necrosis and leads to a twofold increase in keratinocyte differentiation and an eightfold increase in hair follicle formation. Finally, the system is adapted as a microfluidic device to create asymmetrical gradients of differentiation factors and improves the spatial organization of dermal and epidermal cells. This study highlights the pivotal role of ECM and morphogen gradients in promoting and spatially-controlling skin differentiation in the PSO framework.
Collapse
Affiliation(s)
- Cristina Quílez
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Bioengineering, Universidad Carlos III de Madrid, Leganés, 28911 Spain
- Fundación Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, 28040, Spain
| | - Eun Y. Jeon
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Alberto Pappalardo
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Pooja Pathak
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hasan E. Abaci
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| |
Collapse
|
47
|
Li Z, Wu W, Li Q, Heng X, Zhang W, Zhu Y, Chen L, Chen Z, Shen M, Ma N, Xiao Q, Yan Y. BCL6B-dependent suppression of ETV2 hampers endothelial cell differentiation. Stem Cell Res Ther 2024; 15:226. [PMID: 39075623 PMCID: PMC11287929 DOI: 10.1186/s13287-024-03832-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/27/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND B-cell CLL/lymphoma 6 member B (BCL6B) operates as a sequence-specific transcriptional repressor within the nucleus, playing crucial roles in various biological functions, including tumor suppression, immune response, stem cell self-renew, and vascular angiogenesis. However, whether BCL6B is involved in endothelial cell (EC) development has remained largely unknown. ETS variant transcription factor 2 (ETV2) is well known to facilitate EC differentiation. This study aims to determine the important role of BCL6B in EC differentiation and its potential mechanisms. METHODS Doxycycline-inducible human induced pluripotent stem cell (hiPSC) lines with BCL6B overexpression or BCL6B knockdown were established and subjected to differentiate into ECs and vessel organoids (VOs). RNA sequencing analysis was performed to identify potential signal pathways regulated by BCL6B during EC differentiation from hiPSCs. Quantitative real-time PCR (qRT-PCR) was used to detect the expression of pluripotency and vascular-specific marker genes expression. EC differentiation efficiency was determined by Flow cytometry analysis. The performance of EC was evaluated by in vitro Tube formation assay. The protein expression and the vessel-like structures were assessed using immunofluorescence analysis or western blot. Luciferase reporter gene assay and chromatin immunoprecipitation (ChIP)-PCR analysis were used to determine the regulatory relationship between BCL6B and ETV2. RESULTS Functional ECs and VOs were successfully generated from hiPSCs. Notably, overexpression of BCL6B suppressed while knockdown of BCL6B improved EC differentiation from hiPSCs. Additionally, the overexpression of BCL6B attenuated the capacity of derived hiPSC-ECs to form a tubular structure. Furthermore, compared to the control VOs, BCL6B overexpression repressed the growth of VOs, whereas BCL6B knockdown had little effect on the size of VOs. RNA sequencing analysis confirmed that our differentiation protocol induced landscape changes for cell/tissue/system developmental process, particularly vascular development and tube morphogenesis, which were significantly modulated by BCL6B. Subsequent experiments confirmed the inhibitory effect of BCL6B is facilitated by the binding of BCL6B to the promoter region of ETV2, led to the suppression of ETV2's transcriptional activity. Importantly, the inhibitory effect of BCL6B overexpression on EC differentiation from hiPSCs could be rescued by ETV2 overexpression. CONCLUSIONS BCL6B inhibits EC differentiation and hinders VO development by repressing the transcriptional activity of ETV2.
Collapse
Affiliation(s)
- Zhonghao Li
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Wei Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qiushi Li
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Xin Heng
- Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wei Zhang
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Yinghong Zhu
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Lin Chen
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Ziqi Chen
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Mengcheng Shen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, 94305, USA
| | - Ning Ma
- Guangzhou National Laboratory, Guangzhou, 510005, China.
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK.
- Key Laboratory of Cardiovascular Diseases at The Second Affiliated Hospital of Guangzhou Medical University and Guangzhou Municipal, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Yi Yan
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
48
|
Li K, Gu L, Cai H, Lu HC, Mackie K, Guo F. Human brain organoids for understanding substance use disorders. Drug Metab Pharmacokinet 2024; 58:101031. [PMID: 39146603 DOI: 10.1016/j.dmpk.2024.101031] [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: 06/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024]
Abstract
Substance use disorders (SUDs) are complex mental health conditions involving a problematic pattern of substance use. Challenges remain in understanding their neural mechanisms, which are likely to lead to improved SUD treatments. Human brain organoids, brain-like 3D in vitro cultures derived from human stem cells, show unique potential in recapitulating the response of a developing human brain to substances. Here, we review the recent progress in understanding SUDs using human brain organoid models focusing on neurodevelopmental perspectives. We first summarize the background of SUDs in humans. Moreover, we introduce the development of various human brain organoid models and then discuss current progress and findings underlying the abuse of substances like nicotine, alcohol, and other addictive drugs using organoid models. Furthermore, we review efforts to develop organ chips and microphysiological systems to engineer better human brain organoids for advancing SUD studies. Lastly, we conclude by elaborating on the current challenges and future directions of SUD studies using human brain organoids.
Collapse
Affiliation(s)
- Kangle Li
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, United States
| | - Longjun Gu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, United States
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, United States
| | - Hui-Chen Lu
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, United States
| | - Ken Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, United States
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, United States.
| |
Collapse
|
49
|
Ahammed B, Kalangi SK. A Decade of Organoid Research: Progress and Challenges in the Field of Organoid Technology. ACS OMEGA 2024; 9:30087-30096. [PMID: 39035960 PMCID: PMC11256333 DOI: 10.1021/acsomega.4c03683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 07/23/2024]
Abstract
Organoid technology, revolutionizing biomedical research, offers a transformative approach to studying human developmental biology, disease pathology, and drug discovery. Originating from the pioneering work of Henry Van Peters Wilson in 1907 and evolving through subsequent breakthroughs, organoids are three-dimensional structures derived from stem cells or tissue explants that mimic the architecture and function of organs in vitro. With the ability to model various organs such as intestine, liver, brain, kidney, and more, organoids provide unprecedented insights into organ development, disease mechanisms, and drug responses. This review highlights the historical context, generation methods, applications, and challenges of organoid technology. Furthermore, it discusses recent advancements, including strategies to address hypoxia-induced cell death and enhance vascularization within organoids, aiming to refine their physiological relevance and unlock their full potential in personalized medicine and organ transplantation.
Collapse
Affiliation(s)
- Basheer Ahammed
- West BC Colony,
Guduru, Kurnool, Andhra Pradesh 518466, India
| | - Suresh K. Kalangi
- Molecular
Microbiology and Immunology Division, CSIR—Central Drug Research
Institute, Lucknow 226031, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
50
|
Na DH, Cui S, Fang X, Lee H, Eum SH, Shin YJ, Lim SW, Yang CW, Chung BH. Advancements in Research on Genetic Kidney Diseases Using Human-Induced Pluripotent Stem Cell-Derived Kidney Organoids. Cells 2024; 13:1190. [PMID: 39056771 PMCID: PMC11274677 DOI: 10.3390/cells13141190] [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: 05/14/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Genetic or hereditary kidney disease stands as a pivotal cause of chronic kidney disease (CKD). The proliferation and widespread utilization of DNA testing in clinical settings have notably eased the diagnosis of genetic kidney diseases, which were once elusive but are now increasingly identified in cases previously deemed CKD of unknown etiology. However, despite these diagnostic strides, research into disease pathogenesis and novel drug development faces significant hurdles, chiefly due to the dearth of appropriate animal models and the challenges posed by limited patient cohorts in clinical studies. Conversely, the advent and utilization of human-induced pluripotent stem cells (hiPSCs) offer a promising avenue for genetic kidney disease research. Particularly, the development of hiPSC-derived kidney organoid systems presents a novel platform for investigating various forms of genetic kidney diseases. Moreover, the integration of the CRISPR/Cas9 technique into this system holds immense potential for efficient research on genetic kidney diseases. This review aims to explore the applications of in vitro kidney organoids generated from hiPSCs in the study of diverse genetic kidney diseases. Additionally, it will delve into the limitations of this research platform and outline future perspectives for advancing research in this crucial area.
Collapse
Affiliation(s)
- Do Hyun Na
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sheng Cui
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Xianying Fang
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Hanbi Lee
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sang Hun Eum
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Incheon St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Incheon 21431, Republic of Korea
| | - Yoo Jin Shin
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Sun Woo Lim
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Chul Woo Yang
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Byung Ha Chung
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|