1
|
Chen L, Han B, Yang S, Guo L, Zhao L, Liu P, Hong X, Zhao Y, Peng Y, Qi S, Hu L, Chen Y. Toxicological effects and mechanisms of renal injury induced by inhalation exposure to airborne nanoplastics. JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137393. [PMID: 39892132 DOI: 10.1016/j.jhazmat.2025.137393] [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: 08/31/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
Micro-nanoplastics (MNPs) are ubiquitously present in various natural habitats, and the kidney plays a critical role in eliminating metabolic waste from the body. Therefore, nephrotoxicity studies of MNPs are necessary. Consequently, we conducted a study utilizing a mouse model that underwent autonomous inhalation of polystyrene nanoplastics (PS-NPs) to investigate the impact of airborne nanoplastics (NPs) on kidney. The results demonstrated that airborne NPs could accumulate within the kidney subsequent to pulmonary entry. Transcriptome analysis showed that exposure to airborne NPs persistently interfered with important signaling pathways including oxidative stress, inflammation, and coagulation, which activated the NR4A1/CASP3 and TF/F12 signaling pathways. In vitro studies have shown that NPs were internalized by human kidney proximal tubular epithelial (HK-2) cells, leading to a range of pathological responses, and ultimately affecting cell fate. Furthermore, we pioneered the exposure of NPs to human kidney organoids. Our findings revealed a heightened sensitivity in kidney organoids towards NPs as compared to immortalized cell lines. This suggested that exposure to NPs could potentially inflict a more substantial toxic effect on the development of embryonic kidneys. In conclusion, this study has revealed the deleterious effects of exposure to airborne NPs on the mouse kidney.
Collapse
Affiliation(s)
- Liqun Chen
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Bin Han
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, China; Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Shushuai Yang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, China; Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Liqiong Guo
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Lei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Ping Liu
- Tianjin Bioscience Diagnostic Technology Co.Ltd, Tianjin, China
| | - Xiaoming Hong
- Tianjin Mid-Link Biomedical Technology Group, Tianjin, China
| | - Yan Zhao
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, China; Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Yahang Peng
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, China; Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Shiyong Qi
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, China.
| | - Lidan Hu
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Yue Chen
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, China.
| |
Collapse
|
2
|
Huang W, Jeong S, Kim W, Chen L. Biomedical applications of organoids in genetic diseases. MEDICAL REVIEW (2021) 2025; 5:152-163. [PMID: 40224362 PMCID: PMC11987506 DOI: 10.1515/mr-2024-0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/04/2024] [Indexed: 04/15/2025]
Abstract
Organoid technology has significantly transformed biomedical research by providing exceptional prospects for modeling human tissues and disorders in a laboratory setting. It has significant potential for understanding the intricate relationship between genetic mutations, cellular phenotypes, and disease pathology, especially in the field of genetic diseases. The intersection of organoid technology and genetic research offers great promise for comprehending the pathophysiology of genetic diseases and creating innovative treatment approaches customized for specific patients. This review aimed to present a thorough analysis of the current advancements in organoid technology and its biomedical applications for genetic diseases. We examined techniques for modeling genetic disorders using organoid platforms, analyze the approaches for incorporating genetic disease organoids into clinical practice, and showcase current breakthroughs in preclinical application, individualized healthcare, and transplantation. Through the integration of knowledge from several disciplines, such as genetics, regenerative medicine, and biological engineering, our aim is to enhance our comprehension of the complex connection between genetic variations and organoid models in relation to human health and disease.
Collapse
Affiliation(s)
- Wenhua Huang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Seogsong Jeong
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Seoul, Korea
| | - Won Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Divisions of Gastroenterology and Hepatology, Department of Internal Medicine, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Lei Chen
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/National Center for Liver Cancer, Naval Medical University, Shanghai, China
| |
Collapse
|
3
|
Artegiani B, Hendriks D. Organoids from pluripotent stem cells and human tissues: When two cultures meet each other. Dev Cell 2025; 60:493-511. [PMID: 39999776 DOI: 10.1016/j.devcel.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/13/2024] [Accepted: 01/10/2025] [Indexed: 02/27/2025]
Abstract
Human organoids are a widely used tool in cell biology to study homeostatic processes, disease, and development. The term organoids covers a plethora of model systems from different cellular origins that each have unique features and applications but bring their own challenges. This review discusses the basic principles underlying organoids generated from pluripotent stem cells (PSCs) as well as those derived from tissue stem cells (TSCs). We consider how well PSC- and TSC-organoids mimic the different intended organs in terms of cellular complexity, maturity, functionality, and the ongoing efforts to constitute predictive complex models of in vivo situations. We discuss the advantages and limitations associated with each system to answer different biological questions including in the field of cancer and developmental biology, and with respect to implementing emerging advanced technologies, such as (spatial) -omics analyses, CRISPR screens, and high-content imaging screens. We postulate how the two fields may move forward together, integrating advantages of one to the other.
Collapse
Affiliation(s)
| | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
4
|
Kalla J, Pfneissl J, Mair T, Tran L, Egger G. A systematic review on the culture methods and applications of 3D tumoroids for cancer research and personalized medicine. Cell Oncol (Dordr) 2025; 48:1-26. [PMID: 38806997 PMCID: PMC11850459 DOI: 10.1007/s13402-024-00960-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2024] [Indexed: 05/30/2024] Open
Abstract
Cancer is a highly heterogeneous disease, and thus treatment responses vary greatly between patients. To improve therapy efficacy and outcome for cancer patients, more representative and patient-specific preclinical models are needed. Organoids and tumoroids are 3D cell culture models that typically retain the genetic and epigenetic characteristics, as well as the morphology, of their tissue of origin. Thus, they can be used to understand the underlying mechanisms of cancer initiation, progression, and metastasis in a more physiological setting. Additionally, co-culture methods of tumoroids and cancer-associated cells can help to understand the interplay between a tumor and its tumor microenvironment. In recent years, tumoroids have already helped to refine treatments and to identify new targets for cancer therapy. Advanced culturing systems such as chip-based fluidic devices and bioprinting methods in combination with tumoroids have been used for high-throughput applications for personalized medicine. Even though organoid and tumoroid models are complex in vitro systems, validation of results in vivo is still the common practice. Here, we describe how both animal- and human-derived tumoroids have helped to identify novel vulnerabilities for cancer treatment in recent years, and how they are currently used for precision medicine.
Collapse
Affiliation(s)
- Jessica Kalla
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Janette Pfneissl
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Theresia Mair
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Loan Tran
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Vienna, Austria.
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
5
|
Huang J, Wang X, Ge S, Lu X, Sun C. Organoids as Sophisticated Tools for Renal Cancer Research: Extensive Applications and Promising Prospects. Cell Mol Bioeng 2024; 17:527-548. [PMID: 39926385 PMCID: PMC11799493 DOI: 10.1007/s12195-024-00825-y] [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: 08/10/2024] [Accepted: 09/28/2024] [Indexed: 02/11/2025] Open
Abstract
Background Kidney cancer is a significant global health problem that affects nearly 1 in 25 of cancer patients. Prevalence, morbidity and mortality data associated with kidney cancer continue to increase every year, revealing a heavy economic and social burden. Organoid culture is a new research tool with great potential for many applications, particularly in cancer research. The integration of organoids with other emerging technologies has simultaneously expanded their potential applications. However, there is no thorough assessment of organoids in the field of renal cancer research. Objectives This paper presents a comprehensive review of the current development of renal cancer organoids and discusses the corresponding solutions and future directions of renal cancer organoids. Methods In this study, we have compared the operating procedures of different organoid culture protocols and proposed a summary of constituents in culture media. Extensive discussions of renal cancer organoids, including generation and maintenance approaches, application scenarios, current challenges and prospects, have also been made. The information required for this study is extracted from literature databases such as PubMed, SCOPUS and Web of Science. Results In this article, we systematically review thirteen successful methods for generating organoids to kidney cancer and provide practical guidelines for their construction as a reference. In addition, we also elucidate the clinical application of organoids, address the existing challenges and limitations, and highlight promising prospects. Conclusion Ultimately, we firmly believe that as kidney tumour organoids continue to develop and improve, they will become a crucial tool for treating kidney cancer.
Collapse
Affiliation(s)
- Jingqiang Huang
- Department of Urology Surgery, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing’an District, Shanghai, 200040 China
| | - Xianli Wang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Shengyang Ge
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032 China
| | - Xiao Lu
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing’an District, Shanghai, 200040 China
| | - Chuanyu Sun
- Department of Urology Surgery, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing’an District, Shanghai, 200040 China
| |
Collapse
|
6
|
Wang Z, Zong H, Liu W, Lin W, Sun A, Ding Z, Chen X, Wan X, Liu Y, Hu Z, Zhang H, Li H, Liu Y, Li D, Zhang S, Zha X. Augmented ERO1α upon mTORC1 activation induces ferroptosis resistance and tumor progression via upregulation of SLC7A11. J Exp Clin Cancer Res 2024; 43:112. [PMID: 38610018 PMCID: PMC11015652 DOI: 10.1186/s13046-024-03039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 04/08/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND The dysregulated mechanistic target of rapamycin complex 1 (mTORC1) signaling plays a critical role in ferroptosis resistance and tumorigenesis. However, the precise underlying mechanisms still need to be fully understood. METHODS Endoplasmic reticulum oxidoreductase 1 alpha (ERO1α) expression in mTORC1-activated mouse embryonic fibroblasts, cancer cells, and laryngeal squamous cell carcinoma (LSCC) clinical samples was examined by quantitative real-time PCR (qRT-PCR), western blotting, immunofluorescence (IF), and immunohistochemistry. Extensive in vitro and in vivo experiments were carried out to determine the role of ERO1α and its downstream target, member 11 of the solute carrier family 7 (SLC7A11), in mTORC1-mediated cell proliferation, angiogenesis, ferroptosis resistance, and tumor growth. The regulatory mechanism of ERO1α on SLC7A11 was investigated via RNA-sequencing, a cytokine array, an enzyme-linked immunosorbent assay, qRT-PCR, western blotting, IF, a luciferase reporter assay, and a chromatin immunoprecipitation assay. The combined therapeutic effect of ERO1α inhibition and the ferroptosis inducer imidazole ketone erastin (IKE) on mTORC1-activated cells was evaluated using cell line-derived xenografts, LSCC organoids, and LSCC patient-derived xenograft models. RESULTS ERO1α is a functional downstream target of mTORC1. Elevated ERO1α induced ferroptosis resistance and exerted pro-oncogenic roles in mTORC1-activated cells via upregulation of SLC7A11. Mechanically, ERO1α stimulated the transcription of SLC7A11 by activating the interleukin-6 (IL-6)/signal transducer and activator of transcription 3 (STAT3) pathway. Moreover, ERO1α inhibition combined with treatment using the ferroptosis inducer IKE exhibited synergistic antitumor effects on mTORC1-activated tumors. CONCLUSIONS The ERO1α/IL-6/STAT3/SLC7A11 pathway is crucial for mTORC1-mediated ferroptosis resistance and tumor growth, and combining ERO1α inhibition with ferroptosis inducers is a novel and effective treatment for mTORC1-related tumors.
Collapse
Affiliation(s)
- Zixi Wang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui Province, China
- Children's Hospital of Fudan University, National Children's Medical Center, And Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Huaiyuan Zong
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui Province, China
| | - Weiwei Liu
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Wei Lin
- Department of Stomatology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Anjiang Sun
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui Province, China
| | - Zhao Ding
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xu Chen
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui Province, China
| | - Xiaofeng Wan
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Yanyan Liu
- Department of Thyroid and Breast Surgery, Hefei First People's Hospital, Hefei, 230061, China
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hongbing Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Hongwu Li
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Public Health Clinical Center, Hefei, 230011, China
| | - Yehai Liu
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Dapeng Li
- Department of Otorhinolaryngology, Head & Neck Surgery, The Affiliated Bozhou Hospital of Anhui Medical University, No. 616 Duzhong Road, Bozhou, 236800, Anhui Province, China.
| | - Sumei Zhang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui Province, China.
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui Province, China.
- Department of Otorhinolaryngology, Head & Neck Surgery, The Affiliated Bozhou Hospital of Anhui Medical University, No. 616 Duzhong Road, Bozhou, 236800, Anhui Province, China.
| |
Collapse
|
7
|
Dilmen E, Orhon I, Jansen J, Hoenderop JGJ. Advancements in kidney organoids and tubuloids to study (dys)function. Trends Cell Biol 2024; 34:299-311. [PMID: 37865608 DOI: 10.1016/j.tcb.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/14/2023] [Accepted: 09/27/2023] [Indexed: 10/23/2023]
Abstract
The rising prevalence of kidney diseases urges the need for novel therapies. Kidney organoids and tubuloids are advanced in vitro models and have recently been described as promising tools to study kidney (patho)physiology. Recent developments have shown their application in disease modeling, drug screening, and nephrotoxicity. These applications rely on their ability to mimic (dys)function in vitro including endocrine activity and drug, electrolyte, and water transport. This review provides an overview of these emerging kidney models and focuses on the most recent developments that utilize their functional capabilities. In addition, we cover current limitations and provide future perspectives for this rapidly evolving field, including what these functional properties mean for translational and personalized medicine now and in the future.
Collapse
Affiliation(s)
- E Dilmen
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - I Orhon
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J Jansen
- Department of Internal Medicine, Nephrology, and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Experimental Medicine and Systems Biology, University Hospital RWTH Aachen, Aachen, Germany
| | - J G J Hoenderop
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
8
|
Niu W, Siciliano B, Wen Z. Modeling tuberous sclerosis complex with human induced pluripotent stem cells. World J Pediatr 2024; 20:208-218. [PMID: 35759110 DOI: 10.1007/s12519-022-00576-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/23/2022] [Indexed: 12/20/2022]
Abstract
BACKGROUND Tuberous sclerosis complex (TSC) is an autosomal dominant genetic disorder with a birth incidence of 1:6000 in the United States that is characterized by the growth of non-cancerous tumors in multiple organ systems including the brain, kidneys, lungs, and skin. Importantly, TSC is also associated with significant neurological manifestations including epilepsy, TSC-associated neuropsychiatric disorders, intellectual disabilities, and autism spectrum disorder. Mutations in the TSC1 or TSC2 genes are well-established causes of TSC, which lead to TSC1/TSC2 deficiency in organs and hyper-activation of the mammalian target of rapamycin signaling pathway. Animal models have been widely used to study the effect of TSC1/2 genes on the development and function of the brain. Despite considerable progress in understanding the molecular mechanisms underlying TSC in animal models, a human-specific model is urgently needed to investigate the effects of TSC1/2 mutations that are unique to human neurodevelopment. DATA SOURCES Literature reviews and research articles were published in PubMed-indexed journals. RESULTS Human-induced pluripotent stem cells (iPSCs), which capture risk alleles that are identical to their donors and have the capacity to differentiate into virtually any cell type in the human body, pave the way for the empirical study of previously inaccessible biological systems such as the developing human brain. CONCLUSIONS In this review, we present an overview of the recent progress in modeling TSC with human iPSC models, the existing limitations, and potential directions for future research.
Collapse
Affiliation(s)
- Weibo Niu
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Whitehead Research Building 447, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Benjamin Siciliano
- The Graduate Program in Molecular and Systems Pharmacology, Laney Graduate School, Emory University, Atlanta, GA, 30322, USA
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Whitehead Research Building 447, 615 Michael Street, Atlanta, GA, 30322, USA.
- Department of Cell Biology, Emory University School of Medicine, Whitehead Research Building 447, 615 Michael Street, Atlanta, GA, 30322, USA.
- Department of Neurology, Emory University School of Medicine, Whitehead Research Building 447, 615 Michael Street, Atlanta, GA, 30322, USA.
| |
Collapse
|
9
|
Roberto de Barros N, Wang C, Maity S, Peirsman A, Nasiri R, Herland A, Ermis M, Kawakita S, Gregatti Carvalho B, Hosseinzadeh Kouchehbaghi N, Donizetti Herculano R, Tirpáková Z, Mohammad Hossein Dabiri S, Lucas Tanaka J, Falcone N, Choroomi A, Chen R, Huang S, Zisblatt E, Huang Y, Rashad A, Khorsandi D, Gangrade A, Voskanian L, Zhu Y, Li B, Akbari M, Lee J, Remzi Dokmeci M, Kim HJ, Khademhosseini A. Engineered organoids for biomedical applications. Adv Drug Deliv Rev 2023; 203:115142. [PMID: 37967768 PMCID: PMC10842104 DOI: 10.1016/j.addr.2023.115142] [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: 04/18/2023] [Revised: 10/03/2023] [Accepted: 11/10/2023] [Indexed: 11/17/2023]
Abstract
As miniaturized and simplified stem cell-derived 3D organ-like structures, organoids are rapidly emerging as powerful tools for biomedical applications. With their potential for personalized therapeutic interventions and high-throughput drug screening, organoids have gained significant attention recently. In this review, we discuss the latest developments in engineering organoids and using materials engineering, biochemical modifications, and advanced manufacturing technologies to improve organoid culture and replicate vital anatomical structures and functions of human tissues. We then explore the diverse biomedical applications of organoids, including drug development and disease modeling, and highlight the tools and analytical techniques used to investigate organoids and their microenvironments. We also examine the latest clinical trials and patents related to organoids that show promise for future clinical translation. Finally, we discuss the challenges and future perspectives of using organoids to advance biomedical research and potentially transform personalized medicine.
Collapse
Affiliation(s)
| | - Canran Wang
- Andrew and Peggy Cherng Department of Medical Engineering, Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA 91125, USA
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Arne Peirsman
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Plastic and Reconstructive Surgery, Ghent University Hospital, Ghent, Belgium
| | - Rohollah Nasiri
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, 17165 Solna, Sweden
| | - Anna Herland
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, 17165 Solna, Sweden
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Bruna Gregatti Carvalho
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), 13083-970 Campinas, Brazil
| | - Negar Hosseinzadeh Kouchehbaghi
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Department of Textile Engineering, Amirkabir University of Technology (Tehran Polytechnic), Hafez Avenue, 1591634311 Tehran, Iran
| | - Rondinelli Donizetti Herculano
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, USA; São Paulo State University (UNESP), Bioengineering and Biomaterials Group, School of Pharmaceutical Sciences, Araraquara, SP, Brazil
| | - Zuzana Tirpáková
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 04181 Kosice, Slovakia
| | - Seyed Mohammad Hossein Dabiri
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Jean Lucas Tanaka
- Butantan Institute, Viral Biotechnology Laboratory, São Paulo, SP Brazil; University of São Paulo (USP), São Paulo, SP Brazil
| | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Auveen Choroomi
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - RunRun Chen
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, USA
| | - Shuyi Huang
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, USA
| | - Elisheva Zisblatt
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Yixuan Huang
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Ahmad Rashad
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Ankit Gangrade
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Leon Voskanian
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Bingbing Li
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, USA
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Junmin Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | | | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; College of Pharmacy, Korea University, Sejong 30019, Republic of Korea.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA.
| |
Collapse
|
10
|
Nishinakamura R. Advances and challenges toward developing kidney organoids for clinical applications. Cell Stem Cell 2023; 30:1017-1027. [PMID: 37541208 DOI: 10.1016/j.stem.2023.07.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 08/06/2023]
Abstract
Kidney organoids have enabled modeling of human development and disease. While methods of generating the nephron lineage are well established, new protocols to induce another lineage, the ureteric bud/collecting duct, have been reported in the past 5 years. Many reports have described modeling of various hereditary kidney diseases, with polycystic kidney disease serving as the archetypal disease, by using patient-derived or genome-edited kidney organoids. The generation of more organotypic kidneys is also becoming feasible. In this review, I also discuss the significant challenges for more sophisticated disease modeling and for realizing the ambitious goal of generating transplantable synthetic kidneys.
Collapse
Affiliation(s)
- Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan.
| |
Collapse
|
11
|
Guo T, Wei Q. Cell Reprogramming Techniques: Contributions to Cancer Therapy. Cell Reprogram 2023; 25:142-153. [PMID: 37530737 DOI: 10.1089/cell.2023.0011] [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: 08/03/2023] Open
Abstract
The reprogramming of terminally differentiated cells over the past few years has become important for induced pluripotent stem cells (iPSCs) in the field of regenerative medicine and disease drug modeling. At the same time, iPSCs have also played an important role in human cancer research. iPSCs derived from cancer patients can be used to simulate the early progression of cancer, for drug testing, and to study the molecular mechanism of cancer occurrence. In recent years, with the application of cellular immunotherapy in cancer therapy, patient-derived iPSC-induced immune cells (T, natural killer, and macrophage cells) solve the problem of immune rejection and have higher immunogenicity, which greatly improves the therapeutic efficiency of immune cell therapy. With the continuous progress of cancer differentiation therapy, iPSC technology can reprogram cancer cells to a more primitive pluripotent undifferentiated state, and successfully reverse cancer cells to a benign phenotype by changing the epigenetic inheritance of cancer cells. This article reviews the recent progress of cell reprogramming technology in human cancer research, focuses on the application of reprogramming technology in cancer immunotherapy and the problems solved, and summarizes the malignant phenotype changes of cancer cells in the process of reprogramming and subsequent differentiation.
Collapse
Affiliation(s)
- Tongtong Guo
- College of Life Science, Northwest University, Xi'an, China
| | - Qi Wei
- Wuhan Institute of Technology, Wuhan, China
| |
Collapse
|
12
|
Pietrobon A, Stanford WL. Tuberous Sclerosis Complex Kidney Lesion Pathogenesis: A Developmental Perspective. J Am Soc Nephrol 2023; 34:1135-1149. [PMID: 37060140 PMCID: PMC10356159 DOI: 10.1681/asn.0000000000000146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/27/2023] [Indexed: 04/16/2023] Open
Abstract
The phenotypic diversity of tuberous sclerosis complex (TSC) kidney pathology is enigmatic. Despite a well-established monogenic etiology, an incomplete understanding of lesion pathogenesis persists. In this review, we explore the question: How do TSC kidney lesions arise? We appraise literature findings in the context of mutational timing and cell-of-origin. Through a developmental lens, we integrate the critical results from clinical studies, human specimens, and genetic animal models. We also review novel insights gleaned from emerging organoid and single-cell sequencing technologies. We present a new model of pathogenesis which posits a phenotypic continuum, whereby lesions arise by mutagenesis during development from variably timed second-hit events. This model can serve as a conceptual framework for testing hypotheses of TSC lesion pathogenesis, both in the kidney and in other affected tissues.
Collapse
Affiliation(s)
- Adam Pietrobon
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| | - William L. Stanford
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| |
Collapse
|
13
|
Amleh A, Chen HP, Watad L, Abramovich I, Agranovich B, Gottlieb E, Ben-Dov IZ, Nechama M, Volovelsky O. Arginine depletion attenuates renal cystogenesis in tuberous sclerosis complex model. Cell Rep Med 2023:101073. [PMID: 37290438 DOI: 10.1016/j.xcrm.2023.101073] [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: 07/13/2022] [Revised: 03/02/2023] [Accepted: 05/15/2023] [Indexed: 06/10/2023]
Abstract
Cystic kidney disease is a leading cause of morbidity in patients with tuberous sclerosis complex (TSC). We characterize the misregulated metabolic pathways using cell lines, a TSC mouse model, and human kidney sections. Our study reveals a substantial perturbation in the arginine biosynthesis pathway in TSC models with overexpression of argininosuccinate synthetase 1 (ASS1). The rise in ASS1 expression is dependent on the mechanistic target of rapamycin complex 1 (mTORC1) activity. Arginine depletion prevents mTORC1 hyperactivation and cell cycle progression and averts cystogenic signaling overexpression of c-Myc and P65. Accordingly, an arginine-depleted diet substantially reduces the TSC cystic load in mice, indicating the potential therapeutic effects of arginine deprivation for the treatment of TSC-associated kidney disease.
Collapse
Affiliation(s)
- Athar Amleh
- Pediatric Nephrology Unit, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; Wohl Institute for Translational Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Hadass Pri Chen
- Wohl Institute for Translational Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; Department of Nephrology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lana Watad
- Pediatric Nephrology Unit, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; Wohl Institute for Translational Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ifat Abramovich
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Bella Agranovich
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Eyal Gottlieb
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Iddo Z Ben-Dov
- Department of Nephrology, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; Laboratory of Medical Transcriptomics, Department of Nephrology and Hypertension and Internal Medicine B, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Morris Nechama
- Pediatric Nephrology Unit, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; Wohl Institute for Translational Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| | - Oded Volovelsky
- Pediatric Nephrology Unit, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; Wohl Institute for Translational Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
14
|
Vandana JJ, Manrique C, Lacko LA, Chen S. Human pluripotent-stem-cell-derived organoids for drug discovery and evaluation. Cell Stem Cell 2023; 30:571-591. [PMID: 37146581 PMCID: PMC10775018 DOI: 10.1016/j.stem.2023.04.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/27/2023] [Accepted: 04/11/2023] [Indexed: 05/07/2023]
Abstract
Human pluripotent stem cells (hPSCs) and three-dimensional organoids have ushered in a new era for disease modeling and drug discovery. Over the past decade, significant progress has been in deriving functional organoids from hPSCs, which have been applied to recapitulate disease phenotypes. In addition, these advancements have extended the application of hPSCs and organoids for drug screening and clinical-trial safety evaluations. This review provides an overview of the achievements and challenges in using hPSC-derived organoids to conduct relevant high-throughput, high-contentscreens and drug evaluation. These studies have greatly enhanced our knowledge and toolbox for precision medicine.
Collapse
Affiliation(s)
- J Jeya Vandana
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Tri-Institutional PhD Program in Chemical Biology, Weill Cornell Medicine, The Rockefeller University, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Cassandra Manrique
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medical College, New York, NY, USA
| | - Lauretta A Lacko
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
15
|
Kuo TC, Cabrera-Barragan DN, Lopez-Marfil M, Lopez-Cantu DO, Lemos DR. Can Kidney Organoid Xenografts Accelerate Therapeutic Development for Genetic Kidney Disorders? J Am Soc Nephrol 2023; 34:184-190. [PMID: 36344066 PMCID: PMC10103095 DOI: 10.1681/asn.2022080862] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022] Open
Abstract
A number of genetic kidney diseases can now be replicated experimentally, using kidney organoids generated from human pluripotent stem cells. This methodology holds great potential for drug discovery. Under in vitro conditions, however, kidney organoids remain developmentally immature, develop scarce vasculature, and may contain undesired off-target cell types. Those critical deficiencies limit their potential as disease-modeling tools. Orthotopic transplantation under the kidney capsule improves the anatomic maturity and vascularization of kidney organoids, while reducing off-target cell content. The improvements can translate into more accurate representations of disease phenotypes and mechanisms in vivo . Recent studies using kidney organoid xenografts highlighted the unique potential of this novel methodology for elucidating molecular mechanisms driving monogenic kidney disorders and for the development ofnovel pharmacotherapies.
Collapse
Affiliation(s)
- Ting-Chun Kuo
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Dalia N. Cabrera-Barragan
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Marta Lopez-Marfil
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Universitat de Barcelona, Barcelona, Spain
| | - Diana O. Lopez-Cantu
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Dario R. Lemos
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| |
Collapse
|
16
|
Production of kidney organoids arranged around single ureteric bud trees, and containing endogenous blood vessels, solely from embryonic stem cells. Sci Rep 2022; 12:12573. [PMID: 35869233 PMCID: PMC9307805 DOI: 10.1038/s41598-022-16768-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 07/15/2022] [Indexed: 11/09/2022] Open
Abstract
There is intense worldwide effort in generating kidney organoids from pluripotent stem cells, for research, for disease modelling and, perhaps, for making transplantable organs. Organoids generated from pluripotent stem cells (PSC) possess accurate micro-anatomy, but they lack higher-organization. This is a problem, especially for transplantation, as such organoids will not be able to perform their physiological functions. In this study, we develop a method for generating murine kidney organoids with improved higher-order structure, through stages using chimaeras of ex-fetu and PSC-derived cells to a system that works entirely from embryonic stem cells. These organoids have nephrons organised around a single ureteric bud tree and also make vessels, with the endothelial network approaching podocytes.
Collapse
|
17
|
Karp S, Pollak MR, Subramanian B. Disease Modeling with Kidney Organoids. MICROMACHINES 2022; 13:1384. [PMID: 36144007 PMCID: PMC9506184 DOI: 10.3390/mi13091384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 06/16/2023]
Abstract
Kidney diseases often lack optimal treatments, causing millions of deaths each year. Thus, developing appropriate model systems to study human kidney disease is of utmost importance. Some of the most promising human kidney models are organoids or small organ-resembling tissue collectives, derived from human-induced pluripotent stem cells (hiPSCs). However, they are more akin to a first-trimester fetal kidney than an adult kidney. Therefore, new strategies are needed to advance their maturity. They have great potential for disease modeling and eventually auxiliary therapy if they can reach the maturity of an adult kidney. In this review, we will discuss the current state of kidney organoids in terms of their similarity to the human kidney and use as a disease modeling system thus far. We will then discuss potential pathways to advance the maturity of kidney organoids to match an adult kidney for more accurate human disease modeling.
Collapse
|
18
|
Renal organoid modeling of tuberous sclerosis complex reveals lesion features arise from diverse developmental processes. Cell Rep 2022; 40:111048. [PMID: 35793620 DOI: 10.1016/j.celrep.2022.111048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 04/15/2022] [Accepted: 06/13/2022] [Indexed: 02/06/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a multisystem tumor-forming disorder caused by loss of TSC1 or TSC2. Renal manifestations predominately include cysts and angiomyolipomas. Despite a well-described monogenic etiology, the cellular pathogenesis remains elusive. We report a genetically engineered human renal organoid model that recapitulates pleiotropic features of TSC kidney disease in vitro and upon orthotopic xenotransplantation. Time course single-cell RNA sequencing demonstrates that loss of TSC1 or TSC2 affects multiple developmental processes in the renal epithelial, stromal, and glial compartments. First, TSC1 or TSC2 ablation induces transitional upregulation of stromal-associated genes. Second, epithelial cells in the TSC1-/- and TSC2-/- organoids exhibit a rapamycin-insensitive epithelial-to-mesenchymal transition. Third, a melanocytic population forms exclusively in TSC1-/- and TSC2-/- organoids, branching from MITF+ Schwann cell precursors. Together, these results illustrate the pleiotropic developmental consequences of biallelic inactivation of TSC1 or TSC2 and offer insight into TSC kidney lesion pathogenesis.
Collapse
|
19
|
Suhito IR, Kim JW, Koo K, Nam SA, Kim YK, Kim T. In Situ Detection of Kidney Organoid Generation From Stem Cells Using a Simple Electrochemical Method. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200074. [PMID: 35506260 PMCID: PMC9284177 DOI: 10.1002/advs.202200074] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/13/2022] [Indexed: 05/31/2023]
Abstract
Organoids that mimic the structural and cellular characteristics of kidneys in vitro have recently emerged as a promising source for biomedical research. However, uncontrollable cellular heterogeneity after differentiation often results in the generation of off-target cells, one of the most challenging issues in organoid research. This study proposes a new method that enables the real-time assessment of kidney organoids derived from stem cells. When placed on a conductive surface, these organoids generate unique electrochemical signals at ≈0.3 V with intensities proportional to the amount of kidney-specific cell types. Off-target cells (i.e., non-kidney cells) produce an electrical signature at 0 V that is distinguishable from other surrounding cell types, enabling non-destructive assessment of both the differentiation, and maturation levels of kidney organoids. The developed platform can be applied to other types of organoids and is thus highly promising as a tool for organoid-based drug screening, toxicity assessment, and therapeutics.
Collapse
Affiliation(s)
| | - Jin Won Kim
- Cell Death Disease Research CenterCollege of MedicineThe Catholic University of KoreaSeoulKorea
- Department of Internal MedicineCollege of MedicineThe Catholic University of KoreaSt. Vincent's HospitalSuwonRepublic of Korea
| | - Kyeong‐Mo Koo
- School of Integrative EngineeringChung‐Ang UniversitySeoul06974Republic of Korea
| | - Sun Ah Nam
- Cell Death Disease Research CenterCollege of MedicineThe Catholic University of KoreaSeoulKorea
- Department of Internal MedicineCollege of MedicineThe Catholic University of KoreaSt. Vincent's HospitalSuwonRepublic of Korea
| | - Yong Kyun Kim
- Cell Death Disease Research CenterCollege of MedicineThe Catholic University of KoreaSeoulKorea
- Department of Internal MedicineCollege of MedicineThe Catholic University of KoreaSt. Vincent's HospitalSuwonRepublic of Korea
| | - Tae‐Hyung Kim
- School of Integrative EngineeringChung‐Ang UniversitySeoul06974Republic of Korea
| |
Collapse
|
20
|
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Rebecca A Ihrie
- Cell & Developmental Biology and Neurological Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Elizabeth P Henske
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|