{"title":"Stress granule formation as a marker of cellular toxicity in lung organoids","authors":"Seung-Yeon Kim, Kee K. Kim, Eun-Mi Kim","doi":"10.51335/organoid.2022.2.e28","DOIUrl":"https://doi.org/10.51335/organoid.2022.2.e28","url":null,"abstract":"Cells regulate protein synthesis under stressful circumstances by forming cytoplasmic RNA granules, termed stress granules (SGs). SGs are membrane-less organelles that function as a protective mechanism in response to stress. They function through liquid-liquid phase separation, which is a vital process comprising 2 distinct de-mixed liquid phases. The components of SGs, such as G3BP1, can serve as biomarkers of cell toxicity. Respiratory diseases are among the leading causes of death globally. After the humidifier disinfectant disaster in Korea in 2011, social concerns over respiratory disease-related deaths have been raised, and the importance of inhalation toxicity testing has been emphasized. Traditionally, in vivo animal models have been used to assess inhalation toxicity, but these models still have limitations owing to physiological differences between species. To overcome these limitations, human immortalized lung epithelial and lung cancer cell lines have been used to evaluate lung toxicity in vitro. Human stem cell-derived 3-dimensional organoid technology has recently been developed in various research fields, including lung toxicity. This review discusses SG-related proteins as potential biomarkers for lung toxicity assessment, especially in human lung organoids under stress conditions, such as exposure to toxic chemicals.","PeriodicalId":100198,"journal":{"name":"Brain Organoid and Systems Neuroscience Journal","volume":"45 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2022-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"90490692","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jooyeon Lee, Hyosin Baek, Seok-Ho Hong, Jong-Hee Lee, Seung-jun Wang, Ji Young Lee, Myung Ha Song, Se-Ran Yang
{"title":"Alveolar organoids: development of an in vitro assay to facilitate pulmonary toxicity assessments","authors":"Jooyeon Lee, Hyosin Baek, Seok-Ho Hong, Jong-Hee Lee, Seung-jun Wang, Ji Young Lee, Myung Ha Song, Se-Ran Yang","doi":"10.51335/organoid.2022.2.e31","DOIUrl":"https://doi.org/10.51335/organoid.2022.2.e31","url":null,"abstract":"Animal experiments have been performed to predict toxicity in humans in many fields, including toxicology, medicine, and pharmacology, and have contributed to increasing life expectancy. However, animal testing has been a controversial issue for over 100 years due to ethical concerns, and inter-species differences pose limitations for understanding human responses to toxicity. In recent years, many researchers have developed in vitro and in silico alternatives to using animals (e.g., 3-dimensional [3D] organoid culture, organs-on-a-chip, and advanced computer modeling). In this study, we generated 3D alveolar organoids (AOs) for pulmonary toxicity testing following exposure to chemicals, instead of animal models or two-dimensional culture of a single cell type. After human induced pluripotent stem cells were cultured with differentiation medium corresponding to each step for 14 days in 6-well plates, AOs were generated by forced aggregation and cultured with differentiation medium. The AOs were exposed to acrolein and sodium chromate for 24, 72, and 120 hours, and we determined the cytotoxicity of these chemicals using the MTT assay. Exposure to acrolein and sodium chromate for 24 hours decreased proliferation, but the organoid size did not change considerably. However, long-term exposure to acrolein and sodium chromate significantly decreased the organoid size. These findings suggest that AOs could facilitate acute toxicity assessments based on measurements of cell viability in AOs, as well as sub-chronic toxicity assessments based on measurements of both size and viability.","PeriodicalId":100198,"journal":{"name":"Brain Organoid and Systems Neuroscience Journal","volume":"14 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2022-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"82503287","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Generation of proximal tubule spheroids for nephrotoxicity assessment","authors":"D. Kim, J. Lim, Cho-Rok Jung, H. Kang","doi":"10.51335/organoid.2022.2.e30","DOIUrl":"https://doi.org/10.51335/organoid.2022.2.e30","url":null,"abstract":"To date, nephrotoxicity in new drug development has been evaluated through two-dimensional culture of representative cell lines, such as HK-2 and human proximal tubule epithelial cells (hPTECs). Approximately 20% of new drugs that were safe in preclinical studies were withdrawn from clinical trials due to nephrotoxicity, which means the current renal cell lines used in preclinical trials have limitations for the accurate detection of nephrotoxicity. Here, we established proximal tubule cell lines from immortalized mixed primary renal cells and generated functional proximal tubule cell spheroids, which expressed all apical basolateral transporters and showed epithelial polarity. Moreover, they showed a more sensitive drug response than hPTECs, which have been commonly used as in vitro kidney models. Taken together, the proximal tubule cells described in this study provide a more stable, reproducible, and accurate in vitro kidney model for predicting nephrotoxicity, which could help early compound development.","PeriodicalId":100198,"journal":{"name":"Brain Organoid and Systems Neuroscience Journal","volume":"25 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2022-10-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"87201614","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Seon Ju Mun, Jaeseo Lee, Yong-Moon Shin, Vincent Eun, Youngmi Ji, M. Son
{"title":"Recent advances in multicellular human liver models","authors":"Seon Ju Mun, Jaeseo Lee, Yong-Moon Shin, Vincent Eun, Youngmi Ji, M. Son","doi":"10.51335/organoid.2022.2.e26","DOIUrl":"https://doi.org/10.51335/organoid.2022.2.e26","url":null,"abstract":"The liver is the most important metabolic organ in the body. Model systems that recapitulate the complex organ structure and cell composition of the human liver are insufficient to study liver biology and to test toxicity and efficacy during new drug development. Recently established 3-dimensional liver models, including spheroids and organoids, organs-on-a-chip, bioprinting, and the decellularization/recellularization technique, have provided platforms that emulate the structural and functional characteristics of the human liver better than conventional 2-dimensional cell culture models and animal models. This review summarizes the architecture and cell compositions of human liver tissue, focusing on recent studies of multicellular human liver models that recapitulate in vivo-like physiologies with morphological and functional advances by the cellular communication of parenchymal and non-parenchymal cells. We discuss the applications, limitations, and future perspectives of advanced multicellular human liver models.","PeriodicalId":100198,"journal":{"name":"Brain Organoid and Systems Neuroscience Journal","volume":"73 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2022-10-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"85706999","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Toward brain organoid-based precision medicine in neurodegenerative diseases","authors":"Jong-Chan Park, I. Mook-Jung","doi":"10.51335/organoid.2022.2.e21","DOIUrl":"https://doi.org/10.51335/organoid.2022.2.e21","url":null,"abstract":"In recent years, the concept of precision medicine—an approach to developing personalized drugs for disease prevention or treatment—has emerged as an up-and-coming field in medical research. However, there are numerous limitations to applying this approach to the treatment of neurodegenerative diseases, such as Alzheimer disease, because of the invasiveness of obtaining human brain samples. Meanwhile, the development of human brain organoids has become one of the most powerful in vitro research tools because these organoids can be established from induced pluripotent stem cells or embryonic stem cells. In this review, we discuss how we can effectively utilize brain organoids for precision medicine research in conjunction with a variety of high-end techniques such as clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (CRISPR-Cas9) genomic editing, integrative multi-omics analysis, 3D brain tissue clearing, and high-content screening confocal microscopy imaging systems. We herein provide new insights in order to materialize organoid-based precision medicine therapy and future directions.","PeriodicalId":100198,"journal":{"name":"Brain Organoid and Systems Neuroscience Journal","volume":"33 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2022-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"88466466","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mai Tran Thi Nhu, Ulziituya Batjargal, H. Song, Jin-Man Kim, I. Kwak, Byoung-San Moon
{"title":"Generation of human-induced pluripotent stem cell-derived adherent 3-dimensional skin hair-follicle organoids","authors":"Mai Tran Thi Nhu, Ulziituya Batjargal, H. Song, Jin-Man Kim, I. Kwak, Byoung-San Moon","doi":"10.51335/organoid.2022.2.e23","DOIUrl":"https://doi.org/10.51335/organoid.2022.2.e23","url":null,"abstract":"Regenerating hair follicles (HFs) is a critical medical need for patients who suffer from serious hair loss. To generate equivalent hair-bearing skin systems that could mimic the complexity of native tissues with pluripotent stem cells, floating culture has been employed as a standard method; however, it is still necessary to improve limitations such as the heterogeneity of organoids and the difficulty of handling them, which increases during long-term culture. Here, we devise a floating-adherent combinatory culture system to establish skin HF organoids from human-induced pluripotent stem cells (hiPSCs). Specifically, embryoid bodies were generated in a free-floating environment, followed by the induction process, which occurred in adherent conditions. With our approach, hair germ-like buds were shown to protrude and extend faster. After 100 days of culture, mature cystic skin organoids stratified to form the epidermis, dermis, and outer root sheath, as evident from quantitative polymerase chain reaction and immunohistochemistry analysis. Dermal condensate cells (Sox2+, PDGFRα+, P75+), which are the precursors of HFs, were detected together with HF stem cells (NFATC1+, LGR5+), putative bulge stem cells (LHX2+, KRT15+) and melanocytes (PMEL+). Notably, our constructed HFs could recapitulate the sensory function of native tissues, as illustrated by the formation of a network of sensory neurons and Schwann cells connecting towards HF cells and epidermal progenitors. In summary, our results demonstrate a new protocol for the simplified and efficient induction of skin HFs from hiPSCs, thereby contributing to research on optimizing HF growth and investigating novel therapeutic strategies to treat alopecia.","PeriodicalId":100198,"journal":{"name":"Brain Organoid and Systems Neuroscience Journal","volume":"30 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2022-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"87076794","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Methods to identify epithelial stem cells","authors":"Youngtae Jeong","doi":"10.51335/organoid.2022.2.e24","DOIUrl":"https://doi.org/10.51335/organoid.2022.2.e24","url":null,"abstract":"Epithelial tissue is a tissue type that mainly covers the surfaces of the body and organs. Most epithelial tissues are composed of highly proliferative cells, necessitating robust stem cell activity. Epithelial tissue is also the most common site of cancers. Therefore, identifying epithelial stem cells and their self-renewal mechanisms is a prerequisite for promoting epithelial homeostasis, understanding cancer pathogenesis, and developing regenerative therapy and cancer prevention. Over the decades, diverse experimental techniques have been developed to identify epithelial stem cells in many organs and their self-renewal mechanisms from different angles. This review briefly introduces the various experimental methods used in stem cell identification, their rationales, and examples applying those tools to tissue stem cell identification.","PeriodicalId":100198,"journal":{"name":"Brain Organoid and Systems Neuroscience Journal","volume":"116 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2022-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"74424019","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Negative regulation of CDKN1A by the histone methyltransferase EHMT2 for cell growth in colorectal cancer","authors":"Kwangho Kim, Dae-Soo Kim, M. Son, Hyun-Soo Cho","doi":"10.51335/organoid.2022.2.e20","DOIUrl":"https://doi.org/10.51335/organoid.2022.2.e20","url":null,"abstract":"The epigenetic regulation of oncogenes and tumor suppressor genes by histone methyltransferases is an important process for colon cancer growth and metastasis. Although various epigenetic modifiers have been recognized as attractive therapeutic targets for colon cancer treatment, alternative epigenetic regulation in colon cancer for reducing side effects and increasing the effectiveness of treatments has not been thoroughly explored. In this study, we identified CDKN1A as a direct target for EHMT2 by RNA-sequencing and found increased growth suppression via upregulation of CDKN1A by EHMT2 knockdown. In addition, using a 3-dimensional culture system for spheroid formation with an ultralow attachment plate, we confirmed EHMT2-related growth suppression and CDKN1A regulation. Thus, we suggest that EHMT2 may be a therapeutic target for colon cancer treatment, and an EHMT2 inhibitor should be developed for the effective treatment of colon cancer.","PeriodicalId":100198,"journal":{"name":"Brain Organoid and Systems Neuroscience Journal","volume":"1 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2022-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"89508197","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Clinical applications and optimization of patient-derived organoids in intestinal diseases","authors":"Tae Il Kim","doi":"10.51335/organoid.2022.2.e22","DOIUrl":"https://doi.org/10.51335/organoid.2022.2.e22","url":null,"abstract":"Since the first successful establishment of organoids from adult intestinal stem cells, organoid technology has rapidly developed. With advances in normal organoid technology, intestinal disorders, such as colorectal tumors and inflammatory bowel disease, have been major target diseases for patient-derived organoid (PDO) development. PDO biobanking for colorectal cancer has subsequently been developed, and some reports have shown the possibility of using PDO models to predict anticancer drug responses. However, to apply these models to real-world practice, we need more long-term clinical follow-up data from further large-scale PDO biobanks, as well as advanced technology for more rapid and efficient PDO establishment. In addition, in the field of regenerative medicine, the implantation of healthy intestinal PDOs to refractory tissue defects could be a new treatment strategy to accelerate the healing and repair of mucosal defects. This PDO technology could also be applied to inflammatory bowel diseases and serve as a very useful model for drug development via high-throughput screening of useful candidate drugs.","PeriodicalId":100198,"journal":{"name":"Brain Organoid and Systems Neuroscience Journal","volume":"24 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2022-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"88392256","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"A brain metastasis model for breast cancer using human embryonic stem cell-derived cerebral organoids","authors":"M. Choe, M. Lee","doi":"10.51335/organoid.2022.2.e25","DOIUrl":"https://doi.org/10.51335/organoid.2022.2.e25","url":null,"abstract":"Background: Breast cancer is a common cause of brain metastasis. Although breast cancer has relatively high survival rates, its survival rate after metastasis to the brain is lower. Conventional two-dimensional cell culture models and animal models are widely used in metastatic cancer research, and these models have tremendously contributed to the understanding of this disease. However, these models have some limitations, such as different physiological features and genetic backgrounds.Methods: We established a simple metastatic breast cancer model using human pluripotent stem cell-derived cerebral organoids (COs)—in this case, breast cancer cerebral organoids (BC-COs).Results: Using the BC-CO model, we induced the metastasis of MDA-MB-231 cells into COs by co-culture of cells with COs and compared the differences between adapted cancer cells in BC-COs and non-adapted cells. Our results showed that the proliferative capacity increased in adapted cells. Additionally, the expression levels of endothelial-mesenchymal transition markers and cancer stem cells were significantly higher in adapted cancer cells.Conclusion: We conclude that metastasis promotes the metastatic capacity of breast cancer cells. Our results also showed that the BC-CO model could be a novel tool for research on brain metastasis in breast cancer.","PeriodicalId":100198,"journal":{"name":"Brain Organoid and Systems Neuroscience Journal","volume":"2016 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2022-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"78723764","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}