1
|
Xiao Y, Wang L, Li SX, Fang SS, Luo F, Chen SL, Zou X, Ye L, Hou W. Conditional reprogrammed human limbal epithelial cell model for anti-SARS-CoV-2 drug screening. Heliyon 2024; 10:e30044. [PMID: 38698981 PMCID: PMC11064458 DOI: 10.1016/j.heliyon.2024.e30044] [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: 02/06/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
To minimize the global pandemic COVID-19 spread, understanding the possible transmission routes of SARS-CoV-2 and discovery of novel antiviral drugs are necessary. We describe here that the virus can infect ocular surface limbal epithelial, but not other regions. Limbal supports wild type and mutant SARS-CoV-2 entry and replication depending on ACE2, TMPRSS2 and possibly other receptors, resulting in slight CPE and arising IL-6 secretion, which symbolizes conjunctivitis in clinical symptoms. With this limbal model, we have screened two natural product libraries and discovered several unreported drugs. Our data reveal important commonalities between COVID-19 and ocular infection with SARS-CoV-2, and establish an ideal cell model for drug screening and mechanism research.
Collapse
Affiliation(s)
- Yu Xiao
- Shenzhen Research Institute, Wuhan University, Shenzhen 518057, Guangdong Province, China
- Department of Clinical Laboratory, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Ling Wang
- Shenzhen Eye Hospital, Shenzhen 518040, Guangdong Province, China
| | - Shi-xu Li
- Shenzhen Eye Hospital, Shenzhen 518040, Guangdong Province, China
| | - Shi-song Fang
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, Guangdong Province, China
| | - Fan Luo
- State Key Laboratory of Virology/Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Shu-liang Chen
- State Key Laboratory of Virology/Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Xuan Zou
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, Guangdong Province, China
| | - Lin Ye
- Shenzhen Eye Hospital, Shenzhen 518040, Guangdong Province, China
| | - Wei Hou
- Shenzhen Research Institute, Wuhan University, Shenzhen 518057, Guangdong Province, China
- State Key Laboratory of Virology/Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
2
|
Zhou Z, Zhou X, Zhang Y, Yang Y, Wang L, Wu Z. Butyric acid inhibits oxidative stress and inflammation injury in calcium oxalate nephrolithiasis by targeting CYP2C9. Food Chem Toxicol 2023:113925. [PMID: 37414240 DOI: 10.1016/j.fct.2023.113925] [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: 01/29/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023]
Abstract
This study investigates the mechanism by which butyric acid can protect against calcium oxalate (CaOx) nephrolithiasis. To do so, a rat model was used with 0.75% ethylene glycol administration to induce CaOx crystal formation. Histological and von Kossa staining revealed calcium deposits and renal injury, while dihydroethidium fluorescence staining was used to detect reactive oxygen species (ROS) levels. Flow cytometry and TUNEL assays were used to assess apoptosis, respectively. Treatment with sodium butyrate (NaB) was found to partially reverse the oxidative stress, inflammation, and apoptosis associated with CaOx crystallization in the kidney. In addition, in HK-2 cells, NaB reversed the decreased cell viability, increased ROS levels and apoptosis damage caused by oxalate exposure. Network pharmacology was employed to predict the target genes of butyric acid, CYP2C9. Subsequently, NaB was found to significantly reduce CYP2C9 levels in vivo and in vitro, and inhibition of CYP2C9 by Sulfaphenazole (a specific CYP2C9 inhibitor), was able to reduce ROS levels, inflammation injury, and apoptosis in oxalate-induced HK-2 cells. Collectively, these findings suggest that butyric acid may inhibit oxidative stress and reduce inflammation injury in CaOx nephrolithiasis by suppressing CYP2C9.
Collapse
Affiliation(s)
- Zijian Zhou
- Department of Urology, Huashan Hospital & Institute of Urology, Fudan University, Shanghai, 200040, PR China; Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, 200040, PR China
| | - Xuan Zhou
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Yu Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
| | - Yuanyuan Yang
- Department of Urology, Huashan Hospital & Institute of Urology, Fudan University, Shanghai, 200040, PR China; Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, 200040, PR China
| | - Lujia Wang
- Department of Urology, Huashan Hospital & Institute of Urology, Fudan University, Shanghai, 200040, PR China; Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, 200040, PR China.
| | - Zhong Wu
- Department of Urology, Huashan Hospital & Institute of Urology, Fudan University, Shanghai, 200040, PR China; Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, 200040, PR China.
| |
Collapse
|
3
|
Krawczyk E, Kitlińska J. Preclinical Models of Neuroblastoma-Current Status and Perspectives. Cancers (Basel) 2023; 15:3314. [PMID: 37444423 PMCID: PMC10340830 DOI: 10.3390/cancers15133314] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Preclinical in vitro and in vivo models remain indispensable tools in cancer research. These classic models, including two- and three-dimensional cell culture techniques and animal models, are crucial for basic and translational studies. However, each model has its own limitations and typically does not fully recapitulate the course of the human disease. Therefore, there is an urgent need for the development of novel, advanced systems that can allow for efficient evaluation of the mechanisms underlying cancer development and progression, more accurately reflect the disease pathophysiology and complexity, and effectively inform therapeutic decisions for patients. Preclinical models are especially important for rare cancers, such as neuroblastoma, where the availability of patient-derived specimens that could be used for potential therapy evaluation and screening is limited. Neuroblastoma modeling is further complicated by the disease heterogeneity. In this review, we present the current status of preclinical models for neuroblastoma research, discuss their development and characteristics emphasizing strengths and limitations, and describe the necessity of the development of novel, more advanced and clinically relevant approaches.
Collapse
Affiliation(s)
- Ewa Krawczyk
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Joanna Kitlińska
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
4
|
Xia S, Wu M, Zhou X, Zhang X, Ye L, Zhang K, Kang Y, Liu J, Zhang Y, Wu W, Dong D, Chen H, Li H. Treating intrauterine adhesion using conditionally reprogrammed physiological endometrial epithelial cells. Stem Cell Res Ther 2022; 13:178. [PMID: 35505443 PMCID: PMC9066886 DOI: 10.1186/s13287-022-02860-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 04/21/2022] [Indexed: 01/18/2023] Open
Abstract
Background There is unmet need for effective therapies of intrauterine adhesions (IUAs) that are common cause of menstrual disturbance and infertility, since current clinical procedures do not improve prognosis for patients with moderate to severe IUA, with a recurrence rate of 23–50%. Stem cell-based therapy has emerged as a therapeutic option with unsolved issues for IUA patients in the past few years. Primary endometrial epithelial cells for cell therapy are largely hampered with the extremely limited proliferation capacity of uterine epithelial cells. This study was to evaluate whether IUA is curable with conditionally reprogrammed (CR) endometrial epithelial cells. Methods Mouse endometrial epithelial cells (MEECs) were isolated from C57BL female mice, and long-term cultures of MEECs were established and maintained with conditional reprogramming (CR) method. DNA damage response analysis, soft agar assay, and matrigel 3D culture were carried out to determine the normal biological characteristics of CR-MEECs. The tissue-specific differentiation potential of MEECs was analyzed with air–liquid interface (ALI) 3D culture, hematoxylin and eosin (H&E) staining, Masson’s trichrome and DAB staining, immunofluorescence assay. IUA mice were constructed and transplanted with CR-MEECs. Repair and mechanisms of MEECs transplantation in IUA mice were measured with qRT-PCR, Masson’s trichrome, and DAB staining. Results We first successfully established long-term cultures of MEECs using CR approach. CR-MEECs maintained a rapid and stable proliferation in this co-culture system. Our data confirmed that CR-MEECs retained normal biological characteristics and endometrium tissue-specific differentiation potential. CR-MEECs also expressed estrogen and progesterone receptors and maintained the exquisite sensitivity to sex hormones in vitro. Most importantly, allogeneic transplantation of CR-MEECs successfully repaired the injured endometrium and significantly improved the pregnancy rate of IUA mice. Conclusions Conditionally reprogrammed physiological endometrial epithelial cells provide a novel strategy in IUA clinics in a personalized or generalized manner and also serve as a physiological model to explore biology of endometrial epithelial cells and mechanisms of IUA.
Collapse
Affiliation(s)
- Siyu Xia
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Ming Wu
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Xinhao Zhou
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Xiu Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Lina Ye
- Wuhan University Shenzhen Institute, Shenzhen, 518057, Guangdong, China
| | - Kang Zhang
- Wuhan University Shenzhen Institute, Shenzhen, 518057, Guangdong, China
| | - Yiyi Kang
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Jun Liu
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Yunci Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Wang Wu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Dirong Dong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Hong Chen
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| | - Hui Li
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China. .,Wuhan University Shenzhen Institute, Shenzhen, 518057, Guangdong, China.
| |
Collapse
|
5
|
Morgan R, Manfredi C, Easley KF, Watkins LD, Hunt WR, Goudy SL, Sorscher EJ, Koval M, Molina SA. A medium composition containing normal resting glucose that supports differentiation of primary human airway cells. Sci Rep 2022; 12:1540. [PMID: 35087167 PMCID: PMC8795386 DOI: 10.1038/s41598-022-05446-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 01/10/2022] [Indexed: 02/07/2023] Open
Abstract
Primary cells isolated from the human respiratory tract are the state-of-the-art for in vitro airway epithelial cell research. Airway cell isolates require media that support expansion of cells in a basal state to maintain the capacity for differentiation as well as proper cellular function. By contrast, airway cell differentiation at an air-liquid interface (ALI) requires a distinct medium formulation that typically contains high levels of glucose. Here, we expanded and differentiated human basal cells isolated from the nasal and conducting airway to a mature mucociliary epithelial cell layer at ALI using a medium formulation containing normal resting glucose levels. Of note, bronchial epithelial cells expanded and differentiated in normal resting glucose medium showed insulin-stimulated glucose uptake which was inhibited by high glucose concentrations. Normal glucose containing ALI also enabled differentiation of nasal and tracheal cells that showed comparable electrophysiological profiles when assessed for cystic fibrosis transmembrane conductance regulator (CFTR) function and that remained responsive for up to 7 weeks in culture. These data demonstrate that normal glucose containing medium supports differentiation of primary nasal and lung epithelial cells at ALI, is well suited for metabolic studies, and avoids pitfalls associated with exposure to high glucose.
Collapse
Affiliation(s)
- Rachel Morgan
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Candela Manfredi
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Kristen F Easley
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Lionel D Watkins
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| | - William R Hunt
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Steven L Goudy
- Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Eric J Sorscher
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Michael Koval
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA.
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Samuel A Molina
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| |
Collapse
|
6
|
Xu L, Zhao Q, Li D, Luo J, Ma W, Jin Y, Li C, Chen J, Zhao K, Zheng Y, Yu D. MicroRNA-760 resists ambient PM 2.5-induced apoptosis in human bronchial epithelial cells through elevating heme-oxygenase 1 expression. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 284:117213. [PMID: 33933780 DOI: 10.1016/j.envpol.2021.117213] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/31/2021] [Accepted: 04/19/2021] [Indexed: 06/12/2023]
Abstract
PM2.5 (particles matter smaller aerodynamic diameter of 2.5 μm) exposure, a major environmental risk factor for the global burden of diseases, is associated with high risks of respiratory diseases. Heme-oxygenase 1 (HMOX1) is one of the major molecular antioxidant defenses to mediate cytoprotective effects against diverse stressors, including PM2.5-induced toxicity; however, the regulatory mechanism of HMOX1 expression still needs to be elucidated. In this study, using PM2.5 as a typical stressor, we explored whether microRNAs (miRNAs) might modulate HMOX1 expression in lung cells. Systematic bioinformatics analysis showed that seven miRNAs have the potentials to target HMOX1 gene. Among these, hsa-miR-760 was identified as the most responsive miRNA to PM2.5 exposure. More importantly, we revealed a "non-conventional" miRNA function in hsa-miR-760 upregulating HMOX1 expression, by targeting the coding region and interacting with YBX1 protein. In addition, we observed that exogenous hsa-miR-760 effectively elevated HMOX1 expression, reduced the reactive oxygen agents (ROS) levels, and rescued the lung cells from PM2.5-induced apoptosis. Our results revealed that hsa-miR-760 might play an important role in protecting lung cells against PM2.5-induced toxicity, by elevating HMOX1 expression, and offered new clues to elucidate the diverse functions of miRNAs.
Collapse
Affiliation(s)
- Lin Xu
- School of Public Health, Qingdao University, Qingdao, China
| | - Qianwen Zhao
- School of Public Health, Qingdao University, Qingdao, China
| | - Daochuan Li
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jiao Luo
- School of Public Health, Qingdao University, Qingdao, China
| | - Wanli Ma
- School of Public Health, Qingdao University, Qingdao, China
| | - Yuan Jin
- School of Public Health, Qingdao University, Qingdao, China
| | - Chuanhai Li
- School of Public Health, Qingdao University, Qingdao, China
| | - Jing Chen
- School of Public Health, Qingdao University, Qingdao, China
| | - Kunming Zhao
- School of Public Health, Qingdao University, Qingdao, China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao, China
| | - Dianke Yu
- School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
7
|
Zhang C, Wang X, Pi S, Wei Z, Wang C, Yang F, Li G, Nie G, Hu G. Cadmium and molybdenum co-exposure triggers autophagy via CYP450s/ROS pathway in duck renal tubular epithelial cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 759:143570. [PMID: 33243500 DOI: 10.1016/j.scitotenv.2020.143570] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/15/2020] [Accepted: 10/26/2020] [Indexed: 06/11/2023]
Abstract
Cadmium (Cd) and excessive molybdenum (Mo) are detrimental to animals, but the combined nephrotoxic impacts of Cd and Mo on duck are still unclear. To evaluate the combined impacts of Cd and Mo on autophagy via Cytochrome P450s (CYP450s)/reactive oxygen species (ROS) pathway, duck renal tubular epithelial cells were treated with 3CdSO4·8H2O (4.0 μM Cd), (NH4)6Mo7O24·4H2O (500.0 μM Mo), butylated hydroxy anisole (BHA) (100.0 μM) and combination of Cd and Mo or Cd, Mo and BHA for 12 h, and combined cytotoxicity was investigated. The results indicated that Mo or/and Cd induced CYP1A1, CYP1B1, CYP2C9, CYP3A8 and CYP4B1 mRNA levels, decreased superoxide dismutase (SOD), catalase (CAT) activities and glutathione peroxidase (GSH-Px) content, and increased malondialdehyde (MDA) and hydrogen peroxide (H2O2) contents. Besides, Mo or/and Cd elevated the number of autophagosome and microtubule-associated protein light chain 3 (LC3) puncta, upregulated mRNA levels of Beclin-1, LC3A, LC3B, Atg5 and adenosine 5'-monophosphate (AMP)-activated protein kinase α1 (AMPKα-1), inhibited Dynein, p62 and mammalian target of rapamycin (mTOR) mRNA levels, increased Beclin-1 and LC3II/LC3I protein levels. Moreover, the changes of these factors in Mo and Cd co-treated groups were more apparent. Additionally, BHA could efficiently alleviate the changes of above these indicators co-induced by Mo and Cd. Overall, these results manifest Cd and Mo co-exposure may synergistically trigger autophagy via CYP450s/ROS pathway in duck renal tubular epithelial cells.
Collapse
Affiliation(s)
- Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Xueru Wang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Shaoxing Pi
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Zejing Wei
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Chang Wang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Fan Yang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Guyue Li
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Gaohui Nie
- School of Information Technology, Jiangxi University of Finance and Economics, No. 665 Yuping West street, Economic and Technological Development District, Nanchang 330032, Jiangxi, PR China.
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China.
| |
Collapse
|
8
|
Liu X, Mondal AM. Conditional cell reprogramming for modeling host-virus interactions and human viral diseases. J Med Virol 2020; 92:2440-2452. [PMID: 32478897 PMCID: PMC7586785 DOI: 10.1002/jmv.26093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 01/08/2023]
Abstract
Conventional cancer and transformed cell lines are widely used in cancer biology and other fields within biology. These cells usually have abnormalities from the original tumor itself, but may also develop abnormalities due to genetic manipulation, or genetic and epigenetic changes during long-term passages. Primary cultures may maintain lineage functions as the original tissue types, yet they have a very limited life span or population doubling time because of the nature of cellular senescence. Primary cultures usually have very low yields, and the high variability from any original tissue specimens, largely limiting their applications in research. Animal models are often used for studies of virus infections, disease modeling, development of antiviral drugs, and vaccines. Human viruses often need a series of passages in vivo to adapt to the host environment because of variable receptors on the cell surface and may have intracellular restrictions from the cell types or host species. Here, we describe a long-term cell culture system, conditionally reprogrammed cells (CRCs), and its applications in modeling human viral diseases and drug discovery. Using feeder layer coculture in presence of Y-27632 (conditional reprogramming, CR), CRCs can be obtained and rapidly propagated from surgical specimens, core or needle biopsies, and other minimally invasive or noninvasive specimens, for example, nasal cavity brushing. CRCs preserve their lineage functions and provide biologically relevant and physiological conditions, which are suitable for studies of viral entry and replication, innate immune responses of host cells, and discovery of antiviral drugs. In this review, we summarize the applications of CR technology in modeling host-virus interactions and human viral diseases including severe acute respiratory syndrome coronavirus-2 and coronavirus disease-2019, and antiviral discovery.
Collapse
Affiliation(s)
- Xuefeng Liu
- Department of Pathology, Center for Cell ReprogrammingGeorgetown University Medical CenterWashingtonDC
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| | - Abdul M. Mondal
- Department of Pathology, Center for Cell ReprogrammingGeorgetown University Medical CenterWashingtonDC
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| |
Collapse
|
9
|
Wu X, Wang S, Li M, Li J, Shen J, Zhao Y, Pang J, Wen Q, Chen M, Wei B, Kaboli PJ, Du F, Zhao Q, Cho CH, Wang Y, Xiao Z, Wu X. Conditional reprogramming: next generation cell culture. Acta Pharm Sin B 2020; 10:1360-1381. [PMID: 32963937 PMCID: PMC7488362 DOI: 10.1016/j.apsb.2020.01.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
Long-term primary culture of mammalian cells has been always difficult due to unavoidable senescence. Conventional methods for generating immortalized cell lines usually require manipulation of genome which leads to change of important biological and genetic characteristics. Recently, conditional reprogramming (CR) emerges as a novel next generation tool for long-term culture of primary epithelium cells derived from almost all origins without alteration of genetic background of primary cells. CR co-cultures primary cells with inactivated mouse 3T3-J2 fibroblasts in the presence of RHO-related protein kinase (ROCK) inhibitor Y-27632, enabling primary cells to acquire stem-like characteristics while retain their ability to fully differentiate. With only a few years' development, CR shows broad prospects in applications in varied areas including disease modeling, regenerative medicine, drug evaluation, drug discovery as well as precision medicine. This review is thus to comprehensively summarize and assess current progress in understanding mechanism of CR and its wide applications, highlighting the value of CR in both basic and translational researches and discussing the challenges faced with CR.
Collapse
Key Words
- 3T3-J2 fibroblast
- AACR, American Association for Cancer Research
- ACC, adenoid cystic carcinoma
- AR, androgen receptor
- CFTR, cystic fibrosis transmembrane conductance regulators
- CR, conditional reprogramming
- CYPs, cytochrome P450 enzymes
- Conditional reprogramming
- DCIS, ductal carcinoma in situ
- ECM, extracellular matrix
- ESC, embryonic stem cell
- HCMI, human cancer model initiatives
- HGF, hepatocyte growth factor
- HNE, human nasal epithelial
- HPV, human papillomaviruses
- ICD, intracellular domain
- LECs, limbal epithelial cells
- NCI, National Cancer Institute
- NGFR, nerve growth factor receptor
- NSCLC, non-small cell lung cancer
- NSG, NOD/SCID/gamma
- PDAC, pancreatic ductal adenocarcinoma
- PDX, patient derived xenograft
- PP2A, protein phosphatase 2A
- RB, retinoblastoma-associated protein
- ROCK
- ROCK, Rho kinase
- SV40, simian virus 40 large tumor antigen
- Senescence
- UVB, ultraviolet radiation b
- Y-27632
- dECM, decellularized extracellular matrix
- hASC, human adipose stem cells
- hTERT, human telomerase reverse transcriptase
- iPSCs, induction of pluripotent stem cells
- ΔNP63α, N-terminal truncated form of P63α
Collapse
Affiliation(s)
- Xiaoxiao Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Jing Li
- Department of Oncology and Hematology, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Jun Pang
- Center of Radiation Oncology, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou 646000, China
| | - Qinglian Wen
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou 646000, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Bin Wei
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Parham Jabbarzadeh Kaboli
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Qijie Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| |
Collapse
|
10
|
Coupled CRC 2D and ALI 3D Cultures Express Receptors of Emerging Viruses and Are More Suitable for the Study of Viral Infections Compared to Conventional Cell Lines. Stem Cells Int 2020; 2020:2421689. [PMID: 32695180 PMCID: PMC7368225 DOI: 10.1155/2020/2421689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 02/02/2023] Open
Abstract
Infections of emerging and reemerging viruses (SARS-CoVs, influenza H1N1, etc.) largely and globally affect human health. Animal models often fail to reflect a physiological status because of species tropism of virus infection. Conventional cell lines are usually genetically and phenotypically different from primary cells. Developing an in vitro physiological model to study the infection of emerging viruses will facilitate our understanding of virus-host cell interactions, thereby benefiting antiviral drug discovery. In the current work, we first established normal airway epithelial cells (upper and lower airway track) in 2D and 3D culture systems using conditional reprogramming (CR) and air-liquid interface (ALI) techniques. These long-term cultures maintained differentiation potential. More importantly, these cells express two types of influenza virus receptors, α2-6-Gal- and α2-3-Gal-linked sialic acids, and angiotensin-converting enzyme 2 (ACE2), a receptor for SARS-CoVs as well. These cells were permissive to the infection of pandemic influenza H1N1 (H1N1pdm). In contrast, the lung cancer cell line A549 and immortalized airway epithelial cells (16HBE) were not susceptible to H1N1 infection. A virus-induced cytopathic effect (CPE) on 2D CRC cultures developed in a time-dependent manner. The pathological effects were also readily observed spreading from the apical layer to the basal layer of the 3D ALI culture. This integrated 2D CRC and 3D ALI cultures provide a physiological and personalized in vitro model to study the infection of emerging viruses. This novel model can be used for studying virus biology and host response to viral infection and for antiviral drug discovery.
Collapse
|
11
|
Xia S, Wu M, Chen S, Zhang T, Ye L, Liu J, Li H. Long Term Culture of Human Kidney Proximal Tubule Epithelial Cells Maintains Lineage Functions and Serves as an Ex vivo Model for Coronavirus Associated Kidney Injury. Virol Sin 2020; 35:311-320. [PMID: 32602046 PMCID: PMC7322379 DOI: 10.1007/s12250-020-00253-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/09/2020] [Indexed: 12/15/2022] Open
Abstract
The mechanism of how SARS-CoV-2 causes severe multi-organ failure is largely unknown. Acute kidney injury (AKI) is one of the frequent organ damage in severe COVID-19 patients. Previous studies have shown that human renal tubule cells could be the potential host cells targeted by SARS-CoV-2. Traditional cancer cell lines or immortalized cell lines are genetically and phenotypically different from host cells. Animal models are widely used, but often fail to reflect a physiological and pathogenic status because of species tropisms. There is an unmet need for normal human epithelial cells for disease modeling. In this study, we successfully established long term cultures of normal human kidney proximal tubule epithelial cells (KPTECs) in 2D and 3D culture systems using conditional reprogramming (CR) and organoids techniques. These cells had the ability to differentiate and repair DNA damage, and showed no transforming property. Importantly, the CR KPTECs maintained lineage function with expression of specific transporters (SLC34A3 and cubilin). They also expressed angiotensin-converting enzyme 2 (ACE2), a receptor for SARS-CoV and SARS-CoV-2. In contrast, cancer cell line did not express endogenous SLC34A3, cubilin and ACE2. Very interestingly, ACE2 expression was around twofold higher in 3D organoids culture compared to that in 2D CR culture condition. Pseudovirion assays demonstrated that SARS-CoV spike (S) protein was able to enter CR cells with luciferase reporter. This integrated 2D CR and 3D organoid cultures provide a physiological ex vivo model to study kidney functions, innate immune response of kidney cells to viruses, and a novel platform for drug discovery and safety evaluation.
Collapse
Affiliation(s)
- Siyu Xia
- State Key Laboratory of Virology/Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Ming Wu
- State Key Laboratory of Virology/Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Si Chen
- State Key Laboratory of Virology/Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Tao Zhang
- State Key Laboratory of Virology/Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lina Ye
- Wuhan University Shenzhen Institute, Shenzhen, 518057, China
| | - Jun Liu
- State Key Laboratory of Virology/Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Hui Li
- State Key Laboratory of Virology/Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China. .,Wuhan University Shenzhen Institute, Shenzhen, 518057, China.
| |
Collapse
|
12
|
Zhong M, Fu L. Culture and application of conditionally reprogrammed primary tumor cells. Gastroenterol Rep (Oxf) 2020; 8:224-233. [PMID: 32665854 PMCID: PMC7333928 DOI: 10.1093/gastro/goaa023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is still a major public-health problem that threatens human life worldwide and further study needs to be carried out in the basic and preclinical areas. Although high-throughput sequencing technology and individualized precise therapy have made breakthroughs over the years, the high failure rate of clinical translational research has limited the innovation of antitumor drugs and triggered the urgent need for optimal cancer-research models. The development of cancerous cell lines, patient-derived xenograft (PDX) models, and organoid has strongly promoted the development of tumor-biology research, but the prediction values are limited. Conditional reprogramming (CR) is a novel cell-culture method for cancer research combining feeder cells with a Rho-associated coiled-coil kinase (ROCK) inhibitor, which enables the rapid and continuous proliferation of primary epithelial cells. In this review, we summarize the methodology to establish CR model and overview recent functions and applications of CR cell-culture models in cancer research with regard to the study of cancer-biology characterization, the exploration of therapeutic targets, individualized drug screening, the illumination of mechanisms about response to antitumor drugs, and the improvement of patient-derived animal models, and finally discuss in detail the major limitations of this cell-culture system.
Collapse
Affiliation(s)
- Mengjun Zhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
13
|
Liu W, Ju L, Cheng S, Wang G, Qian K, Liu X, Xiao Y, Wang X. Conditional reprogramming: Modeling urological cancer and translation to clinics. Clin Transl Med 2020; 10:e95. [PMID: 32508060 PMCID: PMC7403683 DOI: 10.1002/ctm2.95] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022] Open
Abstract
Patient-derived models, including cell models (organoids and conditionally reprogrammed cells [CRCs]) and patient-derived xenografts, are urgently needed for both basic and translational cancer research. Conditional reprogramming (CR) technique refers to a co-culture system of primary human normal or tumor cells with irradiated murine fibroblasts in the presence of a Rho-associated kinase inhibitor to allow the primary cells to acquire stem cell properties and the ability to proliferate indefinitely in vitro without any exogenous gene or viral transfection. Considering its robust features, the CR technique may facilitate cancer research in many aspects. Under in vitro culturing, malignant CRCs can share certain genetic aberrations and tumor phenotypes with their parental specimens. Thus, tumor CRCs can promisingly be utilized for the study of cancer biology, the discovery of novel therapies, and the promotion of precision medicine. For normal CRCs, the characteristics of normal karyotype maintenance and lineage commitment suggest their potential in toxicity testing and regenerative medicine. In this review, we discuss the applications, limitations, and future potential of CRCs in modeling urological cancer and translation to clinics.
Collapse
Affiliation(s)
- Wei Liu
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Lingao Ju
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhanChina
- Human Genetic Resources Preservation Center of Hubei ProvinceWuhanChina
| | - Songtao Cheng
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Gang Wang
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhanChina
- Human Genetic Resources Preservation Center of Hubei ProvinceWuhanChina
| | - Kaiyu Qian
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhanChina
- Human Genetic Resources Preservation Center of Hubei ProvinceWuhanChina
| | - Xuefeng Liu
- Department of Pathology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| | - Yu Xiao
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhanChina
- Human Genetic Resources Preservation Center of Hubei ProvinceWuhanChina
| | - Xinghuan Wang
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Medical Research InstituteWuhan UniversityWuhanChina
| |
Collapse
|
14
|
Liu X, Wu Y, Rong L. Conditionally Reprogrammed Human Normal Airway Epithelial Cells at ALI: A Physiological Model for Emerging Viruses. Virol Sin 2020; 35:280-289. [PMID: 32557270 PMCID: PMC7298165 DOI: 10.1007/s12250-020-00244-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/22/2020] [Indexed: 01/08/2023] Open
Abstract
Cancer cell lines have been used widely in cancer biology, and as biological or functional cell systems in many biomedical research fields. These cells are usually defective for many normal activities or functions due to significant genetic and epigenetic changes. Normal primary cell yields and viability from any original tissue specimens are usually relatively low or highly variable. These normal cells cease after a few passages or population doublings due to very limited proliferative capacity. Animal models (ferret, mouse, etc.) are often used to study virus-host interaction. However, viruses usually need to be adapted to the animals by several passages due to tropism restrictions including viral receptors and intracellular restrictions. Here we summarize applications of conditionally reprogrammed cells (CRCs), long-term cultures of normal airway epithelial cells from human nose to lung generated by conditional cell reprogramming (CR) technology, as an ex vivo model in studies of emerging viruses. CR allows to robustly propagate cells from non-invasive or minimally invasive specimens, for example, nasal or endobronchial brushing. This process is rapid (2 days) and conditional. The CRCs maintain their differentiation potential and lineage functions, and have been used for studies of adenovirus, rhinovirus, respiratory syncytial virus, influenza viruses, parvovirus, and SARS-CoV. The CRCs can be easily used for air-liquid interface (ALI) polarized 3D cultures, and these coupled CRC/ALI cultures mimic physiological conditions and are suitable for studies of viral entry including receptor binding and internalization, innate immune responses, viral replications, and drug discovery as an ex vivo model for emerging viruses.
Collapse
Affiliation(s)
- Xuefeng Liu
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA.
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.
| | - Yuntao Wu
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, 20110, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinoi at Chicago, Chicago, IL, 60612, USA
| |
Collapse
|
15
|
Krawczyk E, Hong SH, Galli S, Trinh E, Wietlisbach L, Misiukiewicz SF, Tilan JU, Chen YS, Schlegel R, Kitlinska J. Murine neuroblastoma cell lines developed by conditional reprogramming preserve heterogeneous phenotypes observed in vivo. J Transl Med 2020; 100:38-51. [PMID: 31409888 PMCID: PMC6920526 DOI: 10.1038/s41374-019-0297-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 06/14/2019] [Accepted: 06/20/2019] [Indexed: 12/19/2022] Open
Abstract
Neuroblastoma (NB) is a pediatric tumor of the peripheral nervous system. Treatment of the disease represents an unsolved clinical problem, as survival of patients with aggressive form of NB remains below 50%. Despite recent identification of numerous potential therapeutic targets, clinical trials validating them are challenging due to the rarity of the disease and its high patient-to-patient heterogeneity. Hence, there is a need for the accurate preclinical models that would allow testing novel therapeutic approaches and prioritizing the clinical studies, preferentially in personalized way. Here, we propose using conditional reprogramming (CR) technology for rapid development of primary NB cell cultures that could become a new model for such tests. This newly established method allowed for indefinite propagation of normal and tumor cells of epithelial origin in an undifferentiated state by their culture in the presence of Rho-associated kinase (ROCK) inhibitor, Y-27632, and irradiated mouse feeder cells. Using a modification of this approach, we isolated cell lines from tumors arising in the TH-MYCN murine transgenic model of NB (CR-NB). The cells were positive for neuronal markers, including Phox2B and peripherin and consisted of two distinct populations: mesenchymal and adrenergic expressing corresponding markers of their specific lineage. This heterogeneity of the CR-NB cells mimicked the different tumor cell phenotypes in TH-MYCN tumor tissues. The CR-NB cells preserved anchorage-independent growth capability and were successfully passaged, frozen and biobanked. Further studies are required to determine the utility of this method for isolation of human NB cultures, which can become a novel model for basic, translational, and clinical research, including individualized drug testing.
Collapse
Affiliation(s)
- Ewa Krawczyk
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington DC, USA.
| | - Sung-Hyeok Hong
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington DC
| | - Susana Galli
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington DC
| | - Emily Trinh
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington DC
| | - Larissa Wietlisbach
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington DC
| | - Sara F. Misiukiewicz
- Human Science Department, School of Nursing and Health Studies, Georgetown University Medical Center, Washington DC
| | - Jason U. Tilan
- Human Science Department, School of Nursing and Health Studies, Georgetown University Medical Center, Washington DC
| | - You-Shin Chen
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington DC
| | - Richard Schlegel
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington DC
| | - Joanna Kitlinska
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington DC
| |
Collapse
|
16
|
Palechor-Ceron N, Krawczyk E, Dakic A, Simic V, Yuan H, Blancato J, Wang W, Hubbard F, Zheng YL, Dan H, Strome S, Cullen K, Davidson B, Deeken JF, Choudhury S, Ahn PH, Agarwal S, Zhou X, Schlegel R, Furth PA, Pan CX, Liu X. Conditional Reprogramming for Patient-Derived Cancer Models and Next-Generation Living Biobanks. Cells 2019; 8:E1327. [PMID: 31717887 PMCID: PMC6912808 DOI: 10.3390/cells8111327] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/14/2019] [Accepted: 10/25/2019] [Indexed: 12/21/2022] Open
Abstract
Traditional cancer models including cell lines and animal models have limited applications in both basic and clinical cancer research. Genomics-based precision oncology only help 2-20% patients with solid cancer. Functional diagnostics and patient-derived cancer models are needed for precision cancer biology. In this review, we will summarize applications of conditional cell reprogramming (CR) in cancer research and next generation living biobanks (NGLB). Together with organoids, CR has been cited in two NCI (National Cancer Institute, USA) programs (PDMR: patient-derived cancer model repository; HCMI: human cancer model initiatives. HCMI will be distributed through ATCC). Briefly, the CR method is a simple co-culture technology with a Rho kinase inhibitor, Y-27632, in combination with fibroblast feeder cells, which allows us to rapidly expand both normal and malignant epithelial cells from diverse anatomic sites and mammalian species and does not require transfection with exogenous viral or cellular genes. Establishment of CR cells from both normal and tumor tissue is highly efficient. The robust nature of the technique is exemplified by the ability to produce 2 × 106 cells in five days from a core biopsy of tumor tissue. Normal CR cell cultures retain a normal karyotype and differentiation potential and CR cells derived from tumors retain their tumorigenic phenotype. CR also allows us to enrich cancer cells from urine (for bladder cancer), blood (for prostate cancer), and pleural effusion (for non-small cell lung carcinoma). The ability to produce inexhaustible cell populations using CR technology from small biopsies and cryopreserved specimens has the potential to transform biobanking repositories (NGLB: next-generation living biobank) and current pathology practice by enabling genetic, biochemical, metabolomic, proteomic, and biological assays, including chemosensitivity testing as a functional diagnostics tool for precision cancer medicine. We discussed analyses of patient-derived matched normal and tumor models using a case with tongue squamous cell carcinoma as an example. Last, we summarized applications in cancer research, disease modeling, drug discovery, and regenerative medicine of CR-based NGLB.
Collapse
Affiliation(s)
- Nancy Palechor-Ceron
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Ewa Krawczyk
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Aleksandra Dakic
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Vera Simic
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Hang Yuan
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Jan Blancato
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (J.B.); (W.W.); (Y.-L.Z.); (P.A.F.)
| | - Weisheng Wang
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (J.B.); (W.W.); (Y.-L.Z.); (P.A.F.)
| | - Fleesie Hubbard
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland, Baltimore, MD 21201, USA; (F.H.); (H.D.); (S.S.); (K.C.)
| | - Yun-Ling Zheng
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (J.B.); (W.W.); (Y.-L.Z.); (P.A.F.)
| | - Hancai Dan
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland, Baltimore, MD 21201, USA; (F.H.); (H.D.); (S.S.); (K.C.)
| | - Scott Strome
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland, Baltimore, MD 21201, USA; (F.H.); (H.D.); (S.S.); (K.C.)
| | - Kevin Cullen
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland, Baltimore, MD 21201, USA; (F.H.); (H.D.); (S.S.); (K.C.)
| | - Bruce Davidson
- Department of Otorhinolaryngology-Head and Neck Surgery, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - John F. Deeken
- Inova Translational Medicine Institute, Inova Health System, Fairfax, VA 22031, USA;
| | - Sujata Choudhury
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Peter H. Ahn
- Department of Radiation Medicine, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - Seema Agarwal
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Xuexun Zhou
- iCryobiol and iFuture Technologies, Shanghai 200127, China;
| | - Richard Schlegel
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Priscilla A. Furth
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (J.B.); (W.W.); (Y.-L.Z.); (P.A.F.)
| | - Chong-Xian Pan
- University of California at Davis, Sacramento, CA 95817, USA;
| | - Xuefeng Liu
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (J.B.); (W.W.); (Y.-L.Z.); (P.A.F.)
| |
Collapse
|