1
|
Huang Y, Ma C, Zhu L, Kong L, Huang C, Yang W, He J, Yang M, Huang L, Yuan L, Yi J. The Ameliorative Effect of Betulinic Acid on Oxidative Stress in Mice of Cyclophosphamide-Induced Liver Damage. ENVIRONMENTAL TOXICOLOGY 2025; 40:608-623. [PMID: 39601349 DOI: 10.1002/tox.24444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 10/02/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
As a conventional immunosuppressive drug, cyclophosphamide (CYP) exhibits strong hepatotoxicity in clinical applications. Betulinic acid (BA) is a natural triterpenoid that protects against liver damage. However, the underlying mechanism has not yet been elucidated. The purpose of this study was to evaluate the ameliorative effects of BA on CYP-induced hepatotoxicity and further clarify the underlying mechanism. BA pretreatment mitigated CYP-induced liver oxidative damage by alleviating histopathological lesions, reducing reactive oxygen species (ROS) accumulation, and restoring the mRNA expression of antioxidant enzymes (Cu-Sod, Mn-Sod, Cat, and Gsh-Px). BA treatment also suppressed CYP-induced oxidative stress by activating the NRF2 pathway and inhibiting the MAPK signaling pathway. Moreover, BA attenuated CYP-triggered hepatic apoptosis by suppressing excessive mitochondrial fission, boosting mitochondrial fusion, and ameliorating pro-apoptotic protein expression (CASP9 and the ratio of BCL-2/BAX) by blocking the oxidative stress-activated mitochondrial apoptotic pathway. Furthermore, PD98059 (an inhibitor of ERK) and/or BA abated CYP-provoked hepatotoxicity by inhibiting the ERK-MAPK and mitochondrial apoptotic pathways, implying that deactivation of the ERK-mediated mitochondrial apoptotic pathway contributed to the hepatoprotective efficacy of BA against CYP-induced oxidative stress. Therefore, BA could be used as a complementary medicine in patients undergoing CYP treatment owing to its hepatoprotective effects.
Collapse
Affiliation(s)
- You Huang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Chaoyang Ma
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Lijuan Zhu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Li Kong
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Chunlin Huang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Wenjiang Yang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Jiayu He
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Mingqi Yang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Lin Huang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Liyun Yuan
- College of Agronomy, Xiangyang Polytechnic, Xiangyang, China
| | - Jine Yi
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| |
Collapse
|
2
|
Si Y, He M, Li Y, Jiang J, Fan Y, Xue S, Qiu X, Xie M. On-demand treatment of metabolic diseases by a synthetic drug-inducible exocytosis system. Nat Commun 2025; 16:2838. [PMID: 40121196 PMCID: PMC11929842 DOI: 10.1038/s41467-025-58184-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 03/14/2025] [Indexed: 03/25/2025] Open
Abstract
Here, we present StimExo as a rational design strategy allowing various user-defined control signals to trigger calcium-dependent exocytosis and mediate on-demand protein secretion in cell-therapy settings. Using a modular framework incorporating inducible protein-protein interactions into an engineered bipartite activator of calcium release-activated calcium (CRAC) channels, Ca2+ influx mediated by the STIM/Orai1 machinery was flexibly adjusted to depend on different user-defined input signals. Application of StimExo to various endocrine cells enables instant secretion of therapeutic hormones upon administration of safe and patient-compliant trigger compounds. StimExo also mediated insulin exocytosis using a cell-based gene delivery strategy in vivo, accounting for real-time control of blood glucose homeostasis in male diabetic mice in response to the FDA-approved drug grazoprevir. This study achieves true "sense-and-respond" cell-based therapies and provides a platform for remote control of in vivo transgene activities using various trigger signals of interest.
Collapse
Affiliation(s)
- Yaqing Si
- School of Basic Medical Sciences, Fudan University, Shanghai, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Minghui He
- School of Basic Medical Sciences, Fudan University, Shanghai, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Yilin Li
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jian Jiang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- School of Life Sciences, Fudan University, Shanghai, China
| | - Yuxuan Fan
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shuai Xue
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Xinyuan Qiu
- Department of Biology and Chemistry, College of Science, National University of Defense Technology, Changsha, Hunan, China
- College of Computer Science and Technology, National University of Defense Technology, Changsha, Hunan, China
| | - Mingqi Xie
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
- School of Medicine, Westlake University, Hangzhou, Zhejiang, China.
- School of Engineering, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Kulsoom, Ali W, Wang F. Advancement in synthetic gene circuits engineering: An alternative strategy for microRNA imaging and disease theranostics. Biotechnol Adv 2025; 79:108518. [PMID: 39798857 DOI: 10.1016/j.biotechadv.2025.108518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/26/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
Gene circuits, which are genetically engineered systems designed to regulate gene expression, are emerging as powerful tools in disease theranostics, especially in mammalian cells. This review explores the latest advances in the design and application of gene circuits for detecting and treating various diseases. Synthetic gene circuits, inspired by electronic systems, offer precise control over therapeutic gene activity, allowing for real-time, user-defined responses to pathological signals. Notable applications include synZiFTRs for T-cell-based cancer therapies, immunomagnetic circuits for combating antibiotic-resistant infections like MRSA, and caffeine-induced circuits for managing type-2 diabetes. Additionally, advanced designs such as TetR-Elk1 circuits for reversing insulin resistance, RNAi circuits for targeting cancer cells, and synthetic circuits for managing metabolic conditions like urate homeostasis and diet-induced obesity are highlighted. These gene circuits, tailored for mammalian cells, showcase immense potential in gene- and cell-based therapies for complex metabolic and immune-related disorders, paving the way for precise, customizable treatments. The review focuses on the use of these circuits in mammalian systems and emphasizes their therapeutic implications, offering insights into future developments in disease treatment.
Collapse
Affiliation(s)
- Kulsoom
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China; Institute of Medical Engineering, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an 710061, China
| | - Wajahat Ali
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China; Institute of Medical Engineering, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an 710061, China
| | - Fu Wang
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China; Institute of Medical Engineering, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
4
|
Xiao L, Zhao M, Linghu KG, Wu G, Zhang T, Chen C, Guan J, Cao Z, Hu Y, Yu H. Ganweikang extract protects hepatocytes from oxidative injury by activating Nrf2/HO-1 and MAPKs pathways. Fitoterapia 2024; 178:106146. [PMID: 39089591 DOI: 10.1016/j.fitote.2024.106146] [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: 02/13/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Ganweikang tablet (GWK) is a traditional Chinese prescription and has been clinically used in treating liver diseases for decades. Although GWK has been shown to exert potential therapeutic effect for hepatotoxicity protection, the underlying biological mechanisms are still not well clarified. In the present study, the compositional analysis of GWK was performed by HPLC analysis, and the hepato-protective effects of GWK were assessed in H2O2-stimulated acute oxidative injured HL-7702 hepatocytes in vitro. As a result, 7 components in GWK were quantified to be 0.06 ± 0.01% (calycosin), 0.46 ± 0.02% (calycosin-7-glucoside), 0.13 ± 0.01% (liquiritin), 0.17 ± 0.02% (glycyrrhizic acid), 0.45 ± 0.02% (forsythoside A), 0.07 ± 0.01% (5-O-methylvisammioside) and 0.45 ± 0.02% (forsythin), respectively. Furthermore, GWK (100, 200 and 400 μg/mL, 24 h) dose-dependently alleviated HL-7702 hepatocytes from H2O2 (200 μM, 2 h)-induced cell apoptosis by decreasing the intracellular reactive oxygen species (ROS) generation and malondialdehyde (MDA) level, as well as the cellular aminotransferases (ALT and AST) activities. GWK increased the expressions of HO-1, NQO1 and Nrf2, while suppressing the expression of KEAP1 in H2O2-stimulated HL-7702 cells. A specific Nrf2 inhibitor, ML385, was further employed to investigate the regulation of Nrf2 in HL-7702 cells stimulated by H2O2. In addition, the activation of MAPKs (JUN, ERK and p38) was simultaneously detected in H2O2-stimulated HL-7702 cells. In conclusion, GWK exerted potential therapeutic effect to protect hepatocytes from acute oxidative injury through activating the Nrf2/HO-1 and MAPKs pathways.
Collapse
Affiliation(s)
- Linxuan Xiao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Mingming Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Ke-Gang Linghu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Guoping Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Tian Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Chengyu Chen
- Jiaheng Pharmaceutical Technology Co., Ltd, Zhuhai 519000, China
| | - Jianli Guan
- Henan Fusen Pharmaceutical Co., Ltd, Nanyang 473000, China
| | - Zhiming Cao
- Jiaheng Pharmaceutical Technology Co., Ltd, Zhuhai 519000, China; Henan Fusen Pharmaceutical Co., Ltd, Nanyang 473000, China
| | - Yuanjia Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Hua Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China.
| |
Collapse
|
5
|
Yan Y, Ye X, Huang C, Wu J, Liu Y, Zheng P, Shen C, Bai Z, Tingming S. Anoectochilus roxburghii polysaccharide reduces D-GalN/LPS-induced acute liver injury by regulating the activation of multiple inflammasomes. J Pharm Pharmacol 2024; 76:1212-1224. [PMID: 38985664 DOI: 10.1093/jpp/rgae077] [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: 12/05/2023] [Accepted: 06/02/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Acute liver injury (ALI) is a serious syndrome with a high mortality rate due to viral infection, toxic exposure, and autoimmunity, and its severity can range from mildly elevated liver enzymes to severe liver failure. Activation of the nod-like receptor pyrin domain-containing 3 (NLRP3) inflammasome is closely associated with the development of ALI, and the search for an inhibitor targeting this pathway may be a novel therapeutic option. Anoectochilus roxburghii polysaccharide (ARP) is a biologically active ingredient extracted from Anoectochilus roxburghii with immunomodulatory, antioxidant, and anti-inflammatory bioactivities and pharmacological effects. In this study, we focused on D-galactosamine (D-GalN)/lipopolysaccharide (LPS)-induced acute liver injury by ARP through inhibition of NLRP3 inflammasome. METHODS An inflammasome activation model was established in bone marrow-derived macrophages (BMDMs) to investigate the effects of ARP on caspase-1 cleavage, IL-1β secretion, and ASC oligomerization in inflammasomes under different agonists. We used the D-GalN/LPS-induced acute liver injury model in mice, intraperitoneally injected ARP or MCC950, and collected liver tissues, serum, and intraperitoneal lavage fluid for pathological and biochemical indexes. RESULTS ARP effectively inhibited the activation of the NLRP3 inflammasome and had an inhibitory effect on non-classical NLRP3, AIM2, and NLRC4 inflammasomes. It also effectively inhibited the oligomerization of apoptosis-associated speck-like protein (ASC) from a variety of inflammatory vesicles. Meanwhile, ARP has good therapeutic effects on acute liver injury induced by D-GaIN/LPS. CONCLUSION The inhibitory effect of ARP on a wide range of inflammasomes, as well as its excellent protection against acute liver injury, suggests that ARP may be a candidate for acute liver injury.
Collapse
Affiliation(s)
- Yulu Yan
- Ningde Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fujian, 352100, China
| | - Xiqi Ye
- Ningde Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fujian, 352100, China
| | - Chunqing Huang
- Ningde Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fujian, 352100, China
| | - Junjun Wu
- Ningde Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fujian, 352100, China
| | - Yunbiao Liu
- Pingnan County Hospital of Traditional Chinese Medicine, Ningde City, Fujian Province, 352300, China
| | - Pingping Zheng
- Shouning County Hospital of Traditional Chinese Medicine, Ningde City, Fujian Province, 355500, China
| | - Congqi Shen
- Shanxi University of Traditional Chinese Medicine, 030619,China
| | - Zhaofang Bai
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Shen Tingming
- Ningde Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fujian, 352100, China
| |
Collapse
|
6
|
吴 广, 宋 添, 唐 浪, 王 一, 刘 绪, 黄 胜. [Total saponins of Panax japonicus alleviates CCl 4-induced acute liver injury in rats by regulating the PI3K/AktNF-κB signaling pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:244-251. [PMID: 38501409 PMCID: PMC10954515 DOI: 10.12122/j.issn.1673-4254.2024.02.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Indexed: 03/20/2024]
Abstract
OBJECTIVE To investigate the protective effect of total saponins of Panax japonicus (TSPJ) against CCl4-induced acute liver injury (ALI) in rats and explore the underlying pharmacological mechanisms. METHODS Male SD rat models of CCl4-induced ALI were given intraperitoneal injections of distilled water, 100 mg/kg biphenyl bisabololol, or 50, 100, and 200 mg/kg TSPJ during modeling (n=8). Liver functions (AST, ALT, TBil and ALP) of the rats were assessed and liver pathologies were observed with HE staining. Immunohistochemistry was used to detect the expressions of PI3K/Akt/NF-κB signaling pathway molecules in liver tissue; ELISA was used to determine the levels of T-SOD, GSH-Px, and MDA. Western blotting was performed to detect the expression levels of PI3K-Akt and SIRT6-NF-κB pathways in the liver tissue. RESULTS Network pharmacological analysis indicated that the key pathways including PI3K/Akt mediated the therapeutic effect of TSPJ on ALI. In the rat models of ALI, treatments with biphenyl bisabololol and TSPJ significantly ameliorated CCl4-induced increase of serum levels AST, ALT, ALP, TBil and MDA and decrease of T-SOD and GSH-Px levels (all P < 0.01). The rat models of ALI showed significantly increased expression of p-NF-κB (P < 0.01), decreased expressions of PI3K, p-Akt and SIRT6 proteins, and elevated expression levels of p-NF-κB, TNF-α and IL-6 proteins in the liver, which were all significantly improved in the treatment groups (P < 0.05 or 0.01). CONCLUSION TSPJ can effectively alleviate CCl4-induced ALI in rats by suppressing inflammatory responses and oxidative stress in the liver via regulating the PI3K/Akt and SIRT6/NF-κB pathways.
Collapse
Affiliation(s)
- 广阳 吴
- 湖北民族大学医学部,湖北 恩施 445000Department of Medicine, Hubei Minzu University, Enshi 445000, China
- 湖北恩施学院,湖北 恩施 445000Hubei Enshi College, Enshi 445000, China
| | - 添力 宋
- 湖北民族大学医学部,湖北 恩施 445000Department of Medicine, Hubei Minzu University, Enshi 445000, China
- 湖北民族大学风湿性疾病发生与干预实验室,湖北 恩施 445000Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic diseases, Hubei Minzu University, Enshi 445000, China
| | - 浪 唐
- 湖北民族大学医学部,湖北 恩施 445000Department of Medicine, Hubei Minzu University, Enshi 445000, China
| | - 一民 王
- 湖北民族大学医学部,湖北 恩施 445000Department of Medicine, Hubei Minzu University, Enshi 445000, China
| | - 绪 刘
- 湖北民族大学医学部,湖北 恩施 445000Department of Medicine, Hubei Minzu University, Enshi 445000, China
| | - 胜 黄
- 湖北民族大学医学部,湖北 恩施 445000Department of Medicine, Hubei Minzu University, Enshi 445000, China
- 湖北民族大学风湿性疾病发生与干预实验室,湖北 恩施 445000Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic diseases, Hubei Minzu University, Enshi 445000, China
| |
Collapse
|
7
|
Teixeira AP, Fussenegger M. Synthetic Gene Circuits for Regulation of Next-Generation Cell-Based Therapeutics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309088. [PMID: 38126677 PMCID: PMC10885662 DOI: 10.1002/advs.202309088] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Indexed: 12/23/2023]
Abstract
Arming human cells with synthetic gene circuits enables to expand their capacity to execute superior sensing and response actions, offering tremendous potential for innovative cellular therapeutics. This can be achieved by assembling components from an ever-expanding molecular toolkit, incorporating switches based on transcriptional, translational, or post-translational control mechanisms. This review provides examples from the three classes of switches, and discusses their advantages and limitations to regulate the activity of therapeutic cells in vivo. Genetic switches designed to recognize internal disease-associated signals often encode intricate actuation programs that orchestrate a reduction in the sensed signal, establishing a closed-loop architecture. Conversely, switches engineered to detect external molecular or physical cues operate in an open-loop fashion, switching on or off upon signal exposure. The integration of such synthetic gene circuits into the next generation of chimeric antigen receptor T-cells is already enabling precise calibration of immune responses in terms of magnitude and timing, thereby improving the potency and safety of therapeutic cells. Furthermore, pre-clinical engineered cells targeting other chronic diseases are gathering increasing attention, and this review discusses the path forward for achieving clinical success. With synthetic biology at the forefront, cellular therapeutics holds great promise for groundbreaking treatments.
Collapse
Affiliation(s)
- Ana P. Teixeira
- Department of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
- Faculty of ScienceUniversity of BaselKlingelbergstrasse 48BaselCH‐4056Switzerland
| |
Collapse
|
8
|
Zhang ZJ, Ding LY, Zuo XL, Feng H, Xia Q. A new paradigm in transplant immunology: At the crossroad of synthetic biology and biomaterials. MED 2023:S2666-6340(23)00142-3. [PMID: 37244257 DOI: 10.1016/j.medj.2023.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 02/04/2023] [Accepted: 05/02/2023] [Indexed: 05/29/2023]
Abstract
Solid organ transplant (SOT) recipients require meticulously tailored immunosuppressive regimens to minimize graft loss and mortality. Traditional approaches focus on inhibiting effector T cells, while the intricate and dynamic immune responses mediated by other components remain unsolved. Emerging advances in synthetic biology and material science have provided novel treatment modalities with increased diversity and precision to the transplantation community. This review investigates the active interface between these two fields, highlights how living and non-living structures can be engineered and integrated for immunomodulation, and discusses their potential application in addressing the challenges in SOT clinical practice.
Collapse
Affiliation(s)
- Zi-Jie Zhang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; Shanghai Engineering Research Centre of Transplantation and Immunology, Shanghai 200127, China
| | - Lu-Yue Ding
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiao-Lei Zuo
- Shanghai Engineering Research Centre of Transplantation and Immunology, Shanghai 200127, China; School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao Feng
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; Shanghai Engineering Research Centre of Transplantation and Immunology, Shanghai 200127, China; Shanghai Institute of Transplantation, Shanghai 200127, China; Punan Branch (Shanghai Punan Hospital), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; Shanghai Engineering Research Centre of Transplantation and Immunology, Shanghai 200127, China; Shanghai Institute of Transplantation, Shanghai 200127, China.
| |
Collapse
|
9
|
Lu Q, Wang Z, Bai S, Wang Y, Liao C, Sun Y, Zhang Y, Li W, Mei Q. Hydrophobicity Regulation of Energy Acceptors Confined in Mesoporous Silica Enabled Reversible Activation of Optogenetics for Closed-Loop Glycemic Control. J Am Chem Soc 2023; 145:5941-5951. [PMID: 36867047 DOI: 10.1021/jacs.2c13762] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Optogenetics-based synthetic biology holds great promise as a cell-based therapy strategy for many clinical incurable diseases; however, precise control over genetic expression strength and timing through disease state-related closed-loop regulation remains a challenge due to the lack of reversible probes to indicate real-time metabolite fluctuations. Here, based on a novel mechanism of analyte-induced hydrophobicity regulation of energy acceptors confined in mesoporous silica, we developed a smart hydrogel platform comprising glucose reversible responsive upconversion nanoprobes and optogenetic engineered cells, in which the upconverted blue light strength was adaptively tuned through blood glucose levels to control optogenetic expressions for insulin secretion. The intelligent hydrogel system enabled convenient maintenance of glycemic homeostasis through simple near-infrared illuminations without any additional glucose concentration monitoring, which efficiently avoided genetic overexpression-induced hypoglycemia. This proof-of-concept strategy efficiently combines diagnostics with optogenetics-based synthetic biology for mellitus therapy, opening up a new avenue for nano-optogenetics.
Collapse
Affiliation(s)
- Qi Lu
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Zihe Wang
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Shumin Bai
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
| | - Ying Wang
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Cheng Liao
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Yaru Sun
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Yi Zhang
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Wei Li
- Department of Neurosurgery, The Sixth Affiliated Hospital, Jinan University, Dongguan, Guangdong 523560, China
| | - Qingsong Mei
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China.,Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| |
Collapse
|
10
|
Stefanov BA, Fussenegger M. Biomarker-driven feedback control of synthetic biology systems for next-generation personalized medicine. Front Bioeng Biotechnol 2022; 10:986210. [PMID: 36225597 PMCID: PMC9548536 DOI: 10.3389/fbioe.2022.986210] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Many current clinical therapies for chronic diseases involve administration of drugs using dosage and bioavailability parameters estimated for a generalized population. This standard approach carries the risk of under dosing, which may result in ineffective treatment, or overdosing, which may cause undesirable side effects. Consequently, maintaining a drug concentration in the therapeutic window often requires frequent monitoring, adversely affecting the patient’s quality of life. In contrast, endogenous biosystems have evolved finely tuned feedback control loops that govern the physiological functions of the body based on multiple input parameters. To provide personalized treatment for chronic diseases, therefore, we require synthetic systems that can similarly generate a calibrated therapeutic response. Such engineered autonomous closed-loop devices should incorporate a sensor that actively tracks and evaluates the disease severity based on one or more biomarkers, as well as components that utilize these molecular inputs to bio compute and deliver the appropriate level of therapeutic output. Here, we review recent advances in applications of the closed-loop design principle in biomedical implants for treating severe and chronic diseases, highlighting translational studies of cellular therapies. We describe the engineering principles and components of closed-loop therapeutic devices, and discuss their potential to become a key pillar of personalized medicine.
Collapse
Affiliation(s)
| | - Martin Fussenegger
- ETH Zürich, Department of Biosystems Science and Engineering, Basel, Switzerland
- Faculty of Life Science, University of Basel, Basel, Switzerland
- *Correspondence: Martin Fussenegger,
| |
Collapse
|
11
|
Mansouri M, Fussenegger M. Therapeutic cell engineering: designing programmable synthetic genetic circuits in mammalian cells. Protein Cell 2022; 13:476-489. [PMID: 34586617 PMCID: PMC9226217 DOI: 10.1007/s13238-021-00876-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/02/2021] [Indexed: 12/01/2022] Open
Abstract
Cell therapy approaches that employ engineered mammalian cells for on-demand production of therapeutic agents in the patient's body are moving beyond proof-of-concept in translational medicine. The therapeutic cells can be customized to sense user-defined signals, process them, and respond in a programmable and predictable way. In this paper, we introduce the available tools and strategies employed to design therapeutic cells. Then, various approaches to control cell behaviors, including open-loop and closed-loop systems, are discussed. We also highlight therapeutic applications of engineered cells for early diagnosis and treatment of various diseases in the clinic and in experimental disease models. Finally, we consider emerging technologies such as digital devices and their potential for incorporation into future cell-based therapies.
Collapse
Affiliation(s)
- Maysam Mansouri
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland.
- Faculty of Science, University of Basel, Mattenstrasse 26, 4058, Basel, Switzerland.
| |
Collapse
|
12
|
Zhao H, Xue S, Hussherr MD, Teixeira AP, Fussenegger M. Autonomous push button-controlled rapid insulin release from a piezoelectrically activated subcutaneous cell implant. SCIENCE ADVANCES 2022; 8:eabm4389. [PMID: 35704573 PMCID: PMC9200281 DOI: 10.1126/sciadv.abm4389] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 04/28/2022] [Indexed: 05/25/2023]
Abstract
Traceless physical cues are desirable for remote control of the in situ production and real-time dosing of biopharmaceuticals in cell-based therapies. However, current optogenetic, magnetogenetic, or electrogenetic devices require sophisticated electronics, complex artificial intelligence-assisted software, and external energy supplies for power and control. Here, we describe a self-sufficient subcutaneous push button-controlled cellular implant powered simply by repeated gentle finger pressure exerted on the overlying skin. Pushing the button causes transient percutaneous deformation of the implant's embedded piezoelectric membrane, which produces sufficient low-voltage energy inside a semi-permeable platinum-coated cell chamber to mediate rapid release of a biopharmaceutical from engineered electro-sensitive human cells. Release is fine-tuned by varying the frequency and duration of finger-pressing stimulation. As proof of concept, we show that finger-pressure activation of the subcutaneous implant can restore normoglycemia in a mouse model of type 1 diabetes. Self-sufficient push-button devices may provide a new level of convenience for patients to control their cell-based therapies.
Collapse
Affiliation(s)
- Haijie Zhao
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Shuai Xue
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Marie-Didiée Hussherr
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Ana Palma Teixeira
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
- Faculty of Science, University of Basel, Mattenstrasse 26, CH-4058 Basel, Switzerland
| |
Collapse
|
13
|
Fu R, Zu SJ, Liu YJ, Li JC, Dang WZ, Liao LP, Liu LP, Chen PY, Huang HM, Wu KH, Zhou B, Pan Q, Luo C, Zhang YY, Li GM. Selective bromodomain and extra-terminal bromodomain inhibitor inactivates macrophages and hepatic stellate cells to inhibit liver inflammation and fibrosis. Bioengineered 2022; 13:10914-10930. [PMID: 35499161 PMCID: PMC9278415 DOI: 10.1080/21655979.2022.2066756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Liver fibrosis occurs following inflammation triggered by the integrated actions of activated liver-resident macrophages (Kupffer cells) and hepatic stellate cells (HSCs), and the multiplicity of these mechanisms complicates drug therapy. Here, we demonstrate that the selective bromodomain and extra-terminal (BET) bromodomain inhibitor compound38 can block both the Janus kinase-signal transducer and activator of transcription and mitogen-activated protein kinase signaling pathways in macrophages, which decreased their secretion of proinflammatory cytokines in a dose-dependent manner. The inactivation of macrophages attenuated lipopolysaccharide-induced injurious inflammation concurrent with a reduction in F4/80+ cells, proinflammatory cytokine levels, and neutrophil infiltration. Moreover, compound 38 inhibited the Wnt/β-catenin and transforming growth factor-beta/SMAD signaling pathways to abolish the activation of HSCs. In vivo, compound 38 significantly decreased the collagen deposition and fibrotic area of a CCl4-induced liver fibrosis model, and restored the deficiency of activated HSCs and the upregulation of liver inflammation. These results highlight the potential role of compound 38 in treating liver fibrosis considering its simultaneous inhibitory effects on liver inflammation and related fibrosis.
Collapse
Affiliation(s)
- Rong Fu
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, Yangpu District, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, Zuchongzhi District, Shanghai, China
| | - Shi-Jia Zu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, Zuchongzhi District, Shanghai, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, China
| | - Yan-Jun Liu
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, Yangpu District, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, Zuchongzhi District, Shanghai, China
| | - Jia-Cheng Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, Zuchongzhi District, Shanghai, China
| | - Wen-Zhen Dang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, Zuchongzhi District, Shanghai, China
| | - Li-Ping Liao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, Zuchongzhi District, Shanghai, China
| | - Li-Ping Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, Zuchongzhi District, Shanghai, China
| | - Pan-Yu Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, Zuchongzhi District, Shanghai, China
| | - He-Ming Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, Zuchongzhi District, Shanghai, China
| | - Kang-Hui Wu
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, Yangpu District, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, Zuchongzhi District, Shanghai, China
| | - Bing Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, Zuchongzhi District, Shanghai, China
| | - Qin Pan
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, Yangpu District, China
- Research center, Zhoupu Hospital affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, Zhouyuan District, China
| | - Cheng Luo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, Zuchongzhi District, Shanghai, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of SciencesCAS, Hangzhou, Zhejiang, China
| | - Yuan-Yuan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, Zuchongzhi District, Shanghai, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of SciencesCAS, Hangzhou, Zhejiang, China
| | - Guang-Ming Li
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, Yangpu District, China
| |
Collapse
|
14
|
Li M, Luo Q, Tao Y, Sun X, Liu C. Pharmacotherapies for Drug-Induced Liver Injury: A Current Literature Review. Front Pharmacol 2022; 12:806249. [PMID: 35069218 PMCID: PMC8766857 DOI: 10.3389/fphar.2021.806249] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/16/2021] [Indexed: 12/14/2022] Open
Abstract
Drug-induced liver injury (DILI) has become a serious public health problem. For the management of DILI, discontinuation of suspicious drug or medicine is the first step, but the treatments including drugs and supporting approaches are needed. Reference to clinical patterns and disease severity grades of DILI, the treatment drugs were considered to summarize into hepatoprotective drugs (N-acetylcysteine and Glutathione, Glycyrrhizin acid preparation, Polyene phosphatidylcholine, Bicyclol, Silymarin), anticholestatic drug (Ursodeoxycholic acid, S-adenosylmethionine, Cholestyramine), immunosuppressants (Glucocorticoids) and specific treatment agents (L-carnitine, Anticoagulants). The current article reviewed the accumulated literature with evidence-based medicine researches for DILI in clinical practice. Also the drawbacks of the clinical studies involved in the article, unmet needs and prospective development for DILI therapy were discussed.
Collapse
Affiliation(s)
- Meng Li
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiong Luo
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanyan Tao
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Sun
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Chenghai Liu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai, China
- Shanghai Innovation Center of TCM Health Service, Shanghai, China
| |
Collapse
|
15
|
Abstract
The steadfast advance of the synthetic biology field has enabled scientists to use genetically engineered cells, instead of small molecules or biologics, as the basis for the development of novel therapeutics. Cells endowed with synthetic gene circuits can control the localization, timing and dosage of therapeutic activities in response to specific disease biomarkers and thus represent a powerful new weapon in the fight against disease. Here, we conceptualize how synthetic biology approaches can be applied to programme living cells with therapeutic functions and discuss the advantages that they offer over conventional therapies in terms of flexibility, specificity and predictability, as well as challenges for their development. We present notable advances in the creation of engineered cells that harbour synthetic gene circuits capable of biological sensing and computation of signals derived from intracellular or extracellular biomarkers. We categorize and describe these developments based on the cell scaffold (human or microbial) and the site at which the engineered cell exerts its therapeutic function within its human host. The design of cell-based therapeutics with synthetic biology is a rapidly growing strategy in medicine that holds great promise for the development of effective treatments for a wide variety of human diseases.
Collapse
|
16
|
Wu P, Xi X, Li R, Sun G. Engineering Polysaccharides for Tissue Repair and Regeneration. Macromol Biosci 2021; 21:e2100141. [PMID: 34219388 DOI: 10.1002/mabi.202100141] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/11/2021] [Indexed: 12/22/2022]
Abstract
The success of repair or regeneration depends greatly on the architecture of 3D scaffolds that finely mimic natural extracellular matrix to support cell growth and assembly. Polysaccharides have excellent biocompatibility with intrinsic biological cues and they have been extensively investigated as scaffolds for tissue engineering and regenerative medicine (TERM). The physical and biochemical structures of natural polysaccharides, however, can barely meet all the requirements of tissue-engineered scaffolds. To take advantage of their inherent properties, many innovative approaches including chemical, physical, or joint modifications have been employed to improve their properties. Recent advancement in molecular and material building technology facilitates the fabrication of advanced 3D structures with desirable properties. This review focuses on the latest progress of polysaccharide-based scaffolds for TERM, especially those that construct advanced architectures for tissue regeneration.
Collapse
Affiliation(s)
- Pingli Wu
- College of Chemistry and Environmental Science, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Xin Xi
- Affiliated Hospital of Hebei University, College of Clinical Medicine, Institute of Life Science and Green Development, Hebei University, Baoding, 071000, China
| | - Ruochen Li
- College of Chemistry and Environmental Science, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Guoming Sun
- College of Chemistry and Environmental Science, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China.,Affiliated Hospital of Hebei University, College of Clinical Medicine, Institute of Life Science and Green Development, Hebei University, Baoding, 071000, China
| |
Collapse
|
17
|
Wagner HJ, Mohsenin H, Weber W. Synthetic Biology-Empowered Hydrogels for Medical Diagnostics. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2021; 178:197-226. [PMID: 33582837 DOI: 10.1007/10_2020_158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Synthetic biology is strongly inspired by concepts of engineering science and aims at the design and generation of artificial biological systems in different fields of research such as diagnostics, analytics, biomedicine, or chemistry. To this aim, synthetic biology uses an engineering approach relying on a toolbox of molecular sensors and switches that endows cellular hosts with non-natural computing functions and circuits. Importantly, this concept is not only limited to cellular approaches. Synthetic biological building blocks have also conferred sensing and switching capability to otherwise inactive materials. This principle has attracted high interest for the development of biohybrid materials capable of sensing and responding to specific molecular stimuli, such as disease biomarkers, antibiotics, or heavy metals. Moreover, the interconnection of individual sense-and-respond materials to complex materials systems has enabled the processing of, for example, multiple inputs or the amplification of signals using feedback topologies. Such systems holding high potential for applications in the analytical and diagnostic sectors will be described in this chapter.
Collapse
Affiliation(s)
- Hanna J Wagner
- Faculty of Biology, Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg im Breisgau, Germany.,Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Hasti Mohsenin
- Faculty of Biology, Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg im Breisgau, Germany
| | - Wilfried Weber
- Faculty of Biology, Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg im Breisgau, Germany.
| |
Collapse
|
18
|
Li J, Zhao H, Zheng L, An W. Advances in Synthetic Biology and Biosafety Governance. Front Bioeng Biotechnol 2021; 9:598087. [PMID: 33996776 PMCID: PMC8120004 DOI: 10.3389/fbioe.2021.598087] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 02/17/2021] [Indexed: 11/22/2022] Open
Abstract
Tremendous advances in the field of synthetic biology have been witnessed in multiple areas including life sciences, industrial development, and environmental bio-remediation. However, due to the limitations of human understanding in the code of life, any possible intended or unintended uses of synthetic biology, and other unknown reasons, the development and application of this technology has raised concerns over biosafety, biosecurity, and even cyberbiosecurity that they may expose public health and the environment to unknown hazards. Over the past decades, some countries in Europe, America, and Asia have enacted laws and regulations to control the application of synthetic biology techniques in basic and applied research and this has resulted in some benefits. The outbreak of the COVID-19 caused by novel coronavirus SARS-CoV-2 and various speculations about the origin of this virus have attracted more attention on bio-risk concerns of synthetic biology because of its potential power and uncertainty in the synthesis and engineering of living organisms. Therefore, it is crucial to scrutinize the control measures put in place to ensure appropriate use, promote the development of synthetic biology, and strengthen the governance of pathogen-related research, although the true origin of coronavirus remains hotly debated and unresolved. This article reviews the recent progress made in the field of synthetic biology and combs laws and regulations in governing bio-risk issues. We emphasize the urgent need for legislative and regulatory constraints and oversight to address the biological risks of synthetic biology.
Collapse
Affiliation(s)
- Jing Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Huimiao Zhao
- College of Humanities and Law, Beijing University of Chemical Technology, Beijing, China
| | - Lanxin Zheng
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Wenlin An
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
19
|
Rondon R, Wilson CJ. Engineering Alternate Ligand Recognition in the PurR Topology: A System of Novel Caffeine Biosensing Transcriptional Antirepressors. ACS Synth Biol 2021; 10:552-565. [PMID: 33689294 DOI: 10.1021/acssynbio.0c00582] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recent advances in synthetic biology and protein engineering have increased the number of allosteric transcription factors used to regulate independent promoters. These developments represent an important increase in our biological computing capacity, which will enable us to construct more sophisticated genetic programs for a broad range of biological technologies. However, the majority of these transcription factors are represented by the repressor phenotype (BUFFER), and require layered inversion to confer the antithetical logical function (NOT), requiring additional biological resources. Moreover, these engineered transcription factors typically utilize native ligand binding functions paired with alternate DNA binding functions. In this study, we have advanced the state-of-the-art by engineering and redesigning the PurR topology (a native antirepressor) to be responsive to caffeine, while mitigating responsiveness to the native ligand hypoxanthine-i.e., a deamination product of the input molecule adenine. Importantly, the resulting caffeine responsive transcription factors are not antagonized by the native ligand hypoxanthine. In addition, we conferred alternate DNA binding to the caffeine antirepressors, and to the PurR scaffold, creating 38 new transcription factors that are congruent with our current transcriptional programming structure. Finally, we leveraged this system of transcription factors to create integrated NOR logic and related feedback operations. This study represents the first example of a system of transcription factors (antirepressors) in which both the ligand binding site and the DNA binding functions were successfully engineered in tandem.
Collapse
Affiliation(s)
- Ronald Rondon
- Georgia Institute of Technology, School of Chemical & Biomolecular Engineering, 311 Ferst Drive, Atlanta, Georgia 30332-0100, United States
| | - Corey J. Wilson
- Georgia Institute of Technology, School of Chemical & Biomolecular Engineering, 311 Ferst Drive, Atlanta, Georgia 30332-0100, United States
| |
Collapse
|
20
|
Wang Y, Tian L, Wang Y, Zhao T, Khan A, Wang Y, Cao J, Cheng G. Protective effect of Que Zui tea hot-water and aqueous ethanol extract against acetaminophen-induced liver injury in mice via inhibition of oxidative stress, inflammation, and apoptosis. Food Funct 2021; 12:2468-2480. [PMID: 33650604 DOI: 10.1039/d0fo02894k] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The tender leaves and buds of Vaccinium dunalianum Wight have been traditionally processed as folk tea, known as Que Zui tea (QT), with a wide range of benefits to humans. In this study, Que Zui tea hot-water extract (QTW) and aqueous-ethanol extract (QTE) were tested for their effectiveness to alleviate acetaminophen (APAP)-induced liver damage. QTW and QTE significantly inhibited the alanine aminotransaminase, aspartate aminotransaminase, tumor necrosis factor-α, interleukin-6, and interleukin-1β levels in the serum. Both extracts also ameliorated pathological damage and inhibited oxidative stress in the liver of APAP-induced mice. In addition, QTW and QTE activated the nuclear erythroid related factor 2 signal pathway, and inhibited mitogen-activated protein kinase activation. QTW and QTE also suppressed hepatocyte apoptosis by improvement of Bcl-2/Bax and inhibition of caspase-3 and caspase-9 expression. The results demonstrated that QTW and QTE could effectively protect APAP hepatotoxicity, which might be attributed to their antioxidant, anti-inflammatory and anti-apoptosis activities.
Collapse
Affiliation(s)
- Yongpeng Wang
- Faculty of Agriculture and Food, Kunming University of Science and Technology, Kunming, 650500, China.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Brooks SM, Alper HS. Applications, challenges, and needs for employing synthetic biology beyond the lab. Nat Commun 2021; 12:1390. [PMID: 33654085 PMCID: PMC7925609 DOI: 10.1038/s41467-021-21740-0] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
Synthetic biology holds great promise for addressing global needs. However, most current developments are not immediately translatable to 'outside-the-lab' scenarios that differ from controlled laboratory settings. Challenges include enabling long-term storage stability as well as operating in resource-limited and off-the-grid scenarios using autonomous function. Here we analyze recent advances in developing synthetic biological platforms for outside-the-lab scenarios with a focus on three major application spaces: bioproduction, biosensing, and closed-loop therapeutic and probiotic delivery. Across the Perspective, we highlight recent advances, areas for further development, possibilities for future applications, and the needs for innovation at the interface of other disciplines.
Collapse
Affiliation(s)
- Sierra M Brooks
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Hal S Alper
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA.
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
22
|
Feng X, Xia K, Ke Q, Deng R, Zhuang J, Wan Z, Luo P, Wang F, Zang Z, Sun X, Xiang AP, Tu X, Gao Y, Deng C. Transplantation of encapsulated human Leydig-like cells: A novel option for the treatment of testosterone deficiency. Mol Cell Endocrinol 2021; 519:111039. [PMID: 32980418 DOI: 10.1016/j.mce.2020.111039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022]
Abstract
Previous studies have demonstrated that the transplantation of alginate-poly-ʟ-lysine-alginate (APA)-encapsulated rat Leydig cells (LCs) provides a promising approach for treating testosterone deficiency (TD). Nevertheless, LCs have a limited capacity to proliferate, limiting the efficacy of LC transplantation therapy. Here, we established an efficient differentiation system to obtain functional Leydig-like cells (LLCs) from human stem Leydig cells (hSLCs). Then we injected APA-encapsulated LLCs into the abdominal cavities of castrated mice without an immunosuppressor. The APA-encapsulated cells survived and partially restored testosterone production for 90 days in vivo. More importantly, the transplantation of encapsulated LLCs ameliorated the symptoms of TD, such as fat accumulation, muscle atrophy and adipocyte accumulation in bone marrow. Overall, these results suggest that the transplantation of encapsulated LLCs is a promising new method for testosterone supplementation with potential clinical applications in TD.
Collapse
Affiliation(s)
- Xin Feng
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Kai Xia
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Qiong Ke
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Rongda Deng
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; KingMed Center for Clinical Laboratory CO., LTD, Guangzhou, China
| | - Jintao Zhuang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zi Wan
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peng Luo
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fulin Wang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhijun Zang
- Department of Infertility and Sexual Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiangzhou Sun
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiang'an Tu
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Yong Gao
- Reproductive Medicine Center, The Key Laboratory for Reproductive Medicine of Guangdong Province, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Chunhua Deng
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
23
|
Drug repurposing using transcriptome sequencing and virtual drug screening in a patient with glioblastoma. Invest New Drugs 2020; 39:670-685. [PMID: 33313992 PMCID: PMC8068653 DOI: 10.1007/s10637-020-01037-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/19/2020] [Indexed: 12/02/2022]
Abstract
Background Precision medicine and drug repurposing are attractive strategies, especially for tumors with worse prognosis. Glioblastoma is a highly malignant brain tumor with limited treatment options and short survival times. We identified novel BRAF (47-438del) and PIK3R1 (G376R) mutations in a glioblastoma patient by RNA-sequencing. Methods The protein expression of BRAF and PIK3R1 as well as the lack of EGFR expression as analyzed by immunohistochemistry corroborated RNA-sequencing data. The expression of additional markers (AKT, SRC, mTOR, NF-κB, Ki-67) emphasized the aggressiveness of the tumor. Then, we screened a chemical library of > 1500 FDA-approved drugs and > 25,000 novel compounds in the ZINC database to find established drugs targeting BRAF47-438del and PIK3R1-G376R mutated proteins. Results Several compounds (including anthracyclines) bound with higher affinities than the control drugs (sorafenib and vemurafenib for BRAF and PI-103 and LY-294,002 for PIK3R1). Subsequent cytotoxicity analyses showed that anthracyclines might be suitable drug candidates. Aclarubicin revealed higher cytotoxicity than both sorafenib and vemurafenib, whereas idarubicin and daunorubicin revealed higher cytotoxicity than LY-294,002. Liposomal formulations of anthracyclines may be suitable to cross the blood brain barrier. Conclusions In conclusion, we identified novel small molecules via a drug repurposing approach that could be effectively used for personalized glioblastoma therapy especially for patients carrying BRAF47-438del and PIK3R1-G376R mutations.
Collapse
|
24
|
Kojima R, Aubel D, Fussenegger M. Building sophisticated sensors of extracellular cues that enable mammalian cells to work as "doctors" in the body. Cell Mol Life Sci 2020; 77:3567-3581. [PMID: 32185403 PMCID: PMC7452942 DOI: 10.1007/s00018-020-03486-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/27/2020] [Accepted: 02/17/2020] [Indexed: 12/24/2022]
Abstract
Mammalian cells are inherently capable of sensing extracellular environmental signals and activating complex biological functions on demand. Advances in synthetic biology have made it possible to install additional capabilities, which can allow cells to sense the presence of custom biological molecules and provide defined outputs on demand. When implanted/infused in patients, such engineered cells can work as intrabody "doctors" that diagnose disease states and produce and deliver therapeutic molecules when and where necessary. The key to construction of such theranostic cells is the development of a range of sensor systems for detecting various extracellular environmental cues that can be rewired to custom outputs. In this review, we introduce the state-of-art engineering principles utilized in the design of sensor systems to detect soluble factors and also to detect specific cell contact, and we discuss their potential role in treating intractable diseases by delivering appropriate therapeutic functions on demand. We also discuss the challenges facing these emerging technologies.
Collapse
Affiliation(s)
- Ryosuke Kojima
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
- PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan.
| | - Dominque Aubel
- IUTA Département Génie Biologique, Université Claude Bernard Lyon 1, Boulevard du 11 Novembre 1918, 69622, Villeurbanne Cedex, France
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland.
- Faculty of Science, University of Basel, Mattenstrasse 26, 4058, Basel, Switzerland.
| |
Collapse
|
25
|
Orive G, Taebnia N, Erezuma I, Andresen TL, Dolatshahi-Pirouz A. Hacking Human Beings with Machine Biology to Increase Lifespan. Trends Biotechnol 2020; 38:1312-1315. [PMID: 32499063 DOI: 10.1016/j.tibtech.2020.05.007] [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: 12/31/2019] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 11/30/2022]
Abstract
Imagine a world where machines can program cells to deliver therapeutics in a remote-controlled, time-specific, and targeted manner. Or, what if physicians could collect data continuously to establish intimate links between therapy and disease progression? Such machine biology systems could empower physicians beyond imagination and give rise to personalized treatments.
Collapse
Affiliation(s)
- Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore.
| | - Nayere Taebnia
- Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs, Denmark
| | - Itsasne Erezuma
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain
| | - Thomas L Andresen
- Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs, Denmark
| | - Alireza Dolatshahi-Pirouz
- Department of Health Technology, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, 2800 Kgs, Denmark; Department of Dentistry - Regenerative Biomaterials, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Philips van Leydenlaan 25, 6525, EX, Nijmegen, The Netherlands.
| |
Collapse
|
26
|
Xie M, Fussenegger M. Designing cell function: assembly of synthetic gene circuits for cell biology applications. Nat Rev Mol Cell Biol 2019; 19:507-525. [PMID: 29858606 DOI: 10.1038/s41580-018-0024-z] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Synthetic biology is the discipline of engineering application-driven biological functionalities that were not evolved by nature. Early breakthroughs of cell engineering, which were based on ectopic (over)expression of single sets of transgenes, have already had a revolutionary impact on the biotechnology industry, regenerative medicine and blood transfusion therapies. Now, we require larger-scale, rationally assembled genetic circuits engineered to programme and control various human cell functions with high spatiotemporal precision in order to solve more complex problems in applied life sciences, biomedicine and environmental sciences. This will open new possibilities for employing synthetic biology to advance personalized medicine by converting cells into living therapeutics to combat hitherto intractable diseases.
Collapse
Affiliation(s)
- Mingqi Xie
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland. .,University of Basel, Faculty of Science, Basel, Switzerland.
| |
Collapse
|
27
|
Liu Y, Charpin-El Hamri G, Ye H, Fussenegger M. A synthetic free fatty acid-regulated transgene switch in mammalian cells and mice. Nucleic Acids Res 2019; 46:9864-9874. [PMID: 30219861 PMCID: PMC6182168 DOI: 10.1093/nar/gky805] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 08/28/2018] [Indexed: 02/06/2023] Open
Abstract
Trigger-inducible transgene expression systems are utilized in biopharmaceutical manufacturing and also to enable controlled release of therapeutic agents in vivo. We considered that free fatty acids (FFAs), which are dietary components, signaling molecules and important biomarkers, would be attractive candidates as triggers for novel transgene switches with many potential applications, e.g. in future gene- and cell-based therapies. To develop such a switch, we rewired the signal pathway of human G-protein coupled receptor 40 to a chimeric promoter triggering gene expression through an increase of intracellular calcium concentration. This synthetic gene switch is responsive to physiologically relevant FFA concentrations in different mammalian cell types grown in culture or in a bioreactor, or implanted into mice. Animal recipients of microencapsulated sensor cells containing this switch exhibited significant transgene induction following consumption of dietary fat (such as Swiss cheese) or under hyperlipidaemic conditions, including obesity, diabetes and lipodystrophy.
Collapse
Affiliation(s)
- Ying Liu
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Ghislaine Charpin-El Hamri
- Département Génie Biologique, Université Claude Bernard 1, 43 Boulevard du 11 Novembre 1918, F-69100 Villeurbanne, France
| | - Haifeng Ye
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Dongchuan Road 500, Shanghai 200241, China
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland.,Faculty of Science, University of Basel, Mattenstrasse 26, CH-4058 Basel, Switzerland
| |
Collapse
|
28
|
A fully human transgene switch to regulate therapeutic protein production by cooling sensation. Nat Med 2019; 25:1266-1273. [PMID: 31285633 DOI: 10.1038/s41591-019-0501-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 05/29/2019] [Indexed: 12/15/2022]
Abstract
The ability to safely control transgene expression with simple synthetic gene switches is critical for effective gene- and cell-based therapies. In the present study, the signaling pathway controlled by human transient receptor potential (TRP) melastatin 8 (hTRPM8), a TRP channel family member1, is harnessed to control transgene expression. Human TRPM8 signaling is stimulated by menthol, an innocuous, natural, cooling compound, or by exposure to a cool environment (15-18 °C). By functionally linking hTRPM8-induced signaling to a synthetic promoter containing elements that bind nuclear factor of activated T cells, a synthetic gene circuit was designed that can be adjusted by exposure to either a cool environment or menthol. It was shown that this gene switch is functional in various cell types and human primary cells, as well as in mice implanted with engineered cells. In response to transdermal delivery of menthol, microencapsulated cell implants harboring this gene circuit, coupled to expression of either of two therapeutic proteins, insulin or a modified, activin type IIB, receptor ligand trap protein (mActRIIBECD-hFc), could alleviate hyperglycemia in alloxan-treated mice (a model of type 1 diabetes) or reverse muscle atrophy in dexamethasone-treated mice (a model of muscle wasting), respectively. This fully human-derived orthogonal transgene switch should be amenable to a wide range of clinical applications.
Collapse
|
29
|
Cella F, Siciliano V. Protein-based parts and devices that respond to intracellular and extracellular signals in mammalian cells. Curr Opin Chem Biol 2019; 52:47-53. [PMID: 31158655 DOI: 10.1016/j.cbpa.2019.04.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 03/14/2019] [Accepted: 04/15/2019] [Indexed: 01/07/2023]
Abstract
Synthetic biology aims to rewire cellular activities and functionality by implementing genetic circuits with high biocomputing capabilities. Recent efforts led to the development of smart sensing interfaces which integrate multiple inputs to activate desired outputs in a highly specific and sensitive manner. In this review, we highlight protein-based interfaces that sense intracellular or extracellular cues providing information about dynamic environmental changes and cellular state. We will also discuss different mechanisms of regulation of gene expression connected to the sensors to develop diagnostic and therapeutic devices. We conclude discussing challenges and opportunities for biomedical applications of synthetic mammalian protein-based devices.
Collapse
Affiliation(s)
- Federica Cella
- Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci, Naples, Italy; University of Genoa, Genoa, Italy
| | - Velia Siciliano
- Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci, Naples, Italy.
| |
Collapse
|
30
|
P Teixeira A, Fussenegger M. Engineering mammalian cells for disease diagnosis and treatment. Curr Opin Biotechnol 2019; 55:87-94. [DOI: 10.1016/j.copbio.2018.08.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/11/2018] [Accepted: 08/17/2018] [Indexed: 12/11/2022]
|
31
|
Lainšček D, Kadunc L, Keber MM, Bratkovič IH, Romih R, Jerala R. Delivery of an Artificial Transcription Regulator dCas9-VPR by Extracellular Vesicles for Therapeutic Gene Activation. ACS Synth Biol 2018; 7:2715-2725. [PMID: 30513193 DOI: 10.1021/acssynbio.8b00192] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The CRISPR/Cas system has been developed as a potent tool for genome engineering and transcription regulation. However, the efficiency of the delivery of the system into cells, particularly for therapeutic in vivo applications, remains a major bottleneck. Extracellular vesicles (EVs), released by eukaryotic cells, can mediate the transfer of various molecules, including nucleic acids and proteins. We show the packaging and delivery of the CRISPR/Cas system via EVs to the target cells, combining the advantages of both technological platforms. A genome editing with designed extracellular vesicles (GEDEX) system generated by the producer cells can transfer the designed transcriptional regulator dCas9-VPR complexed with appropriate targeting gRNAs enabling activation of gene transcription. We show functional delivery in mammalian cells as well in the animals. The therapeutic efficiency of in vivo delivery of dCas9-VPR/sgRNA GEDEX is demonstrated in a mouse model of liver damage counteracted by upregulation of the endogenous hepatocyte growth factor, demonstrating the potential for therapeutic applications.
Collapse
Affiliation(s)
- Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
| | - Lucija Kadunc
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
- Graduate School of Biomedicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Mateja Manček Keber
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, Ljubljana 1000, Slovenia
| | - Iva Hafner Bratkovič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
| | - Rok Romih
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana 1000, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, Ljubljana 1000, Slovenia
| |
Collapse
|
32
|
Hughes RM. A compendium of chemical and genetic approaches to light-regulated gene transcription. Crit Rev Biochem Mol Biol 2018; 53:453-474. [PMID: 30040498 DOI: 10.1080/10409238.2018.1487382] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
On-cue regulation of gene transcription is an invaluable tool for the study of biological processes and the development and integration of next-generation therapeutics. Ideal reagents for the precise regulation of gene transcription should be nontoxic to the host system, highly tunable, and provide a high level of spatial and temporal control. Light, when coupled with protein or small molecule-linked photoresponsive elements, presents an attractive means of meeting the demands of an ideal system for regulating gene transcription. In this review, we cover recent developments in the burgeoning field of light-regulated gene transcription, covering both genetically encoded and small-molecule based strategies for optical regulation of transcription during the period 2012 till present.
Collapse
Affiliation(s)
- Robert M Hughes
- a Department of Chemistry , East Carolina University , Greenville , NC , USA
| |
Collapse
|
33
|
Bojar D, Scheller L, Hamri GCE, Xie M, Fussenegger M. Caffeine-inducible gene switches controlling experimental diabetes. Nat Commun 2018; 9:2318. [PMID: 29921872 PMCID: PMC6008335 DOI: 10.1038/s41467-018-04744-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 05/08/2018] [Indexed: 02/08/2023] Open
Abstract
Programming cellular behavior using trigger-inducible gene switches is integral to synthetic biology. Although significant progress has been achieved in trigger-induced transgene expression, side-effect-free remote control of transgenes continues to challenge cell-based therapies. Here, utilizing a caffeine-binding single-domain antibody we establish a caffeine-inducible protein dimerization system, enabling synthetic transcription factors and cell-surface receptors that enable transgene expression in response to physiologically relevant concentrations of caffeine generated by routine intake of beverages such as tea and coffee. Coffee containing different caffeine concentrations dose-dependently and reversibly controlled transgene expression by designer cells with this caffeine-stimulated advanced regulators (C-STAR) system. Type-2 diabetic mice implanted with microencapsulated, C-STAR-equipped cells for caffeine-sensitive expression of glucagon-like peptide 1 showed substantially improved glucose homeostasis after coffee consumption compared to untreated mice. Biopharmaceutical production control by caffeine, which is non-toxic, inexpensive and only present in specific beverages, is expected to improve patient compliance by integrating therapy with lifestyle.
Collapse
Affiliation(s)
- Daniel Bojar
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland
| | - Leo Scheller
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland
| | - Ghislaine Charpin-El Hamri
- IUT, Département Génie Biologique, Institut Universitaire de Technologie, F-69622, Villeurbanne Cedex, France
| | - Mingqi Xie
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland. .,Faculty of Life Science, University of Basel, Mattenstrasse 26, CH-4058, Basel, Switzerland.
| |
Collapse
|
34
|
Sedlmayer F, Aubel D, Fussenegger M. Synthetic gene circuits for the detection, elimination and prevention of disease. Nat Biomed Eng 2018; 2:399-415. [PMID: 31011195 DOI: 10.1038/s41551-018-0215-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/05/2018] [Indexed: 12/13/2022]
Abstract
In living organisms, naturally evolved sensors that constantly monitor and process environmental cues trigger corrective actions that enable the organisms to cope with changing conditions. Such natural processes have inspired biologists to construct synthetic living sensors and signalling pathways, by repurposing naturally occurring proteins and by designing molecular building blocks de novo, for customized diagnostics and therapeutics. In particular, designer cells that employ user-defined synthetic gene circuits to survey disease biomarkers and to autonomously re-adjust unbalanced pathological states can coordinate the production of therapeutics, with controlled timing and dosage. Furthermore, tailored genetic networks operating in bacterial or human cells have led to cancer remission in experimental animal models, owing to the network's unprecedented specificity. Other applications of designer cells in infectious, metabolic and autoimmune diseases are also being explored. In this Review, we describe the biomedical applications of synthetic gene circuits in major disease areas, and discuss how the first genetically engineered devices developed on the basis of synthetic-biology principles made the leap from the laboratory to the clinic.
Collapse
Affiliation(s)
- Ferdinand Sedlmayer
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Dominique Aubel
- IUTA Département Génie Biologique, Université Claude Bernard Lyon 1, Lyon, France
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland. .,Faculty of Science, University of Basel, Basel, Switzerland.
| |
Collapse
|
35
|
Yang W, Chen Q, Xia R, Zhang Y, Shuai L, Lai J, You X, Jiang Y, Bie P, Zhang L, Zhang H, Bai L. A novel bioscaffold with naturally-occurring extracellular matrix promotes hepatocyte survival and vessel patency in mouse models of heterologous transplantation. Biomaterials 2018; 177:52-66. [PMID: 29885586 DOI: 10.1016/j.biomaterials.2018.05.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/14/2018] [Accepted: 05/16/2018] [Indexed: 01/26/2023]
Abstract
BACKGROUND Naïve decellularized liver scaffold (nDLS)-based tissue engineering has been impaired by the lack of a suitable extracellular matrix (ECM) to provide "active micro-environmental" support. AIM The present study aimed to examine whether a novel, regenerative DLS (rDLS) with an active ECM improves primary hepatocyte survival and prevents thrombosis. METHODS rDLS was obtained from a 30-55% partial hepatectomy that was maintained in vivo for 3-5 days and then perfused with detergent in vitro. Compared to nDLS generated from normal livers, rDLS possesses bioactive molecules due to the regenerative period in vivo. Primary mouse hepatocyte survival was evaluated by staining for Ki-67 and Trypan blue exclusion. Thrombosis was assessed by immunohistochemistry and ex vivo diluted whole-blood perfusion. Hemocompatibility was determined by near-infrared laser-Doppler flowmetry and heterotopic transplantation. RESULTS After recellularization, rDLS contained more Ki-67-positive primary hepatocytes than nDLS. rDLS had a higher oxygen saturation and blood flow velocity and a lower expression of integrin αIIb and α4 than nDLS. Tumor necrosis factor-α, hepatocyte growth factor, interleukin-10, interleukin-6 and interleukin-1β were highly expressed throughout the rDLS, whereas expression of collagen-I, collagen-IV and thrombopoietin were lower in rDLS than in nDLS. Improved blood vessel patency was observed in rDLS both in vitro and in vivo. The results in mice were confirmed in large animals (pigs). CONCLUSION rDLS is an effective DLS with an "active microenvironment" that supports primary hepatocyte survival and promotes blood vessel patency. This is the first study to demonstrate a rDLS with a blood microvessel network that promotes hepatocyte survival and resists thrombosis.
Collapse
Affiliation(s)
- Wei Yang
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China; Hepatobiliary Institute, Southwest Hospital, The Army Medical University, Chongqing 400038, China
| | - Quanyu Chen
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China; Hepatobiliary Institute, Southwest Hospital, The Army Medical University, Chongqing 400038, China
| | - Renpei Xia
- Hepatobiliary Institute, Southwest Hospital, The Army Medical University, Chongqing 400038, China
| | - Yujun Zhang
- Hepatobiliary Institute, Southwest Hospital, The Army Medical University, Chongqing 400038, China
| | - Ling Shuai
- Hepatobiliary Institute, Southwest Hospital, The Army Medical University, Chongqing 400038, China
| | - Jiejuan Lai
- Hepatobiliary Institute, Southwest Hospital, The Army Medical University, Chongqing 400038, China
| | - Xiaolin You
- Hepatobiliary Institute, Southwest Hospital, The Army Medical University, Chongqing 400038, China
| | - Yan Jiang
- Hepatobiliary Institute, Southwest Hospital, The Army Medical University, Chongqing 400038, China
| | - Ping Bie
- Hepatobiliary Institute, Southwest Hospital, The Army Medical University, Chongqing 400038, China
| | - Leida Zhang
- Hepatobiliary Institute, Southwest Hospital, The Army Medical University, Chongqing 400038, China.
| | - Hongyu Zhang
- Hepatobiliary Institute, Southwest Hospital, The Army Medical University, Chongqing 400038, China.
| | - Lianhua Bai
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China; Hepatobiliary Institute, Southwest Hospital, The Army Medical University, Chongqing 400038, China.
| |
Collapse
|
36
|
Dou J, Bennett MR. Synthetic Biology and the Gut Microbiome. Biotechnol J 2018; 13:e1700159. [PMID: 28976641 PMCID: PMC5882594 DOI: 10.1002/biot.201700159] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/23/2017] [Indexed: 12/19/2022]
Abstract
The gut microbiome plays a crucial role in maintaining human health. Functions performed by gastrointestinal microbes range from regulating metabolism to modulating immune and nervous system development. Scientists have attempted to exploit this importance through the development of engineered probiotics that are capable of producing and delivering small molecule therapeutics within the gut. However, existing synthetic probiotics are simplistic and fail to replicate the complexity and adaptability of native homeostatic mechanisms. In this review, the ways in which the tools and approaches of synthetic biology have been applied to improve the efficacy of therapeutic probiotics, and the ways in which they might be applied in the future is discussed. Simple devices, such as a bistable switches and integrase memory arrays, have been successfully implemented in the mammalian gut, and models for targeted delivery in this environment have also been developed. In the future, it will be necessary to introduce concepts such as logic-gating and biocontainment mechanisms into synthetic probiotics, as well as to expand the collection of relevant biosensors. Ideally, this will bring us closer to a reality in which engineered therapeutic microbes will be able to accurately diagnose and effectively respond to a variety of disease states.
Collapse
Affiliation(s)
- Jennifer Dou
- Department of Biosciences, Rice University, Houston, TX 77005
| | - Matthew R. Bennett
- Department of Biosciences, Rice University, Houston, TX 77005
- Department of Bioengineering, Rice University, Houston, TX 77005
| |
Collapse
|
37
|
Kitada T, DiAndreth B, Teague B, Weiss R. Programming gene and engineered-cell therapies with synthetic biology. Science 2018; 359:359/6376/eaad1067. [PMID: 29439214 DOI: 10.1126/science.aad1067] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Gene and engineered-cell therapies promise to treat diseases by genetically modifying cells to carry out therapeutic tasks. Although the field has had some success in treating monogenic disorders and hematological malignancies, current approaches are limited to overexpression of one or a few transgenes, constraining the diseases that can be treated with this approach and leading to potential concerns over safety and efficacy. Synthetic gene networks can regulate the dosage, timing, and localization of gene expression and therapeutic activity in response to small molecules and disease biomarkers. Such "programmable" gene and engineered-cell therapies will provide new interventions for incurable or difficult-to-treat diseases.
Collapse
Affiliation(s)
- Tasuku Kitada
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Breanna DiAndreth
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Brian Teague
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ron Weiss
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
38
|
Abstract
The programming of new functions into mammalian cells has tremendous application in research and medicine. Continued improvements in the capacity to sequence and synthesize DNA have rapidly increased our understanding of mechanisms of gene function and regulation on a genome-wide scale and have expanded the set of genetic components available for programming cell biology. The invention of new research tools, including targetable DNA-binding systems such as CRISPR/Cas9 and sensor-actuator devices that can recognize and respond to diverse chemical, mechanical, and optical inputs, has enabled precise control of complex cellular behaviors at unprecedented spatial and temporal resolution. These tools have been critical for the expansion of synthetic biology techniques from prokaryotic and lower eukaryotic hosts to mammalian systems. Recent progress in the development of genome and epigenome editing tools and in the engineering of designer cells with programmable genetic circuits is expanding approaches to prevent, diagnose, and treat disease and to establish personalized theranostic strategies for next-generation medicines. This review summarizes the development of these enabling technologies and their application to transforming mammalian synthetic biology into a distinct field in research and medicine.
Collapse
Affiliation(s)
- Joshua B Black
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708; , .,Center for Genomic and Computational Biology, Duke University, Durham, North Carolina 27708
| | - Pablo Perez-Pinera
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; .,Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708; , .,Center for Genomic and Computational Biology, Duke University, Durham, North Carolina 27708.,Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
39
|
|
40
|
Wang Y, Wang M, Dong K, Ye H. Engineering Mammalian Designer Cells for the Treatment of Metabolic Diseases. Biotechnol J 2017; 13:e1700160. [PMID: 29144600 DOI: 10.1002/biot.201700160] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/03/2017] [Indexed: 12/22/2022]
Abstract
Synthetic biology applies engineering principles to biological systems and has significantly advanced the design of synthetic gene circuits that can reprogram cell activities to perform new functions. The ability to engineer mammalian designer cells with robust therapeutic behaviors has brought new opportunities for treating metabolic diseases. In this review, the authors highlight the most recent advances in the development of synthetic designer cells uploaded with open- or closed-loop gene circuits for the treatment of metabolic disorders including diabetes, hypertension, hyperuricemia, and obesity, and discuss the current technologies and future perspectives in applying these designer cells for clinical applications. In the future, more and more rationally designed cells will be constructed and revolutionized to treat a number of metabolic disorders in an intelligent manner.
Collapse
Affiliation(s)
- Yidan Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Dongchuan Road 500, Shanghai 200241, China
| | - Meiyan Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Dongchuan Road 500, Shanghai 200241, China
| | - Kaili Dong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Dongchuan Road 500, Shanghai 200241, China
| | - Haifeng Ye
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Dongchuan Road 500, Shanghai 200241, China
| |
Collapse
|
41
|
Muldoon JJ, Donahue PS, Dolberg TB, Leonard JN. Building with intent: technologies and principles for engineering mammalian cell-based therapies to sense and respond. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2017; 4:127-133. [PMID: 29450405 DOI: 10.1016/j.cobme.2017.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The engineering of cells as programmable devices has enabled therapeutic strategies that could not otherwise be achieved. Such strategies include recapitulating and enhancing native cellular functions and composing novel functions. These novel functions may be composed using both natural and engineered biological components, with the latter exemplified by the development of synthetic receptor and signal transduction systems. Recent advances in implementing these approaches include the treatment of cancer, where the most clinical progress has been made to date, and the treatment of diabetes. Principles for engineering cell-based therapies that are safe and effective are increasingly needed and beginning to emerge, and will be essential in the development of this new class of therapeutics.
Collapse
Affiliation(s)
- Joseph J Muldoon
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States.,Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, Illinois 60208, United States
| | - Patrick S Donahue
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States.,Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, Illinois 60208, United States.,Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Taylor B Dolberg
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Joshua N Leonard
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States.,Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, Illinois 60208, United States.,Center for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, United States.,Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States.,Member, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
42
|
|
43
|
Higashikuni Y, Chen WC, Lu TK. Advancing therapeutic applications of synthetic gene circuits. Curr Opin Biotechnol 2017; 47:133-141. [PMID: 28750201 DOI: 10.1016/j.copbio.2017.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 06/21/2017] [Indexed: 02/07/2023]
Abstract
Synthetic biology aims to introduce new sense-and-respond capabilities into living cells, which would enable novel therapeutic strategies. The development of regulatory elements, molecular computing devices, and effector screening technologies has enabled researchers to design synthetic gene circuits in many organisms, including mammalian cells. Engineered gene networks, such as closed-loop circuits or Boolean logic gate circuits, can be used to program cells to perform specific functions with spatiotemporal control and restoration of homeostasis in response to the extracellular environment and intracellular signaling. In addition, genetically modified microbes can be designed as local delivery of therapeutic molecules. In this review, we will discuss recent advances in therapeutic applications of synthetic gene circuits, as well as challenges and future opportunities for biomedicine.
Collapse
Affiliation(s)
- Yasutomi Higashikuni
- Research Laboratory of Electronics, Massachusetts Institute of Technology, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, MA 02139, USA
| | - William Cw Chen
- Research Laboratory of Electronics, Massachusetts Institute of Technology, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, MA 02139, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Timothy K Lu
- Research Laboratory of Electronics, Massachusetts Institute of Technology, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, MA 02139, USA.
| |
Collapse
|
44
|
|
45
|
Teixeira AP, Fussenegger M. Synthetic biology-inspired therapies for metabolic diseases. Curr Opin Biotechnol 2017; 47:59-66. [PMID: 28662442 DOI: 10.1016/j.copbio.2017.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 06/13/2017] [Indexed: 12/15/2022]
Abstract
Our ability to engineer mammalian cells with effective therapeutic behaviors has brought new hope for treating metabolic diseases. Synthetic gene networks have been customized to interface with the host metabolism, discriminate between healthy and diseased states, and respond by producing an adjusted dose of the therapeutic molecule. Such devices have the potential to treat a range of dysfunctions that are simply not addressable using conventional therapies. Recently, the repurposing of native signaling pathways has formed the basis of autonomous therapeutic programs genetically installed in mammalian cells and has greatly expanded the possibilities to effectively tackle metabolic disorders. Here, we outline network topologies that have been successfully validated in animal models of metabolic diseases and discuss future developments that will be important for bringing this technology closer to clinical application.
Collapse
Affiliation(s)
- Ana Palma Teixeira
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland; Instituto de Tecnologia Química e Biológica António Xavier, ITQB-NOVA, Avenida da República, 2780-157 Oeiras, Portugal; IBET, Instituto de Biologia Experimental e Tecnológica, Avenida da República, 2781-157 Oeiras, Portugal
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland; University of Basel, Faculty of Science, Mattenstrasse 26, CH-4058 Basel, Switzerland.
| |
Collapse
|
46
|
Ho P, Chen YY. Mammalian synthetic biology in the age of genome editing and personalized medicine. Curr Opin Chem Biol 2017. [PMID: 28628856 DOI: 10.1016/j.cbpa.2017.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The recent expansion of molecular tool kits has propelled synthetic biology toward the design of increasingly sophisticated mammalian systems. Specifically, advances in genome editing, protein engineering, and circuitry design have enabled the programming of cells for diverse applications, including regenerative medicine and cancer immunotherapy. The ease with which molecular and cellular interactions can be harnessed promises to yield novel approaches to elucidate genetic interactions, program cellular functions, and design therapeutic interventions. Here, we review recent advancements in the development of enabling technologies and the practical applications of mammalian synthetic biology.
Collapse
Affiliation(s)
- Patrick Ho
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, 420 Westwood Plaza, Boelter Hall 5531, Los Angeles, CA 90095, USA
| | - Yvonne Y Chen
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, 420 Westwood Plaza, Boelter Hall 5531, Los Angeles, CA 90095, USA.
| |
Collapse
|
47
|
Abstract
By using tools from synthetic biology, sophisticated genetic devices can be assembled to reprogram mammalian cell activities. Here, we demonstrate that a self-adjusting synthetic gene circuit can be designed to sense and reverse the insulin-resistance syndrome in different mouse models. By functionally rewiring the mitogen-activated protein kinase (MAPK) signalling pathway to produce MAPK-mediated activation of the hybrid transcription factor TetR-ELK1, we assembled a synthetic insulin-sensitive transcription-control device that self-sufficiently distinguished between physiological and increased blood insulin levels and correspondingly fine-tuned the reversible expression of therapeutic transgenes from synthetic TetR-ELK1-specific promoters. In acute experimental hyperinsulinemia, the synthetic insulin-sensing designer circuit reversed the insulin-resistance syndrome by coordinating expression of the insulin-sensitizing compound adiponectin. Engineering synthetic gene circuits to sense pathologic markers and coordinate the expression of therapeutic transgenes may provide opportunities for future gene- and cell-based treatments of multifactorial metabolic disorders.
Collapse
|