1
|
Liu H, Su P, Li Y, Hoover A, Hu S, King SA, Zhao J, Guan JL, Chen SY, Zhao Y, Tan M, Wu X. VAMP2 controls murine epidermal differentiation and carcinogenesis by regulation of nucleophagy. Dev Cell 2024; 59:2005-2016.e4. [PMID: 38810653 PMCID: PMC11303110 DOI: 10.1016/j.devcel.2024.05.004] [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: 09/26/2023] [Revised: 02/16/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024]
Abstract
Differentiation of murine epidermal stem/progenitor cells involves the permanent withdrawal from the cell cycle, the synthesis of various protein and lipid components for the cornified envelope, and the controlled dissolution of cellular organelles and nuclei. Deregulated epidermal differentiation contributes to the development of various skin diseases, including skin cancers. With a genome-wide shRNA screen, we identified vesicle-associated membrane protein 2 (VAMP2) as a critical factor involved in skin differentiation. Deletion of VAMP2 leads to aberrant skin stratification and enucleation in vivo. With quantitative proteomics, we further identified an autophagy protein, focal adhesion kinase family interacting protein of 200 kDa (FIP200), as a binding partner of VAMP2. Additionally, we showed that both VAMP2 and FIP200 are critical for murine keratinocyte enucleation and epidermal differentiation. Loss of VAMP2 or FIP200 enhances cutaneous carcinogenesis in vivo. Together, our findings identify important molecular mechanisms underlying epidermal differentiation and skin tumorigenesis.
Collapse
Affiliation(s)
- Han Liu
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Peihong Su
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Yuanyuan Li
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Alex Hoover
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Sophie Hu
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Sarah A King
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Jing Zhao
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Shao-Yu Chen
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Yingming Zhao
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Minjia Tan
- The Chemical Proteomics Center and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Xiaoyang Wu
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
2
|
Zhou S, Zhao L, Zuo W, Zheng Y, Zhang P, Sun Y, Wang Y, Du G, Kang Z. Minimizing endogenous cryptic plasmids to construct antibiotic-free expression systems for Escherichia coli Nissle 1917. Synth Syst Biotechnol 2024; 9:165-175. [PMID: 38348398 PMCID: PMC10859263 DOI: 10.1016/j.synbio.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/25/2023] [Accepted: 01/11/2024] [Indexed: 02/15/2024] Open
Abstract
The probiotic bacterium Escherichia coli Nissle 1917 (EcN) holds significant promise for use in clinical and biological industries. However, the reliance on antibiotics to maintain plasmid-borne genes has overshadowed its benefits. In this study, we addressed this issue by engineering the endogenous cryptic plasmids pMUT1 and pMUT2. The non-essential elements were removed to create more stable derivatives pMUT1NR△ and pMUT2HBC△. Synthetic promoters by integrating binding motifs on sigma factors were further constructed and applied for expression of Bacteroides thetaiotaomicron heparinase III and the biosynthesis of ectoine. Compared to traditional antibiotic-dependent expression systems, our newly constructed antibiotic-free expression systems offer considerable advantages for clinical and synthetic biology applications.
Collapse
Affiliation(s)
- Siyan Zhou
- The Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Linlin Zhao
- The Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Wenjie Zuo
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Yilin Zheng
- The Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Ping Zhang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Yanan Sun
- The Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Yang Wang
- The Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Guocheng Du
- The Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Zhen Kang
- The Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| |
Collapse
|
3
|
Abstract
Diseases associated with nicotine dependence in the form of habitual tobacco use are a major cause of premature death in the United States. The majority of tobacco smokers will relapse within the first month of attempted abstinence. Smoking cessation agents increase the likelihood that smokers can achieve long-term abstinence. Nevertheless, currently available smoking cessation agents have limited utility and fail to prevent relapse in the majority of smokers. Pharmacotherapy is therefore an effective strategy to aid smoking cessation efforts but considerable risk of relapse persists even when the most efficacious medications currently available are used. The past decade has seen major breakthroughs in our understanding of the molecular, cellular, and systems-level actions of nicotine in the brain that contribute to the development and maintenance of habitual tobacco use. In parallel, large-scale human genetics studies have revealed allelic variants that influence vulnerability to tobacco use disorder. These advances have revealed targets for the development of novel smoking cessation agents. Here, we summarize current efforts to develop smoking cessation therapeutics and highlight opportunities for future efforts.
Collapse
Affiliation(s)
- Dana Lengel
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paul J. Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Drug Discovery Institute (DDI), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
4
|
Current advances and future prospects in production of recombinant insulin and other proteins to treat diabetes mellitus. Biotechnol Lett 2022; 44:643-669. [DOI: 10.1007/s10529-022-03247-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 03/16/2022] [Indexed: 12/14/2022]
|
5
|
Prasathkumar M, Becky R, Anisha S, Dhrisya C, Sadhasivam S. Evaluation of hypoglycemic therapeutics and nutritional supplementation for type 2 diabetes mellitus management: An insight on molecular approaches. Biotechnol Lett 2022; 44:203-238. [PMID: 35119572 DOI: 10.1007/s10529-022-03232-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 01/28/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This review aims to summarize the current management of type 2 diabetes principles, including oral hypoglycemic agents, types of insulin administration, diet maintenance, and various molecular approaches. METHODS A literature search was conducted in different databases such as Scopus, ScienceDirect, Google Scholar, and Web of Science by using the following keywords: type-2 diabetes mellitus (T2DM), first-line and second-line treatment, oral hypoglycemic agents, insulin administration, diet/nutritional therapy, gene and stem cell therapy, and diabetic complications. RESULTS The first-line treatment of T2DM includes administering oral hypoglycemic agents (OHAs) and second-line treatment by insulin therapy and some OHAs like Sulfonylurea's (SU). The oral hypoglycemic or oral antidiabetic drugs have the function of lowering glucose in the blood. Insulin therapy is recommended for people with A1C levels > 7.0, and insulin administration is evolved drastically from the syringe, pump, pen, inhalation, insulin jet, and patch. The use of OHAs and insulin therapy during glycemic control has a severe effect on weight gain and other side effects. Hence, diet maintenance (macro and micronutrients) and nutritional therapy guidelines were also reviewed/recommended for safe T2DM management. Besides, the recent progress in molecular approaches that focuses on identifying new targets for T2DM (i.e.) consisting of gene therapy, stem cell therapy, and the modulation of insulin signaling pathways for the regulation of glucose storage and uptake also discussed. CONCLUSION The analysis of all these key factors is necessary to develop a potential agent to cure T2DM and suggest that a combination of therapies will pave the way for advanced management of T2DM.
Collapse
Affiliation(s)
- Murugan Prasathkumar
- Bioprocess and Biomaterials Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, India
| | - Robert Becky
- Bioprocess and Biomaterials Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, India
| | - Salim Anisha
- Bioprocess and Biomaterials Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, India
| | - Chenthamara Dhrisya
- Bioprocess and Biomaterials Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, India
| | - Subramaniam Sadhasivam
- Bioprocess and Biomaterials Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, India.
- Department of Extension and Career Guidance, Bharathiar University, Coimbatore, 641046, India.
| |
Collapse
|
6
|
Smits JP, Meesters LD, Maste BG, Zhou H, Zeeuwen PL, van den Bogaard EH. CRISPR-Cas9 based genomic engineering in keratinocytes: from technology to application. JID INNOVATIONS 2021; 2:100082. [PMID: 35146483 PMCID: PMC8819031 DOI: 10.1016/j.xjidi.2021.100082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/13/2021] [Accepted: 11/18/2021] [Indexed: 12/14/2022] Open
Affiliation(s)
- Jos P.H. Smits
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Luca D. Meesters
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Berber G.W. Maste
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Huiqing Zhou
- Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center (Radboudumc), Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Patrick L.J.M. Zeeuwen
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Ellen H. van den Bogaard
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
- Correspondence: Ellen H. van den Bogaard, Department of Dermatology, Radboud University Medical Center (Radboudumc), Radboud Institute for Molecular Life Sciences (RIMLS), Rene Descartesdreef 1, Nijmegen 6525 GL, The Netherlands.
| |
Collapse
|
7
|
Goldenberg D, McLaughlin C, Koduru SV, Ravnic DJ. Regenerative Engineering: Current Applications and Future Perspectives. Front Surg 2021; 8:731031. [PMID: 34805257 PMCID: PMC8595140 DOI: 10.3389/fsurg.2021.731031] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/13/2021] [Indexed: 12/12/2022] Open
Abstract
Many pathologies, congenital defects, and traumatic injuries are untreatable by conventional pharmacologic or surgical interventions. Regenerative engineering represents an ever-growing interdisciplinary field aimed at creating biological replacements for injured tissues and dysfunctional organs. The need for bioengineered replacement parts is ubiquitous among all surgical disciplines. However, to date, clinical translation has been limited to thin, small, and/or acellular structures. Development of thicker tissues continues to be limited by vascularization and other impediments. Nevertheless, currently available materials, methods, and technologies serve as robust platforms for more complex tissue fabrication in the future. This review article highlights the current methodologies, clinical achievements, tenacious barriers, and future perspectives of regenerative engineering.
Collapse
Affiliation(s)
- Dana Goldenberg
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, United States
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Caroline McLaughlin
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, United States
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Srinivas V. Koduru
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, United States
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Dino J. Ravnic
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, United States
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, United States
| |
Collapse
|
8
|
Kong Q, Wu X, Xu M. A Genetically Modified Skin Graft for Treating Alcohol Use Disorder and/or Polysubstance Abuse With Cocaine. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2021; 1:10007. [PMID: 38390403 PMCID: PMC10880775 DOI: 10.3389/adar.2021.10007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/21/2021] [Indexed: 02/24/2024]
Abstract
Alcohol use disorder (AUD) is one of the foremost public health problems. Alcohol is also frequently co-abused with cocaine. There is a huge unmet need for the treatment of AUD and/or cocaine co-abuse. We have developed and used a skin stem cell-based gene delivery platform and found that production of the glucagon-like peptide-1 (GLP1) from the grafted genetically modified skin reduced development and reinstatement of alcohol-induced drug-taking and seeking, voluntary oral alcohol consumption and alcohol-induced increase in dopamine (DA) levels in the nucleus accumbens (NAc). Moreover, we have developed a novel co-grafting procedure for both modified human butyrylcholinesterase (hBChE)- and GLP1-expressing cells. Skin grafts-derived hBChE and GLP1 reduced acquisition of drug-taking and toxicity induced by concurrent alcohol and cocaine injections. These results imply that gene delivery through skin transplants may add a new option to treat drug abuse and co-abuse.
Collapse
Affiliation(s)
- Qingyao Kong
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, United States
| | - Xiaoyang Wu
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, United States
| | - Ming Xu
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
9
|
Shi H, Smits JPH, van den Bogaard EH, Brewer MG. Research Techniques Made Simple: Delivery of the CRISPR/Cas9 Components into Epidermal Cells. J Invest Dermatol 2021; 141:1375-1381.e1. [PMID: 34024338 PMCID: PMC8224101 DOI: 10.1016/j.jid.2021.01.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 10/21/2022]
Abstract
CRISPR/Cas9 technology is a powerful tool used to alter the genetic landscape of various hosts. This has been exemplified by its success in the transgenic animal world where it has been utilized to develop novel mouse lines modeling numerous disease states. The technology has helped to develop both in vitro and in vivo systems that simulate diseases within the fields of epithelial biology, skin cancer biology, dermatology, and beyond. Importantly, the delivery of the single-guide RNA/Cas9 editing complex to the host cell is key for its success. In this paper, we discuss the various methods that have been utilized as delivery techniques for CRISPR/Cas9 components, the benefits and pitfalls of each, and how successful they have been at genetically modifying epidermal cells. In addition, we acknowledge recent advances in the field of dermatology that have harnessed these methods to better understand epidermal biology, identify potential therapeutic targets, or serve as novel methods to treat disease states.
Collapse
Affiliation(s)
- Huishan Shi
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Jos P H Smits
- Department of Dermatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ellen H van den Bogaard
- Department of Dermatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Matthew G Brewer
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA.
| |
Collapse
|
10
|
Liu J, Cao J, Li Y, Guo F. Beneficial Flavonoid in Foods and Anti-obesity Effect. FOOD REVIEWS INTERNATIONAL 2021. [DOI: 10.1080/87559129.2021.1923730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jingwen Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiaoxian Cao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fujiang Guo
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
11
|
Lee J, Wu Y, Harada BT, Li Y, Zhao J, He C, Ma Y, Wu X. N 6 -methyladenosine modification of lncRNA Pvt1 governs epidermal stemness. EMBO J 2021; 40:e106276. [PMID: 33729590 DOI: 10.15252/embj.2020106276] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 02/02/2021] [Accepted: 02/15/2021] [Indexed: 11/09/2022] Open
Abstract
Dynamic chemical modifications of RNA represent novel and fundamental mechanisms that regulate stemness and tissue homeostasis. Rejuvenation and wound repair of mammalian skin are sustained by epidermal progenitor cells, which are localized within the basal layer of the skin epidermis. N6 -methyladenosine (m6 A) is one of the most abundant modifications found in eukaryotic mRNA and lncRNA (long noncoding RNA). In this report, we survey changes of m6 A RNA methylomes upon epidermal differentiation and identify Pvt1, a lncRNA whose m6 A modification is critically involved in sustaining stemness of epidermal progenitor cells. With genome-editing and a mouse genetics approach, we show that ablation of m6 A methyltransferase or Pvt1 impairs the self-renewal and wound healing capability of skin. Mechanistically, methylation of Pvt1 transcripts enhances its interaction with MYC and stabilizes the MYC protein in epidermal progenitor cells. Our study presents a global view of epitranscriptomic dynamics that occur during epidermal differentiation and identifies the m6 A modification of Pvt1 as a key signaling event involved in skin tissue homeostasis and wound repair.
Collapse
Affiliation(s)
- Jimmy Lee
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Yuchen Wu
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA.,Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bryan T Harada
- Department of Chemistry, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.,Department of Biochemistry and Molecular Biology, Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Yuanyuan Li
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Jing Zhao
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Chuan He
- Department of Chemistry, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.,Department of Biochemistry and Molecular Biology, Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Yanlei Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoyang Wu
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
12
|
A pulsatile release platform based on photo-induced imine-crosslinking hydrogel promotes scarless wound healing. Nat Commun 2021; 12:1670. [PMID: 33723267 PMCID: PMC7960722 DOI: 10.1038/s41467-021-21964-0] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
Effective healing of skin wounds is essential for our survival. Although skin has strong regenerative potential, dysfunctional and disfiguring scars can result from aberrant wound repair. Skin scarring involves excessive deposition and misalignment of ECM (extracellular matrix), increased cellularity, and chronic inflammation. Transforming growth factor-β (TGFβ) signaling exerts pleiotropic effects on wound healing by regulating cell proliferation, migration, ECM production, and the immune response. Although blocking TGFβ signaling can reduce tissue fibrosis and scarring, systemic inhibition of TGFβ can lead to significant side effects and inhibit wound re-epithelization. In this study, we develop a wound dressing material based on an integrated photo-crosslinking strategy and a microcapsule platform with pulsatile release of TGF-β inhibitor to achieve spatiotemporal specificity for skin wounds. The material enhances skin wound closure while effectively suppressing scar formation in murine skin wounds and large animal preclinical models. Our study presents a strategy for scarless wound repair. Dysfunctional and disfiguring scars can result from aberrant wound repair. Here, the authors develop a wound dressing material based on an integrated photo-crosslinking strategy and a microcapsule platform with pulsatile release of TGF-β inhibitor to achieve spatiotemporal specificity for scarless wound repair.
Collapse
|
13
|
Kong Q, Li Y, Yue J, Wu X, Xu M. Reducing alcohol and/or cocaine-induced reward and toxicity via an epidermal stem cell-based gene delivery platform. Mol Psychiatry 2021; 26:5266-5276. [PMID: 33619338 PMCID: PMC8380265 DOI: 10.1038/s41380-021-01043-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/14/2021] [Accepted: 02/01/2021] [Indexed: 01/31/2023]
Abstract
Alcohol use disorder (AUD) is one of the foremost public health problems. Alcohol is also frequently co-abused with cocaine. There is a huge unmet need for the treatment of AUD and/or cocaine co-abuse. We recently demonstrated that skin grafts generated from mouse epidermal stem cells that had been engineered by CRISPR-mediated genome editing could be transplanted onto mice as a gene delivery platform. Here, we show that expression of the glucagon-like peptide-1 (GLP1) gene delivered by epidermal stem cells attenuated development and reinstatement of alcohol-induced drug-taking and seeking as well as voluntary oral alcohol consumption. GLP1 derived from the skin grafts decreased alcohol-induced increase in dopamine levels in the nucleus accumbens. In exploring the potential of this platform in reducing concurrent use of drugs, we developed a novel co-grafting procedure for both modified human butyrylcholinesterase (hBChE)- and GLP1-expressing cells. Epidermal stem cell-derived hBChE and GLP1 reduced acquisition of drug-taking and toxicity induced by alcohol and cocaine co-administration. These results imply that cutaneous gene delivery through skin transplants may add a new option to treat drug abuse and co-abuse.
Collapse
Affiliation(s)
- Qingyao Kong
- grid.170205.10000 0004 1936 7822Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL USA
| | - Yuanyuan Li
- grid.170205.10000 0004 1936 7822Ben May Department for Cancer Research, The University of Chicago, Chicago, IL USA
| | - Jiping Yue
- grid.170205.10000 0004 1936 7822Ben May Department for Cancer Research, The University of Chicago, Chicago, IL USA
| | - Xiaoyang Wu
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA.
| | - Ming Xu
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
14
|
Yin J, Yang L, Mou L, Dong K, Jiang J, Xue S, Xu Y, Wang X, Lu Y, Ye H. A green tea-triggered genetic control system for treating diabetes in mice and monkeys. Sci Transl Med 2020; 11:11/515/eaav8826. [PMID: 31645456 DOI: 10.1126/scitranslmed.aav8826] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 05/20/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022]
Abstract
Cell-based therapies are recognized as the next frontier in medicine, but the translation of many promising technologies into the clinic is currently limited by a lack of remote-control inducers that are safe and can be tightly regulated. Here, we developed therapeutically active engineered cells regulated by a control system that is responsive to protocatechuic acid (PCA), a metabolite found in green tea. We constructed multiple genetic control technologies that could toggle a PCA-responsive ON/OFF switch based on a transcriptional repressor from Streptomyces coelicolor We demonstrated that PCA-controlled switches can be used for guide RNA expression-mediated control of the CRISPR-Cas9 systems for gene editing and epigenetic remodeling. We showed how these technologies could be used as implantable biocomputers in live mice to perform complex logic computations that integrated signals from multiple food metabolites. Last, we used our system to treat type 1 and type 2 diabetes in mice and cynomolgus monkeys. This biocompatible and versatile food phenolic acid-controlled transgenic device opens opportunities for dynamic interventions in gene- and cell-based precision medicine.
Collapse
Affiliation(s)
- Jianli Yin
- Synthetic Biology and Biomedical Engineering Laboratory, Biomedical Synthetic Biology Research Center, 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
| | - Linfeng Yang
- Synthetic Biology and Biomedical Engineering Laboratory, Biomedical Synthetic Biology Research Center, 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
| | - Lisha Mou
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, China
| | - Kaili Dong
- Synthetic Biology and Biomedical Engineering Laboratory, Biomedical Synthetic Biology Research Center, 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
| | - Jian Jiang
- Synthetic Biology and Biomedical Engineering Laboratory, Biomedical Synthetic Biology Research Center, 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
| | - Shuai Xue
- Synthetic Biology and Biomedical Engineering Laboratory, Biomedical Synthetic Biology Research Center, 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
| | - Ying Xu
- Synthetic Biology and Biomedical Engineering Laboratory, Biomedical Synthetic Biology Research Center, 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
| | - Xinyi Wang
- Synthetic Biology and Biomedical Engineering Laboratory, Biomedical Synthetic Biology Research Center, 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
| | - Ying Lu
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, China
| | - Haifeng Ye
- Synthetic Biology and Biomedical Engineering Laboratory, Biomedical Synthetic Biology Research Center, 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
|
15
|
Fortunel NO, Chadli L, Coutier J, Lemaître G, Auvré F, Domingues S, Bouissou-Cadio E, Vaigot P, Cavallero S, Deleuze JF, Roméo PH, Martin MT. KLF4 inhibition promotes the expansion of keratinocyte precursors from adult human skin and of embryonic-stem-cell-derived keratinocytes. Nat Biomed Eng 2019; 3:985-997. [PMID: 31636412 DOI: 10.1038/s41551-019-0464-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 09/13/2019] [Indexed: 01/01/2023]
Abstract
Expanded autologous skin keratinocytes are currently used in cutaneous cell therapy, and embryonic-stem-cell-derived keratinocytes could become a complementary alternative. Regardless of keratinocyte provenance, for efficient therapy it is necessary to preserve immature keratinocyte precursors during cell expansion and graft processing. Here, we show that stable and transient downregulation of the transcription factor Krüppel-like factor 4 (KLF4) in keratinocyte precursors from adult skin, using anti-KLF4 RNA interference or kenpaullone, promotes keratinocyte immaturity and keratinocyte self-renewal in vitro, and enhances the capacity for epidermal regeneration in mice. Both stable and transient KLF4 downregulation had no impact on the genomic integrity of adult keratinocytes. Moreover, transient KLF4 downregulation in human-embryonic-stem-cell-derived keratinocytes increased the efficiency of skin-orientated differentiation and of keratinocyte immaturity, and was associated with improved generation of epidermis. As a regulator of the cell fate of keratinocyte precursors, KLF4 could be used for promoting the ex vivo expansion and maintenance of functional immature keratinocyte precursors.
Collapse
Affiliation(s)
- Nicolas O Fortunel
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France. .,INSERM U967, Université Paris-Diderot, Paris, France. .,Université Paris-Saclay, Paris, France.
| | - Loubna Chadli
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France
| | - Julien Coutier
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France
| | - Gilles Lemaître
- Université d'Evry Val d'Essonne, Université Paris-Saclay, INSERM U861, Institut des Cellules Souches pour le Traitement et l'Etude des Maladies Monogéniques, Corbeil Essonne, France
| | - Frédéric Auvré
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France
| | - Sophie Domingues
- Centre d'Etude des Cellules Souches, Institut des Cellules Souches pour le Traitement et l'Etude des Maladies Monogéniques, Corbeil Essonne, France
| | - Emmanuelle Bouissou-Cadio
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France
| | - Pierre Vaigot
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France
| | - Sophie Cavallero
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France
| | | | - Paul-Henri Roméo
- INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France.,Laboratoire de Recherche sur la Réparation et la Transcription dans les Cellules Souches, CEA/DRF/IBFJ/IRCM, Fontenay-aux-Roses, France
| | - Michèle T Martin
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France. .,INSERM U967, Université Paris-Diderot, Paris, France. .,Université Paris-Saclay, Paris, France.
| |
Collapse
|
16
|
El-Kenawy A, Benarba B, Neves AF, de Araujo TG, Tan BL, Gouri A. Gene surgery: Potential applications for human diseases. EXCLI JOURNAL 2019; 18:908-930. [PMID: 31762718 PMCID: PMC6868916 DOI: 10.17179/excli2019-1833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022]
Abstract
Gene therapy became in last decade a new emerging therapeutic era showing promising results against different diseases such as cancer, cardiovascular diseases, diabetes, and neurological disorders. Recently, the genome editing technique for eukaryotic cells called CRISPR-Cas (Clustered Regulatory Interspaced Short Palindromic Repeats) has enriched the field of gene surgery with enhanced applications. In the present review, we summarized the different applications of gene surgery for treating human diseases such as cancer, diabetes, nervous, and cardiovascular diseases, besides the molecular mechanisms involved in these important effects. Several studies support the important therapeutic applications of gene surgery in a large number of health disorders and diseases including β-thalassemia, cancer, immunodeficiencies, diabetes, and neurological disorders. In diabetes, gene surgery was shown to be effective in type 1 diabetes by triggering different signaling pathways. Furthermore, gene surgery, especially that using CRISPR-Cas possessed important application on diagnosis, screening and treatment of several cancers such as lung, liver, pancreatic and colorectal cancer. Nevertheless, gene surgery still presents some limitations such as the design difficulties and costs regarding ZFNs (Zinc Finger Nucleases) and TALENs (Transcription Activator-Like Effector Nucleases) use, off-target effects, low transfection efficiency, in vivo delivery-safety and ethical issues.
Collapse
Affiliation(s)
- Ayman El-Kenawy
- Department of Pathology, College of Medicine, Taif University, Saudi Arabia
- Department of Molecular Biology, GEBRI, University of Sadat City, P.O. Box 79, Sadat City, Egypt
| | - Bachir Benarba
- Laboratory Research on Biological Systems and Geomatics, Faculty of Nature and Life Sciences, University of Mascara, Algeria
| | - Adriana Freitas Neves
- Institute of Biotechnology, Molecular Biology Laboratory, Universidade Federal de Goias, Catalao, Brazil
| | - Thaise Gonçalves de Araujo
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas, MG, Brazil
| | - Bee Ling Tan
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Adel Gouri
- Laboratory of Medical Biochemistry, Faculty of Medicine, University of Annaba, Algeria
| |
Collapse
|
17
|
Kotagama OW, Jayasinghe CD, Abeysinghe T. Era of Genomic Medicine: A Narrative Review on CRISPR Technology as a Potential Therapeutic Tool for Human Diseases. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1369682. [PMID: 31687377 PMCID: PMC6800964 DOI: 10.1155/2019/1369682] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/07/2019] [Accepted: 09/10/2019] [Indexed: 01/07/2023]
Abstract
Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) provides acquired immunity in microorganisms against exogenous DNA that may hinder the survival of the organism. Pioneering work by Doudna and Charpentier in 2012 resulted in the creation of the CRISPR/Cas9 genome editing tool on the basis of this concept. The aim of this was to create a rapid, efficient, and versatile genome-editing tool to facilitate genetic manipulation. The mechanism relies on two components: the RNA guide which acts as a sentinel and a Cas protein complex which functions as a highly precise molecular knife. The guide RNA can be modified to match a DNA sequence of interest in the cell and accordingly be used to rectify mutations that may otherwise cause disease. Within a few years following the development of the CRISPR/Cas9 tool, its usage has become ubiquitous. Its influence extends into many fields of biological sciences from biotechnology and biochemistry to molecular biology and biomedical sciences. The following review aims at shedding some light on to the applications of the CRISPR/Cas9 tool in the field of biomedical sciences, particularly gene therapy. An insight with relation to a few of the many diseases that are being tackled with the aid of the CRISPR/Cas9 mechanism and the trends, successes, and challenges of this application as a gene therapy are discussed in this review.
Collapse
Affiliation(s)
- Odatha W. Kotagama
- Department of Chemistry, Faculty of Natural Sciences, The Open University of Sri Lanka, Nawala, Nugegoda, Sri Lanka
| | - Chanika D. Jayasinghe
- Department of Zoology, Faculty of Natural Sciences, The Open University of Sri Lanka, Nawala, Nugegoda, Sri Lanka
| | - Thelma Abeysinghe
- Department of Chemistry, Faculty of Natural Sciences, The Open University of Sri Lanka, Nawala, Nugegoda, Sri Lanka
| |
Collapse
|
18
|
Li Y, Kong Q, Yue J, Gou X, Xu M, Wu X. Genome-edited skin epidermal stem cells protect mice from cocaine-seeking behaviour and cocaine overdose. Nat Biomed Eng 2019; 3:105-113. [PMID: 30899600 PMCID: PMC6423967 DOI: 10.1038/s41551-018-0293-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 08/15/2018] [Indexed: 12/20/2022]
Abstract
Cocaine addiction is associated with compulsive drug-seeking, and exposure to the drug or to drug-associated cues leads to relapse, even after long periods of abstention. A variety of pharmacological targets and behavioral interventions have been explored to counteract cocaine addiction, but to date no market-approved medications for treating cocaine addiction or relapse exist, and effective interventions for acute emergencies resulting from cocaine overdose are lacking. We recently demonstrated that skin epidermal stem cells can be readily edited by using CRISPR (clustered regularly interspaced short palindromic repeats) and then transplanted back into the donor mice. Here, we show that the transplantation, into mice, of skin cells modified to express an enhanced form of butyrylcholinesterase, an enzyme that hydrolyzes cocaine, enables the long-term release of the enzyme and efficiently protects the mice from cocaine-seeking behavior and cocaine overdose. Cutaneous gene therapy through skin transplants that elicit drug elimination may offer a therapeutic option to address drug abuse.
Collapse
Affiliation(s)
- Yuanyuan Li
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Qingyao Kong
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL, USA
| | - Jiping Yue
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Xuewen Gou
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Ming Xu
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL, USA.
| | - Xiaoyang Wu
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
19
|
|
20
|
|
21
|
Uppal S, Italiya KS, Chitkara D, Mittal A. Nanoparticulate-based drug delivery systems for small molecule anti-diabetic drugs: An emerging paradigm for effective therapy. Acta Biomater 2018; 81:20-42. [PMID: 30268916 DOI: 10.1016/j.actbio.2018.09.049] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/14/2018] [Accepted: 09/26/2018] [Indexed: 12/11/2022]
Abstract
Emergence of nanoparticulate drug delivery systems in diabetes has facilitated improved delivery of small molecule drugs which could dramatically improve the quality of life for diabetics. Conventional dosage forms of the anti-diabetic drugs exhibit variable/less bioavailability and short half-life, demanding frequent dosing and causing increased side-effects resulting in ineffectiveness of therapy and non-compliance with the patients. Considering the chronic nature of diabetes, nanotechnology-based approaches are more promising in terms of providing site-specific delivery of drugs with higher bioavailability and reduced dosage regimen. Nanomedicines act at the cellular and molecular levels to enhance the uptake of the drug into the cells or block the efflux mechanisms thus retaining the drug inside the cell for a longer duration of time. Many studies have hinted at the possibility of administering peptide drugs like glucagon like peptides orally by encapsulation into nanoparticles. Nanoparticles also allow further modifications including their encapsulation into microparticles, polyethylene glycol (PEG)-PEGylation- or functionalization with ligands for active targeting. Nevertheless, such remarkable benefits are fraught with their long-term safety concerns, regulatory hurdles, limitations of scale-up and ineffective patent protection which have hindered their commercialization. This review summarizes the latest advances in the area of nanoformulations as applied to the delivery of anti-diabetics. STATEMENT OF SIGNIFICANCE: The present work describes the latest advancements in the area of nanoformulations for anti-diabetic therapy along with highlighting the advantages that these nanoformulations offer at molecular level for diabetes. Although several potent orally active anti-hyperglycemic agents are available, the current challenges in efficient management of diabetes include optimization of the present therapies to ensure an optimum and stable level of glucose, and also to reduce the occurrence of long term complications associated with diabetes. Nanoformulations because of their high surface area to volume ratio provide improved efficacy, targeting their delivery to the desired site of action tends to minimize adverse effects and administration of peptide drugs by oral route is also possible by encapsulating them in nanoparticles. As we reflect on the success and failures of latest research on nanoformulations for the treatment of diabetes, it is important not to dwell on lack of FDA approvals but rather define future directions that guarantee more effective anti-diabetic treatment. In proposed review we have explored the latest advancement in anti-diabetic nanotechnology based formulations.
Collapse
Affiliation(s)
- Siddharth Uppal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS-PILANI), Pilani, Rajasthan 333031, India
| | - Kishan S Italiya
- Department of Pharmacy, Birla Institute of Technology and Science (BITS-PILANI), Pilani, Rajasthan 333031, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science (BITS-PILANI), Pilani, Rajasthan 333031, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS-PILANI), Pilani, Rajasthan 333031, India.
| |
Collapse
|
22
|
|
23
|
Tjin MS, Chua AWC, Moreno-Moral A, Chong LY, Tang PY, Harmston NP, Cai Z, Petretto E, Tan BK, Tryggvason K. Biologically relevant laminin as chemically defined and fully human platform for human epidermal keratinocyte culture. Nat Commun 2018; 9:4432. [PMID: 30377295 PMCID: PMC6207750 DOI: 10.1038/s41467-018-06934-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 09/27/2018] [Indexed: 12/18/2022] Open
Abstract
The current expansion of autologous human keratinocytes to resurface severe wound defects still relies on murine feeder layer and calf serum in the cell culture system. Through our characterization efforts of the human skin basement membrane and murine feeder layer 3T3-J2, we identified two biologically relevant recombinant laminins-LN-511 and LN-421- as potential candidates to replace the murine feeder. Herein, we report a completely xeno-free and defined culture system utilizing these laminins which enables robust expansion of adult human skin keratinocytes. We demonstrate that our laminin system is comparable to the 3T3-J2 co-culture system in terms of basal markers' profile, colony-forming efficiency and the ability to form normal stratified epidermal structure in both in vitro and in vivo models. These results show that the proposed system may not only provide safer keratinocyte use in the clinics, but also facilitate the broader use of other cultured human epithelial cells in regenerative medicine.
Collapse
Affiliation(s)
- Monica Suryana Tjin
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Alvin Wen Choong Chua
- Department of Plastic Reconstructive & Aesthetic Surgery, Singapore General Hospital, 20 College Road, Singapore, 169856, Singapore.
- Skin Bank Unit, Singapore General Hospital, Singapore, 169608, Singapore.
| | - Aida Moreno-Moral
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Li Yen Chong
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Po Yin Tang
- Department of Anatomical Pathology, Singapore General Hospital, 20 College Road, Singapore, 169856, Singapore
| | - Nathan Peter Harmston
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Zuhua Cai
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Enrico Petretto
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Bien Keem Tan
- Department of Plastic Reconstructive & Aesthetic Surgery, Singapore General Hospital, 20 College Road, Singapore, 169856, Singapore
- Skin Bank Unit, Singapore General Hospital, Singapore, 169608, Singapore
| | - Karl Tryggvason
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore.
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17771, Stockholm, Sweden.
| |
Collapse
|
24
|
Luan XR, Chen XL, Tang YX, Zhang JY, Gao X, Ke HP, Lin ZY, Zhang XN. CRISPR/Cas9-Mediated Treatment Ameliorates the Phenotype of the Epidermolytic Palmoplantar Keratoderma-like Mouse. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:220-228. [PMID: 30195761 PMCID: PMC6023945 DOI: 10.1016/j.omtn.2018.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 12/25/2022]
Abstract
CRISPR/Cas9 has been confirmed as a distinctly efficient, simple-to-configure, highly specific genome-editing tool that has been used to treat monogenetic disorders. Epidermolytic palmoplantar keratoderma (EPPK) is a common autosomal dominant keratin disease resulting from dominant-negative mutation of the KRT9 gene, and it has no effective therapy. We performed CRISPR/Cas9-mediated treatment on a knockin (KI) transgenic mouse model that carried a small indel heterozygous mutation of Krt9, c.434delAinsGGCT (p.Tyr144delinsTrpLeu), which caused a humanized EPPK-like phenotype. The mutation within exon 1 of Krt9 generated a novel protospacer adjacent motif site, TGG, for Cas9 recognition and cutting. By delivering lentivirus vectors (LVs) encoding single-guide RNAs (sgRNAs) and Cas9 that targeted Krt9 sequence into HeLa cells engineered to constitutively express wild-type and mutant keratin 9 (K9), we found the sgRNA was highly effective in reducing expression of the mutant K9 protein in vitro. We injected the LV into the fore-paws of adult KI-Krt9 mice three times every 8 days and found that the expression of K9 decreased ∼14.6%. The phenotypic mitigation was revealed by restoration of the abnormal differentiation and aberrant proliferation of the epidermis. Our data are the first to show that CRISPR/Cas9 is a potentially powerful therapeutic option for EPPK and other PPK subtypes.
Collapse
Affiliation(s)
- Xiao-Rui Luan
- Department of Genetics, Research Center for Molecular Medicine, Institute of Cell Biology, Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Xiao-Ling Chen
- Department of Biological Chemistry, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yue-Xiao Tang
- Department of Genetics, Research Center for Molecular Medicine, Institute of Cell Biology, Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Jin-Yan Zhang
- Department of Genetics, Research Center for Molecular Medicine, Institute of Cell Biology, Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Xiang Gao
- Key Laboratory of Model Animals for Disease Study of The Ministry of Education, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu 210061, China
| | - Hai-Ping Ke
- Department of Biology, Ningbo College of Health Sciences, Ningbo, Zhejiang 315100, China
| | - Zhao-Yu Lin
- Key Laboratory of Model Animals for Disease Study of The Ministry of Education, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu 210061, China
| | - Xian-Ning Zhang
- Department of Genetics, Research Center for Molecular Medicine, Institute of Cell Biology, Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
25
|
Wu CAM, Roth TL, Baglaenko Y, Ferri DM, Brauer P, Zuniga-Pflucker JC, Rosbe KW, Wither JE, Marson A, Allen CDC. Genetic engineering in primary human B cells with CRISPR-Cas9 ribonucleoproteins. J Immunol Methods 2018; 457:33-40. [PMID: 29614266 DOI: 10.1016/j.jim.2018.03.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/24/2018] [Accepted: 03/26/2018] [Indexed: 12/27/2022]
Abstract
Genome editing in human cells with targeted nucleases now enables diverse experimental and therapeutic genome engineering applications, but extension to primary human B cells remains limited. Here we report a method for targeted genetic engineering in primary human B cells, utilizing electroporation of CRISPR-Cas9 ribonucleoproteins (RNPs) to introduce gene knockout mutations at protein-coding loci with high efficiencies that in some cases exceeded 80%. Further, we demonstrate knock-in editing of targeted nucleotides with efficiency exceeding 10% through co-delivery of oligonucleotide templates for homology directed repair. We delivered Cas9 RNPs in two distinct in vitro culture systems to achieve editing in both undifferentiated B cells and activated B cells undergoing differentiation, reflecting utility in diverse experimental conditions. In summary, we demonstrate a powerful and scalable research tool for functional genetic studies of human B cell biology that may have further applications in engineered B cell therapeutics.
Collapse
Affiliation(s)
- Chung-An M Wu
- Cardiovascular Research Institute, Sandler Asthma Basic Research Center, 555 Mission Bay Blvd S, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Theodore L Roth
- Department of Microbiology and Immunology, 513 Parnassus Ave, University of California, San Francisco, CA 94143, USA
| | - Yuriy Baglaenko
- Krembil Research Institute, 60 Leonard Ave, University Health Network, Toronto, Ontario, Canada; Department of Immunology, 60 Leonard Ave, University of Toronto, Toronto, Ontario, Canada
| | - Dario M Ferri
- Krembil Research Institute, 60 Leonard Ave, University Health Network, Toronto, Ontario, Canada; Department of Immunology, 60 Leonard Ave, University of Toronto, Toronto, Ontario, Canada
| | - Patrick Brauer
- Department of Immunology, 60 Leonard Ave, University of Toronto, Toronto, Ontario, Canada; Sunnybrook Research Institute, 2075 Bayview Ave, University of Toronto, Toronto, Ontario, Canada
| | - Juan Carlos Zuniga-Pflucker
- Department of Immunology, 60 Leonard Ave, University of Toronto, Toronto, Ontario, Canada; Sunnybrook Research Institute, 2075 Bayview Ave, University of Toronto, Toronto, Ontario, Canada
| | - Kristina W Rosbe
- Department of Otolaryngology, 550 16th St, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joan E Wither
- Krembil Research Institute, 60 Leonard Ave, University Health Network, Toronto, Ontario, Canada; Department of Immunology, 60 Leonard Ave, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, 60 Leonard Ave, University of Toronto, Toronto, Ontario, Canada.
| | - Alexander Marson
- Department of Microbiology and Immunology, 513 Parnassus Ave, University of California, San Francisco, CA 94143, USA; Department of Medicine, Diabetes Center, Helen Diller Family Comprehensive Cancer Center, 513 Parnassus Ave, University of California, San Francisco, CA 94143, USA; Innovative Genomics Institute, 2151 Berkeley Way, University of California, Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, 499 Illinois St, San Francisco, CA 94158, USA.
| | - Christopher D C Allen
- Cardiovascular Research Institute, Sandler Asthma Basic Research Center, 555 Mission Bay Blvd S, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Anatomy, 555 Mission Bay Blvd S, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
26
|
Böttcher A, Lickert H, Tschöp M. Engineering Skin with Skinny Genes. Cell Stem Cell 2017; 21:153-155. [PMID: 28777938 DOI: 10.1016/j.stem.2017.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The increasing prevalence of obesity and diabetes is an urgent worldwide health challenge. Now in Cell Stem Cell, Yue et al. (2017) report a proof-of-concept study using genetically engineered skin transplants that produce the incretin GLP-1 to prevent diet-induced obesity, suggesting a powerful approach for treating metabolic disorders.
Collapse
Affiliation(s)
- Anika Böttcher
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Am Parkring 11, 85748 Garching, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Am Parkring 11, 85748 Garching, Germany; Technische Universität München, Ismaninger Strasse 22, 81675 München, Germany.
| | - Matthias Tschöp
- Institute of Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Am Parkring 13, 85748 Garching, Germany; Division of Metabolic Diseases, Technische Universität München, Ismaninger Strasse 22, 81675 München, Germany.
| |
Collapse
|