1
|
Zhang W, Li S, Wang Y, Liu S, Liu L, Deng Z, Mo S, Chen M, Li Z, Wang R, Zhou X, Xu L, Yu L, Liu Z, Li H, Liang J, Wang C. Arginine-Rich Peptides Regulate the Pathogenic Galectin-10 Crystallization and Mitigate Crystallopathy-Associated Inflammation. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 39894983 DOI: 10.1021/acsami.4c18411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Protein self-assembly into a crystal in vivo triggers acute or chronic organ injury that can lead to intractable diseases lacking specific treatment options. In this study, we report the discovery of ionic arginine-rich peptides to disrupt the pathogenic galectin-10 (gal-10) crystallization, where the aberrant deposition of gal-10 crystals in airways causes the activation of IL-1β-dependent inflammation and the stimulation of epithelial cells to produce TNF-α. Gal-10 crystals show susceptibility to pH changes and charged residue substitutions at the protein packing interfaces, manifesting the role of charge-charge attractions across protein-protein interaction interfaces in governing gal-10 crystallization. To dissolve the gal-10 crystal, the ionic peptides R9 and R12Y8 were identified to eliminate the interprotein charge-charge interactions. The efficacy of R12Y8 in mitigating the gal-10 crystallopathy in vivo was assessed in a crystal-induced lung inflammation mice model. The mice intratracheally administrated by R12Y8 exhibited a downregulated release of proinflammatory cytokines and reduced infiltration of inflammatory cells in the lungs. Our study demonstrates that the pathogenic gal-10 crystallization is readily eliminated by R-rich peptides, which may display translational potentials for the treatment of gal-10 crystallopathy.
Collapse
Affiliation(s)
- Wenbo Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, P. R. China
| | - Shuyuan Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, P. R. China
| | - Yang Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, P. R. China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Shuli Liu
- Department of Clinical Laboratory, Peking University Civil Aviation School of Clinical Medicine, Beijing 100123, P. R. China
| | - Lei Liu
- Department of Cardiology, Boston Children's Hospital, 320 Longwood Avenue, Boston, Massachusetts 02115, United States
- Department of Pediatrics, Harvard Medical School, 320 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Zhun Deng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, P. R. China
| | - Shanshan Mo
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, P. R. China
| | - Mingrui Chen
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, P. R. China
| | - Zhenyan Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, P. R. China
| | - Ruonan Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, P. R. China
| | - Xin Zhou
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, P. R. China
| | - Longxin Xu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, P. R. China
| | - Lanlan Yu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, P. R. China
| | - Zhenlin Liu
- Department of Medical Engineering, Peking University Third Hospital, Beijing 100191, P. R. China
| | - Hongwei Li
- Beijing Nuclear Magnetic Resonance Center, Peking University, Beijing 100871, P. R. China
| | - Junbo Liang
- Center for Bioinformatics, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, P. R. China
| | - Chenxuan Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, P. R. China
| |
Collapse
|
2
|
Hao M, Zhang L, Chen P. Membrane Internalization Mechanisms and Design Strategies of Arginine-Rich Cell-Penetrating Peptides. Int J Mol Sci 2022; 23:ijms23169038. [PMID: 36012300 PMCID: PMC9409441 DOI: 10.3390/ijms23169038] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 11/29/2022] Open
Abstract
Cell-penetrating peptides (CPPs) have been discovered to deliver chemical drugs, nucleic acids, and macromolecules to permeate cell membranes, creating a novel route for exogenous substances to enter cells. Up until now, various sequence structures and fundamental action mechanisms of CPPs have been established. Among them, arginine-rich peptides with unique cell penetration properties have attracted substantial scientific attention. Due to the positively charged essential amino acids of the arginine-rich peptides, they can interact with negatively charged drug molecules and cell membranes through non-covalent interaction, including electrostatic interactions. Significantly, the sequence design and the penetrating mechanisms are critical. In this brief synopsis, we summarize the transmembrane processes and mechanisms of arginine-rich peptides; and outline the relationship between the function of arginine-rich peptides and the number of arginine residues, arginine optical isomers, primary sequence, secondary and ternary structures, etc. Taking advantage of the penetration ability, biomedical applications of arginine-rich peptides have been refreshed, including drug/RNA delivery systems, biosensors, and blood-brain barrier (BBB) penetration. Understanding the membrane internalization mechanisms and design strategies of CPPs will expand their potential applications in clinical trials.
Collapse
Affiliation(s)
- Minglu Hao
- Advanced Materials Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Lei Zhang
- Department of Chemical Engineering, Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L3G1, Canada
- Correspondence: (L.Z.); (P.C.)
| | - Pu Chen
- Advanced Materials Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Department of Chemical Engineering, Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L3G1, Canada
- Correspondence: (L.Z.); (P.C.)
| |
Collapse
|
3
|
Sadeghian I, Heidari R, Raee MJ, Negahdaripour M. Cell-penetrating peptide-mediated delivery of therapeutic peptides/proteins to manage the diseases involving oxidative stress, inflammatory response and apoptosis. J Pharm Pharmacol 2022; 74:1085-1116. [PMID: 35728949 DOI: 10.1093/jpp/rgac038] [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: 12/10/2021] [Accepted: 05/22/2022] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Peptides and proteins represent great potential for modulating various cellular processes including oxidative stress, inflammatory response, apoptosis and consequently the treatment of related diseases. However, their therapeutic effects are limited by their inability to cross cellular barriers. Cell-penetrating peptides (CPPs), which can transport cargoes into the cell, could resolve this issue, as would be discussed in this review. KEY FINDINGS CPPs have been successfully exploited in vitro and in vivo for peptide/protein delivery to treat a wide range of diseases involving oxidative stress, inflammatory processes and apoptosis. Their in vivo applications are still limited due to some fundamental issues of CPPs, including nonspecificity, proteolytic instability, potential toxicity and immunogenicity. SUMMARY Totally, CPPs could potentially help to manage the diseases involving oxidative stress, inflammatory response and apoptosis by delivering peptides/proteins that could selectively reach proper intracellular targets. More studies to overcome related CPP limitations and confirm the efficacy and safety of this strategy are needed before their clinical usage.
Collapse
Affiliation(s)
- Issa Sadeghian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Biotechnology Incubator, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Javad Raee
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manica Negahdaripour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
4
|
Dong G, Zhao X, Guo J, Ma L, Zhou H, Liu Q, Zhao X, Wang C, Wu K. Functional expression and purification of recombinant full-length human ATG7 protein with HIV-1 Tat peptide in Escherichia coli. Protein Expr Purif 2021; 182:105844. [PMID: 33592251 DOI: 10.1016/j.pep.2021.105844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/18/2021] [Accepted: 02/09/2021] [Indexed: 01/07/2023]
Abstract
The human autophagy-related protein ATG7 (hATG7), an E1-like ubiquitin enzyme, activates two ubiquitin-like proteins, LC3 (Atg8) and Atg12, and promotes autophagosome formation. While hATG7 plays an essential role for the autophagy conjugation system, the production of full-length functional hATG7 in bacterial systems remains challenging. Previous studies have demonstrated that the HIV-1 virus-encoded Tat peptide ('GRKKRRQRRR') can increase the yield and solubility of heterologous proteins. Here, functional full-length hATG7 was expressed using the pET28b-Tat expression vector in the Escherichia coli BL21 (DE3) strain. Recombinant hATG7 protein aggregated as inclusion bodies while expressed with widely used prokaryotic expression plasmids. In contrast, the solubility of Tat-tagged hATG7 increased significantly with prolonged time compared to Tat-free hATG7. The recombinant proteins were purified to >90% homogeneity under native conditions with a single step of affinity chromatography purification. The results of in vitro pull-down and LC3B-I lipidation assays showed that Tat-tagged hATG7 directly interacted with LC3B-I and promoted LC3B-I lipidation, suggesting that Tat-tagged hATG7 has significant catalytic activity. Overall, this study provides a novel method for improving the functional expression of full-length hATG7 in bacterial systems by fusion with the Tat peptide, a process which may be applied in future studies of hATG7 structure and function.
Collapse
Affiliation(s)
- Guofu Dong
- Beijing Key Laboratory of Radiation Biology (No. BZ0325) and Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xi Zhao
- Beijing Key Laboratory of Radiation Biology (No. BZ0325) and Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Junwang Guo
- Beijing Key Laboratory of Radiation Biology (No. BZ0325) and Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Lei Ma
- Beijing Key Laboratory of Radiation Biology (No. BZ0325) and Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Hongmei Zhou
- Beijing Key Laboratory of Radiation Biology (No. BZ0325) and Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Qi Liu
- Beijing Key Laboratory of Radiation Biology (No. BZ0325) and Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xuelong Zhao
- Beijing Key Laboratory of Radiation Biology (No. BZ0325) and Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Changzhen Wang
- Beijing Key Laboratory of Radiation Biology (No. BZ0325) and Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Ke Wu
- Beijing Key Laboratory of Radiation Biology (No. BZ0325) and Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
5
|
Vedadghavami A, Zhang C, Bajpayee AG. Overcoming negatively charged tissue barriers: Drug delivery using cationic peptides and proteins. NANO TODAY 2020; 34:100898. [PMID: 32802145 PMCID: PMC7425807 DOI: 10.1016/j.nantod.2020.100898] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Negatively charged tissues are ubiquitous in the human body and are associated with a number of common diseases yet remain an outstanding challenge for targeted drug delivery. While the anionic proteoglycans are critical for tissue structure and function, they make tissue matrix dense, conferring a high negative fixed charge density (FCD) that makes drug penetration through the tissue deep zones and drug delivery to resident cells extremely challenging. The high negative FCD of these tissues is now being utilized by taking advantage of electrostatic interactions to create positively charged multi-stage delivery methods that can sequentially penetrate through the full thickness of tissues, create a drug depot and target cells. After decades of work on attempting delivery using strong binding interactions, significant advances have recently been made using weak and reversible electrostatic interactions, a characteristic now considered essential to drug penetration and retention in negatively charged tissues. Here we discuss these advances using examples of negatively charged tissues (cartilage, meniscus, tendons and ligaments, nucleus pulposus, vitreous of eye, mucin, skin), and delve into how each of their structures, tissue matrix compositions and high negative FCDs create barriers to drug entry and explore how charge interactions are being used to overcome these barriers. We review work on tissue targeting cationic peptide and protein-based drug delivery, compare and contrast drug delivery designs, and also present examples of technologies that are entering clinical trials. We also present strategies on further enhancing drug retention within diseased tissues of lower FCD by using synergistic effects of short-range binding interactions like hydrophobic and H-bonds that stabilize long-range charge interactions. As electrostatic interactions are incorporated into design of drug delivery materials and used as a strategy to create properties that are reversible, tunable and dynamic, bio-electroceuticals are becoming an exciting new direction of research and clinical work.
Collapse
Affiliation(s)
- Armin Vedadghavami
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Chenzhen Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Ambika G. Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
- Department of Mechanical Engineering, Northeastern University, Boston, MA, 02115, USA
| |
Collapse
|
6
|
Li B, Qi P, Qu Y, Wang B, Chen J, Chang Z. Effects of dechlorane plus on oxidative stress, inflammatory response, and cell apoptosis in Cyprinus carpio. Drug Chem Toxicol 2019; 45:378-386. [PMID: 31826665 DOI: 10.1080/01480545.2019.1701001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The levels of the chlorinated organic compound Dechlorane Plus (DP) are increasing in aquatic ecosystems. To investigate the adverse effects of DP on aquatic animals, common carp (Cyprinus carpio) were subjected to three different DP concentrations (30 μg L-1, 60 μg L-1, and 120 μg L-1) for 1 d, 15 d, and 30 d. Histology and the hepatic and cerebral expression levels of several key antioxidant, detoxification, and apoptotic factors were then examined. Histopathological inspections showed that the liver and brain were severely damaged in carp exposed to 60 μg L-1 and 120 μg L-1 DP. Relative to the controls, the superoxide dismutase and glutathione activity levels and the malondialdehyde content were also changed in livers and brains exposed to DP. Besides, significant alterations in the expression levels of the inflammatory cytokines IL-1β, IL-6, and IL-10 were observed in the livers of carp subjected to DP. Relative to the control, the brains of DP-exposed carp presented with significantly upregulated IL-1β and IL-6 in carp treated with 120 μg L-1 DP for 30 d. The transcription levels of hepatic cyp2b4, cyp1b1, and cyp3a138 were all increased compared with the untreated at all DP exposure concentrations. The aforementioned results suggest that DP exposure perturbs fish metabolism and causes liver injury by inhibiting antioxidant enzyme activity, increasing lipid peroxidation, promoting inflammation, and inducing cell apoptosis. This information and the analytical methodology used to acquire it may form the basis for future ecological risk assessments on DP and related xenobiotics in aquatic animals.
Collapse
Affiliation(s)
- Baohua Li
- College of Life Science, Henan Normal University, Xinxiang, PR China.,College of Fisheries, Henan Normal University, Xinxiang, PR China
| | - Pengju Qi
- College of Life Science, Henan Normal University, Xinxiang, PR China
| | - Ying Qu
- College of Life Science, Henan Normal University, Xinxiang, PR China
| | - Beibei Wang
- College of Life Science, Henan Normal University, Xinxiang, PR China
| | - Jianjun Chen
- College of Life Science, Henan Normal University, Xinxiang, PR China
| | - Zhongjie Chang
- College of Life Science, Henan Normal University, Xinxiang, PR China
| |
Collapse
|
7
|
Role of Catalase in Oxidative Stress- and Age-Associated Degenerative Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9613090. [PMID: 31827713 PMCID: PMC6885225 DOI: 10.1155/2019/9613090] [Citation(s) in RCA: 505] [Impact Index Per Article: 84.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/18/2019] [Accepted: 08/14/2019] [Indexed: 12/22/2022]
Abstract
Reactive species produced in the cell during normal cellular metabolism can chemically react with cellular biomolecules such as nucleic acids, proteins, and lipids, thereby causing their oxidative modifications leading to alterations in their compositions and potential damage to their cellular activities. Fortunately, cells have evolved several antioxidant defense mechanisms (as metabolites, vitamins, and enzymes) to neutralize or mitigate the harmful effect of reactive species and/or their byproducts. Any perturbation in the balance in the level of antioxidants and the reactive species results in a physiological condition called “oxidative stress.” A catalase is one of the crucial antioxidant enzymes that mitigates oxidative stress to a considerable extent by destroying cellular hydrogen peroxide to produce water and oxygen. Deficiency or malfunction of catalase is postulated to be related to the pathogenesis of many age-associated degenerative diseases like diabetes mellitus, hypertension, anemia, vitiligo, Alzheimer's disease, Parkinson's disease, bipolar disorder, cancer, and schizophrenia. Therefore, efforts are being undertaken in many laboratories to explore its use as a potential drug for the treatment of such diseases. This paper describes the direct and indirect involvement of deficiency and/or modification of catalase in the pathogenesis of some important diseases such as diabetes mellitus, Alzheimer's disease, Parkinson's disease, vitiligo, and acatalasemia. Details on the efforts exploring the potential treatment of these diseases using a catalase as a protein therapeutic agent have also been described.
Collapse
|
8
|
Chen X, Xu Y, Li X, Liao S. Fluorogenic approach to evaluating prodrug hydrolysis and stability in live cells. Bioorg Med Chem 2019; 27:851-858. [PMID: 30723042 DOI: 10.1016/j.bmc.2019.01.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/03/2019] [Accepted: 01/25/2019] [Indexed: 01/21/2023]
Abstract
Fluorescein diester which is conjugated with cell membrane permeable Arg9 peptide was proposed as probe for ester prodrug stability and drug release study in living cells. α-Amino protected d-Val and l-Ala which bear differently hindered side chains were used to afford model diesters of 5-maleimide-fluorescein. Such fluorescein diesters were further conjugated with a Cys containing cell membrane permeable Arg9 peptide via thiol-ene crosslink reaction. The resulted conjugates of fluorescein diester and Arg9 peptide were purified with HPLC and characterized with MALDI-TOF MS. Upon incubation with cultured cells, the fluorescein diesters were delivered into the cells, the following hydrolysis of fluorescein diesters and release of fluorescein inside living cells were observed by monitoring the fluorescence accumulation. Fluorescence microscopic imaging studies of HeLa cells treated with fluorescein l-Ala diester show strong fluorescence accumulation in 30 min indicating fast hydrolysis of fluorescein diester and fluorescein release; in contrast d-Val diester remains stable inside cells evidenced by margin fluorescence formation. Further flowcytometry studies on the fluorescein diester-Arg9 conjugate treated cells show that the hydrolysis t1/2 for l-Ala diester is 15 min. The results also show that Arg9 peptide not only transports the ester probes into cell efficiently but also can retain and concentrate hydrolytic product fluorescein inside cells so that the accumulated fluorescence can be accurately quantified. This fluorogenic probe approach provides feasible applications in dynamic studies on ester prodrug hydrolysis and release, facilitating screening and optimization of prodrug structures in living cell settings.
Collapse
Affiliation(s)
- Xin Chen
- ME Genomics Inc. Software Industry Base, Shenzhen 518061, PR China; Department of Chemical Engineering, Columbia University, 550 W 120th Street, 1001A, New York, NY 10027, USA
| | - Yunzhen Xu
- The First Clinical Medical School of Lanzhou University, Lanzhou 730000, PR China
| | - Xiaoxu Li
- Department of Chemical Engineering, Columbia University, 550 W 120th Street, 1001A, New York, NY 10027, USA.
| | - Shiqi Liao
- Laboratory of Molecular Biology, Institute of Gansu Medical Science Research, Lanzhou 730050, PR China.
| |
Collapse
|
9
|
Kiseleva RY, Glassman PM, Greineder CF, Hood ED, Shuvaev VV, Muzykantov VR. Targeting therapeutics to endothelium: are we there yet? Drug Deliv Transl Res 2018; 8:883-902. [PMID: 29282646 DOI: 10.1007/s13346-017-0464-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Vascular endothelial cells represent an important therapeutic target in many pathologies, including inflammation, oxidative stress, and thrombosis; however, delivery of drugs to this site is often limited by the lack of specific affinity of therapeutics for these cells. Selective delivery of both small molecule drugs and therapeutic proteins to the endothelium has been achieved through the use of targeting ligands, such as monoclonal antibodies, directed against endothelial cell surface markers, particularly cell adhesion molecules (CAMs). Careful selection of target molecules and targeting agents allows for precise delivery to sites of inflammation, thereby maximizing therapeutic drug concentrations at the site of injury. A good understanding of the physiological and pathological determinants of drug and drug carrier pharmacokinetics and biodistribution may allow for a priori identification of optimal properties of drug carrier and targeting agent. Targeted delivery of therapeutics such as antioxidants and antithrombotic agents to the injured endothelium has shown efficacy in preclinical models, suggesting the potential for translation into clinical practice. As with all therapeutics, demonstration of both efficacy and safety are required for successful clinical implementation, which must be considered not only for the individual components (drug, targeting agent, etc.) but also for the sum of the parts (e.g., the drug delivery system), as unexpected toxicities may arise with complex delivery systems. While the use of endothelial targeting has not been translated into the clinic to date, the preclinical results summarized here suggest that there is hope for successful implementation of these agents in the years to come.
Collapse
Affiliation(s)
- Raisa Yu Kiseleva
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA, 19104-5158, USA
| | - Patrick M Glassman
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA, 19104-5158, USA
| | - Colin F Greineder
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA, 19104-5158, USA
| | - Elizabeth D Hood
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA, 19104-5158, USA
| | - Vladimir V Shuvaev
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA, 19104-5158, USA
| | - Vladimir R Muzykantov
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA, 19104-5158, USA.
| |
Collapse
|
10
|
Khaliq H, Jing W, Ke X, Ke-Li Y, Peng-Peng S, Cui L, Wei-Wei Q, Zhixin L, Hua-Zhen L, Hui S, Ju-Ming Z, Ke-Mei P. Boron Affects the Development of the Kidney Through Modulation of Apoptosis, Antioxidant Capacity, and Nrf2 Pathway in the African Ostrich Chicks. Biol Trace Elem Res 2018. [PMID: 29536335 DOI: 10.1007/s12011-018-1280-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The nuclear-related factor 2 (Nrf2) pathway is the most important mechanism in antioxidant capacity, which regulates the cell's redox homeostasis. In addition, Nrf2 pathway also can inhibit cell apoptosis. The mechanism of boron actions on various organs is well documented. But, it is not known whether boron can also regulate the Nrf2 pathway in the kidneys. Therefore, in this research, the actions of boron on the kidneys of ostrich chicks, especially the antioxidant effects, have been studied. The ostrich chicks were divided into six groups and supplemented with boric acid (BA) (source of boron) in the drinking water (0, 40, 80, 160, 320, 640 mg respectively) to examine apoptotic, antioxidant, biochemical, and histochemical alterations induced by boron administration in the ostrich chick's kidney. The cellular apoptosis was assessed by terminal deoxynucleotidyl transferase dUTP Nick-End Labeling (TUNEL) assay. The relative antioxidant enzymes (T-AOC, MDA, GSH-Px, SOD, GR, CAT) and biochemical indices (ALT, AST, ALP, CK, LDH, BUN, CREA, UA) in the kidney were determined by spectrophotometric method. The expression of three important genes in the antioxidant pathway (Nrf2, HO-1, GCLc) was measured by quantitative real-time PCR (qPCR), and the localization of key regulator Nrf2 was examined by immunohistochemistry (IHC) method. Western blotting was also performed to further validate our results. Our results revealed that low doses of boron (up to 160 mg) had positive effect, while high doses (especially 640 mg) caused negative effect on the development of the kidney. The cellular apoptosis was in a biphasic manner by altering the boron quantities. The low doses regulate the oxidative and enzyme activity in the kidney. The IHC and western blot showed maximum localization of Nrf2 in 80 mg/L BA dose group. Furthermore, supplementation of boron at low doses upregulated the expression of genes involved in the antioxidant pathway. Taken together, the study demonstrated that low levels of boron (up to 160 mg) inhibited the cell apoptosis, regulate the enzyme activity, and improved the antioxidant system, thus may encourage the development of the ostrich chick's kidney, while a high amount of boron especially 640 mg/L promoted cell apoptosis and reduced the antioxidant capacity, thus caused negative effect to the ostrich chick's kidney.
Collapse
Affiliation(s)
- Haseeb Khaliq
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Wang Jing
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Xiao Ke
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Yang Ke-Li
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Sun Peng-Peng
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Lei Cui
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Qiu Wei-Wei
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Lei Zhixin
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Liu Hua-Zhen
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Song Hui
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Zhong Ju-Ming
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
- College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Peng Ke-Mei
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.
| |
Collapse
|
11
|
Lin Q, Dong L, Xu Y, Di G. Studies on effects of static electric field exposure on liver in mice. Sci Rep 2018; 8:15507. [PMID: 30341322 PMCID: PMC6195622 DOI: 10.1038/s41598-018-33447-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/26/2018] [Indexed: 02/08/2023] Open
Abstract
With the development of ultra-high-voltage direct-current transmission, the intensity of static electric field (SEF) under transmission lines increased, which has aroused public attention on its potential health effects. In order to examine effects of SEF exposure on liver, institute of cancer research mice were exposed to SEF with intensities of 27.5 kV/m, 34.7 kV/m and 56.3 kV/m, respectively. In each intensity of SEF exposure, a corresponding sham exposure group was used. Several indices relating to liver function (aspartate aminotransferase (AST) and alanine aminotransferase (ALT)) and oxidative stress (superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and malondialdehyde (MDA)) were tested after exposure of 7, 14, 21 and 35 days. Results showed that exposure to SEF with intensities of 27.5 kV/m and 34.7 kV/m for 35 days did not significantly influence any detected indices above. Under SEF exposure with intensity of 56.3 kV/m, the SOD activity in liver was significantly increased after exposure of 7 and 14 days. However, no significant increase was found in MDA content as well as the activities of AST and ALT between exposure group and sham exposure group during SEF exposure of 56.3 kV/m. It suggested that from three SEF intensities, only exposure to SEF with intensity of 56.3 kV/m (7 and 14 days) caused a temporary oxidative stress response in liver expressed by the increase in activity of SOD, but it did not produce oxidative damage. This biological effect may be related to the increase of mitochondrial membrane potential of hepatocytes caused by SEF exposure. When the membrane potential exceeds a threshold, Q cycle in mitochondria will be affected, which will result in an increase of superoxide anion concentration and ultimately an oxidative stress.
Collapse
Affiliation(s)
- Qinhao Lin
- Institute of Environmental Process, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Li Dong
- Institute of Environmental Process, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | | | - Guoqing Di
- Institute of Environmental Process, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, PR China.
| |
Collapse
|
12
|
Sadeghian I, Khalvati B, Ghasemi Y, Hemmati S. TAT-mediated intracellular delivery of carboxypeptidase G2 protects against methotrexate-induced cell death in HepG2 cells. Toxicol Appl Pharmacol 2018; 346:9-18. [PMID: 29574210 DOI: 10.1016/j.taap.2018.03.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/05/2018] [Accepted: 03/20/2018] [Indexed: 12/20/2022]
Abstract
Side effects of methotrexate (MTX) especially hepatotoxicity limits clinical applications of this anticancer agent. Carboxypeptidase G2 (CPG2) is administrated for the treatment of elevated plasma concentrations of MTX. In this study, we have investigated the intracellular delivery of CPG2 fused to the transactivator transduction domain (TAT) and its protective effects against MTX-induced cell death of HepG2 cells. We have observed that both native and denatured forms of the enzyme transduced into the HepG2 cells efficiently in a concentration and time-dependent manner. The denatured protein transduced with higher efficiency than the native form and was functional inside the cells. MTX exposure significantly decreased HepG2 cell viability in a dose- and time-dependent manner. The cell viability after 24 and 48 h of incubation with 100 μM MTX was reduced to 44.37% and 17.69%, respectively. In cells pretreated with native and denatured TAT-CPG2 protein the cell viability was 98.63% and 86.31% after 24 and 48 h, respectively. Treatment with MTX increased the number of apoptotic HepG2 cells to 90.23% after 48 h. However, the apoptosis percentage in cells pretreated with native and denatured TAT-CPG2 was 21.49% and 22.28%, respectively. Our results showed that TAT-CPG2 significantly prevents MTX-induced oxidative stress by decreasing the formation of ROS and increasing the content of glutathione (GSH) and catalase activity. Our finding indicates that both native and denatured TAT-CPG2 strongly protect HepG2 cells against MTX-induced oxidative stress and apoptosis. Hence, intracellular delivery of CPG2 might provide a new therapeutic strategy for protecting against MTX mediated cytotoxicity.
Collapse
Affiliation(s)
- Issa Sadeghian
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahman Khalvati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shiva Hemmati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
13
|
Chaulagain B, Jain A, Tiwari A, Verma A, Jain SK. Passive delivery of protein drugs through transdermal route. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:472-487. [PMID: 29378433 DOI: 10.1080/21691401.2018.1430695] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Skin is the largest external organ in the human body but its use for therapeutic purposes has been minimal. Stratum corneum residing on the uppermost layer of the skin provides a tough barrier to transport the drugs across the skin. Very small group of drugs sharing Lipinski properties, i.e. drugs having molecular weight not larger than 500 Da, having high lipophilicity and optimum polarity are fortunate enough to be used on skin therapeutics. But, at a time where modern therapeutics is slowly shifting from use of small molecular drugs towards the use of macromolecular therapeutic agents such as peptides, proteins and nucleotides in origin, skin therapeutics need to be evolved accordingly to cater the delivery of these agents. Physical technologies like iontophoresis, laser ablation, micro-needles and ultrasound, etc. have been introduced to enhance skin permeability. But their success is limited due to their complex working mechanisms and involvement of certain irreversible skin damage in some or other way. This review therefore explores the delivery strategies for transport of mainly peptide and protein drugs that do not involve any injuries (non-invasive) to the skin termed as passive delivery techniques. Chemical enhancers, nanocarriers, certain biological peptides and miscellaneous approaches like prodrugs are also thoroughly reviewed for their applications in protein delivery.
Collapse
Affiliation(s)
- Bivek Chaulagain
- a Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory , Dr. Hari Singh Gour Central University , Sagar , India
| | - Ankit Jain
- b Institute of Pharmaceutical Research, GLA University , Mathura , India
| | - Ankita Tiwari
- a Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory , Dr. Hari Singh Gour Central University , Sagar , India
| | - Amit Verma
- a Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory , Dr. Hari Singh Gour Central University , Sagar , India
| | - Sanjay K Jain
- a Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory , Dr. Hari Singh Gour Central University , Sagar , India
| |
Collapse
|
14
|
TAT-IL-24-KDEL-induced apoptosis is inhibited by survivin but restored by the small molecular survivin inhibitor, YM155, in cancer cells. Oncotarget 2018; 7:37030-37042. [PMID: 27203744 PMCID: PMC5095056 DOI: 10.18632/oncotarget.9458] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/19/2016] [Indexed: 11/25/2022] Open
Abstract
Interleukin-24 (IL-24) is a cytokine belonging to the IL-10 gene family. This cytokine selectively induces apoptosis in cancer cells, without harming normal cells, through a mechanism involving endoplasmic reticulum (ER) stress response. TAT-IL-24-KDEL is a fusion protein that efficiently enters the tumor cells and locates in the ER. Here we report that TAT-IL-24-KDEL induced apoptosis in human cancer cells, mediated by the ER stress cell death pathway. This process was accompanied by the inhibition of the transcription of an antiapoptotic protein, survivin. The forced expression of survivin partially protected cancer cells from the induction of apoptosis by TAT-IL-24-KDEL, increased their clonogenic survival, and attenuated TAT-IL-24-KDEL-induced activation of caspase-3/7. RNA interference of survivin markedly sensitized the transformed cells to TAT-IL-24-KDEL. Survivin was expressed at higher levels among isolated clones that resistant to TAT-IL-24-KDEL. These observations show the important role of survivin in attenuating cancer-specific apoptosis induced by TAT-IL-24-KDEL. The pharmacological inhibition of survivin expression by a selective small-molecule survivin suppressant YM155 synergistically sensitized cancer cells to TAT-IL-24-KDEL-induced apoptosis in vitro and in vivo. The combined regimen caused significantly higher activation of ER stress and dysfunction of mitochondria than either treatment alone. As survivin is overexpressed in a majority of cancers, the combined TAT-IL-24-KDEL and YM155 treatment provides a promising alternative to the existing therapies.
Collapse
|
15
|
Brenner JS, Kiseleva RY, Glassman PM, Parhiz H, Greineder CF, Hood ED, Shuvaev VV, Muzykantov VR. The new frontiers of the targeted interventions in the pulmonary vasculature: precision and safety (2017 Grover Conference Series). Pulm Circ 2017; 8:2045893217752329. [PMID: 29261028 PMCID: PMC5768280 DOI: 10.1177/2045893217752329] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The pulmonary vasculature plays an important role in many lung pathologies, such as pulmonary arterial hypertension, primary graft dysfunction of lung transplant, and acute respiratory distress syndrome. Therapy for these diseases is quite limited, largely due to dose-limiting side effects of numerous drugs that have been trialed or approved. High doses of drugs targeting the pulmonary vasculature are needed due to the lack of specific affinity of therapeutic compounds to the vasculature. To overcome this problem, the field of targeted drug delivery aims to target drugs to the pulmonary endothelial cells, especially those in pathological regions. The field uses a variety of drug delivery systems (DDSs), ranging from nano-scale drug carriers, such as liposomes, to methods of conjugating drugs to affinity moieites, such as antibodies. These DDSs can deliver small molecule drugs, protein therapeutics, and imaging agents. Here we review targeted drug delivery to the pulmonary endothelium for the treatment of pulmonary diseases. Cautionary notes are made of the risk–benefit ratio and safety—parameters one should keep in mind when developing a translational therapeutic.
Collapse
Affiliation(s)
- Jacob S Brenner
- 1 14640 Pulmonary, Allergy, & Critical Care Division, University of Pennsylvania, Philadelphia, PA, USA
| | - Raisa Yu Kiseleva
- 2 14640 Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick M Glassman
- 2 14640 Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hamideh Parhiz
- 2 14640 Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Colin F Greineder
- 2 14640 Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth D Hood
- 2 14640 Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vladimir V Shuvaev
- 2 14640 Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vladimir R Muzykantov
- 2 14640 Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
16
|
Chen J, Li H, Chen J. Human epidermal growth factor coupled to different structural classes of cell penetrating peptides: A comparative study. Int J Biol Macromol 2017; 105:336-345. [DOI: 10.1016/j.ijbiomac.2017.07.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/30/2017] [Accepted: 07/06/2017] [Indexed: 12/25/2022]
|
17
|
Nordin F, Ahmad RNR, Farzaneh F. Transactivator protein: An alternative for delivery of recombinant proteins for safer reprogramming of induced Pluripotent Stem Cell. Virus Res 2017; 235:106-114. [DOI: 10.1016/j.virusres.2017.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/08/2017] [Indexed: 10/19/2022]
|
18
|
Srivastava S, Singh D, Patel S, Singh MR. Role of enzymatic free radical scavengers in management of oxidative stress in autoimmune disorders. Int J Biol Macromol 2017; 101:502-517. [PMID: 28342757 DOI: 10.1016/j.ijbiomac.2017.03.100] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/17/2017] [Accepted: 03/18/2017] [Indexed: 02/03/2023]
Abstract
Autoimmune disorders are distinct with over production and accumulation of free radicals due to its undisclosed genesis. The cause of numerous disorders as cancer, arthritis, psoriasis, diabetes, alzheimer's, cardiovascular disease, Parkinson's, respiratory distress syndrome, colitis, crohn's, pulmonary fibrosis, obesity and ageing have been associated with immune dysfunction and oxidative stress. In an oxidative stress, reactive oxygen species generally provoke the series of oxidation at cellular level. The buildup of free radicals in turn triggers various inflammatory cells causing release of various inflammatory interleukins, cytokines, chemokines, and tumor necrosis factors which mediate signal transduction and transcription pathways as nuclear factor- kappa B (NF-κB), signal transducer and activator of transcription 3 (STAT3), hypoxia-inducible factor-1 (HIF-1α) and nuclear factor-erythroid 2-related factor (Nrf2). The imbalance could only be combat by supplementing natural defensive antioxidant enzymes such as superoxide dismutase and catalase. The efficiency of these enzymes is enhanced by use of colloidal carriers which include cellular carriers, vesicular and particulate systems like erythrocytes, leukocytes, platelets, liposomes, transferosomes, solid lipid nanoparticles, microspheres, emulsions. Thus this review provides a platform for understanding importance of antioxidant enzymes and its therapeutic applications in treatment of various autoimmune disorders.
Collapse
Affiliation(s)
- Shikha Srivastava
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chattissgarh, 492010, India
| | - Deependra Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chattissgarh, 492010, India
| | - Satish Patel
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chattissgarh, 492010, India
| | - Manju R Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chattissgarh, 492010, India.
| |
Collapse
|
19
|
Topical Application of TAT-Superoxide Dismutase in Acupoints LI 20 on Allergic Rhinitis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2016:3830273. [PMID: 28119757 PMCID: PMC5227158 DOI: 10.1155/2016/3830273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 12/12/2016] [Indexed: 11/17/2022]
Abstract
Reactive oxygen species are products of cellular metabolism and assigned important roles in biomedical science as deleterious factors in pathologies. In fact, some studies have shown that the therapeutic benefits of taking antioxidants were limited and the potential for therapeutic intervention remains unclear. New evidences showed that ROS have some ability of intercellular transportation. For treating allergic rhinitis, as a novel intracellular superoxide quencher, TAT-SOD applied to acupoints LI 20 instead of directly to nasal cavity can be used to test that. TTA group apply TAT-SOD cream prepared by adding purified TAT-SOD to the vehicle cream to acupoints LI 20, while placebo group used the vehicle cream instead. TTN group applied the same TAT-SOD cream directly to nasal cavity three times daily. Symptom scores were recorded at baseline and days 8 and 15. For the overall efficacy rate, TTA group was 81.0%, while placebo group was 5.9% and TTN was 0%. Malondialdehyde levels decreased observably in TTA group, and superoxide dismutase, catalase, and glutathione peroxidase levels remained basically unaffected. Enzymatic scavenging of the intracellular superoxide at acupoints LI 20 proved to be effective in treating allergic rhinitis, while no improvement was observed with the placebo group and TTN group.
Collapse
|
20
|
Coyle CH, Mendralla S, Lanasa S, Kader KN. Endothelial Cell Seeding onto Various Biomaterials Causes Superoxide-induced Cell Death. J Biomater Appl 2016; 22:55-69. [PMID: 16920759 DOI: 10.1177/0885328206069152] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The seeding and/or in-growth of endothelial cells on a number of blood-contacting implants are a concern for both biomaterials and tissue engineering. While endothelialization has been viewed positively, owing to their ability to regulate both smooth muscle and blood, there is evidence which suggests that endothelial cells on a nonoptimized surface may be counterproductive. The present study describes the experimentation designed to elucidate the effect of culture substrate on intracellular superoxide (SO) levels, a marker for endothelial cell dysfunction. The adaptation of the use of dihydroethidium under physiologically relevant shearing conditions is also reported. The present study describes a standardized method for the use of dihydroethidium as a marker for intracellular oxidative stress under physiologic shear. Levels of hydrogen peroxide (oxidative stress producing agent) are optimized to a minimum of 60 μM (under static conditions) to allow for the detection of SO within the free radical scavenging environment. A flow rate of 24.4 mL/min is applied and found to produce physiologically relevant shear stress (8.2 dynes/cm2) within the system under study. Dihydroethidium is a useful marker for assessing intracellular oxidative stress in studies that require shear.
Collapse
Affiliation(s)
- Christian H Coyle
- Cell and Synthetic Interface Engineering Laboratory Department of Biomedical Engineering, University of Iowa Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
21
|
Cell penetrating peptides as an innovative approach for drug delivery; then, present and the future. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2016. [DOI: 10.1007/s40005-016-0253-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
22
|
Zhang J, Sun A, Xu R, Tao X, Dong Y, Lv X, Wei D. Cell-penetrating and endoplasmic reticulum-locating TAT-IL-24-KDEL fusion protein induces tumor apoptosis. J Cell Physiol 2016; 231:84-93. [PMID: 26031207 DOI: 10.1002/jcp.25054] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 05/20/2015] [Indexed: 01/27/2023]
Abstract
Interleukin-24 (IL-24) is a unique IL-10 family cytokine that could selectively induce apoptosis in cancer cells without harming normal cells. Previous research demonstrated that intracellular IL-24 protein induces an endoplasmic reticulum (ER) stress response only in cancer cells, culminating in apoptosis. In this study, we developed a novel recombinant fusion protein to penetrate into cancer cells and locate on ER. It is composed of three distinct functional domains, IL-24, and the targeting domain of transactivator of transcription (TAT) and an ER retention four-peptide sequence KDEL (Lys-Asp-Glu-Leu) that link at its NH2 and COOH terminal, respectively. The in vitro results indicated that TAT-IL-24-KDEL inhibited growth in bladder cancer cells, as well as in non-small cell lung cancer cell line and breast cancer cell line, but the normal human lung fibroblast cell line was not affected, indicating the cancer specificity of TAT-IL-24-KDEL. Western blot analysis showed that apoptosis activation was induced by TAT-IL-24-KDEL through the ER stress-mediated cell death pathway. Treatment with TAT-IL-24-KDEL significantly inhibited the growth of human H460 xenografts in nude mice, and the tumor growth inhibition was correlated with increased hematoxylin and eosin (H&E) staining and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining. These findings suggest that the artificially designed recombinant fusion protein TAT-IL-24-KDEL may be highly effective in cancer therapy and worthy of further evaluation and development.
Collapse
Affiliation(s)
- Jian Zhang
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Aiyou Sun
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Rui Xu
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Xinyi Tao
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Yuguo Dong
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Xinxin Lv
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Dongzhi Wei
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai, People's Republic of China
| |
Collapse
|
23
|
Xu C, Zhao H, Chen H, Yao Q. CXCR4 in breast cancer: oncogenic role and therapeutic targeting. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:4953-64. [PMID: 26356032 PMCID: PMC4560524 DOI: 10.2147/dddt.s84932] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Chemokines are 8–12 kDa peptides that function as chemoattractant cytokines and are involved in cell activation, differentiation, and trafficking. Chemokines bind to specific G-protein-coupled seven-span transmembrane receptors. Chemokines play a fundamental role in the regulation of a variety of cellular, physiological, and developmental processes. Their aberrant expression can lead to a variety of human diseases including cancer. C-X-C chemokine receptor type 4 (CXCR4), also known as fusin or CD184, is an alpha-chemokine receptor specific for stromal-derived-factor-1 (SDF-1 also called CXCL12). CXCR4 belongs to the superfamily of the seven transmembrane domain heterotrimeric G protein-coupled receptors and is functionally expressed on the cell surface of various types of cancer cells. CXCR4 also plays a role in the cell proliferation and migration of these cells. Recently, CXCR4 has been reported to play an important role in cell survival, proliferation, migration, as well as metastasis of several cancers including breast cancer. This review is mainly focused on the current knowledge of the oncogenic role and potential drugs that target CXCR4 in breast cancer. Additionally, CXCR4 proangiogenic molecular mechanisms will be reviewed. Strict biunivocal binding affinity and activation of CXCR4/CXCL12 complex make CXCR4 a unique molecular target for prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Chao Xu
- First Clinical College of Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Hong Zhao
- First Clinical College of Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Haitao Chen
- First Clinical College of Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Qinghua Yao
- Department of Integrated Traditional Chinese and Western Medicine, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China ; Key Laboratory of Integrated Traditional Chinese and Western Medicine, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| |
Collapse
|
24
|
Abstract
Nearly 30years ago, certain small, relatively nontoxic peptides were discovered to be capable of traversing the cell membrane. These cell-penetrating peptides, as they are now called, have been shown to not only be capable of crossing the cell membrane themselves but can also carry many different therapeutic agents into cells, including small molecules, plasmid DNA, siRNA, therapeutic proteins, viruses, imaging agents, and other various nanoparticles. Many cell-penetrating peptides have been derived from natural proteins, but several other cell-penetrating peptides have been developed that are either chimeric or completely synthetic. How cell-penetrating peptides are internalized into cells has been a topic of debate, with some peptides seemingly entering cells through an endocytic mechanism and others by directly penetrating the cell membrane. Although the entry mechanism is still not entirely understood, it seems to be dependent on the peptide type, the peptide concentration, the cargo the peptide transports, and the cell type tested. With new intracellular disease targets being discovered, cell-penetrating peptides offer an exciting approach for delivering drugs to these intracellular targets. There are hundreds of cell-penetrating peptides being studied for drug delivery, and ongoing studies are demonstrating their success both in vitro and in vivo.
Collapse
Affiliation(s)
- Joshua D Ramsey
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK 74078, United States.
| | - Nicholas H Flynn
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK 74078, United States
| |
Collapse
|
25
|
Ren Y, Li X, Liu Q, Deng Y, Shi D. An improved Tet-on system to tightly conditionally regulate reporter gene expression. BIOTECHNOL BIOPROC E 2015. [DOI: 10.1007/s12257-014-0422-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
26
|
Wu Y, Ng DYW, Kuan SL, Weil T. Protein–polymer therapeutics: a macromolecular perspective. Biomater Sci 2015. [DOI: 10.1039/c4bm00270a] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The development of protein–polymer hybrids emerged several decades ago with the vision that their synergistic combination will offer macromolecular hybrids with manifold features to succeed as the next generation therapeutics.
Collapse
Affiliation(s)
- Yuzhou Wu
- Institute of Organic Chemistry III
- Macromolecular Chemistry
- Albert-Einstein-Allee 11
- 89081 Ulm
- Germany
| | - David Y. W. Ng
- Institute of Organic Chemistry III
- Macromolecular Chemistry
- Albert-Einstein-Allee 11
- 89081 Ulm
- Germany
| | - Seah Ling Kuan
- Institute of Organic Chemistry III
- Macromolecular Chemistry
- Albert-Einstein-Allee 11
- 89081 Ulm
- Germany
| | - Tanja Weil
- Institute of Organic Chemistry III
- Macromolecular Chemistry
- Albert-Einstein-Allee 11
- 89081 Ulm
- Germany
| |
Collapse
|
27
|
Daundkar PS, Rampal S. Evaluation of ameliorative potential of selenium on carbendazim induced oxidative stress in male goats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 38:711-719. [PMID: 25299847 DOI: 10.1016/j.etap.2014.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 08/26/2014] [Accepted: 09/11/2014] [Indexed: 06/04/2023]
Abstract
In the present investigation, ameliorative effect of selenium on carbendazim induced oral sub chronic toxicity in bucks was assessed by studying various indices of antioxidant defense system. Bucks were randomly divided into four groups of four animals each. Group I served as control, Group II was orally drenched carbendazim at the dose rate of 50mg/kg body weight for 90 consecutive days. Group III was orally administered selenium in the form of sodium selenite at the dose rate of 0.05mg/kg body weight for 90 consecutive days. Group IV was orally administered carbendazim along with selenium at the same dosages as Group II and III. Prolonged administration of carbendazim produced oxidative stress in goat bucks as evidenced by increase in lipid peroxidation and decline in total antioxidant capacity. The increase in the activity of antioxidant enzymes was not sufficient to prevent pesticide induced oxidative stress. Selenium supplementation provides some amelioration against this effect. Further study is needed to prove ameliorative potential of this antioxidant against carbendazim induced toxicity in goat bucks.
Collapse
Affiliation(s)
- Prashant S Daundkar
- Department of Pharmacology and Toxicology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India.
| | - S Rampal
- Department of Pharmacology and Toxicology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| |
Collapse
|
28
|
Eiamphungporn W, Yainoy S, Prachayasittikul V. Angiopep-2-Mediated Delivery of Human Manganese Superoxide Dismutase in Brain Endothelial Cells and its Protective Effect Against Oxidative Stress. Int J Pept Res Ther 2014. [DOI: 10.1007/s10989-014-9433-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
29
|
Liu J, Gaj T, Patterson JT, Sirk SJ, Barbas CF. Cell-penetrating peptide-mediated delivery of TALEN proteins via bioconjugation for genome engineering. PLoS One 2014; 9:e85755. [PMID: 24465685 PMCID: PMC3896395 DOI: 10.1371/journal.pone.0085755] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 12/02/2013] [Indexed: 12/21/2022] Open
Abstract
Transcription activator-like (TAL) effector nucleases (TALENs) have enabled the introduction of targeted genetic alterations into a broad range of cell lines and organisms. These customizable nucleases are comprised of programmable sequence-specific DNA-binding modules derived from TAL effector proteins fused to the non-specific FokI cleavage domain. Delivery of these nucleases into cells has proven challenging as the large size and highly repetitive nature of the TAL effector DNA-binding domain precludes their incorporation into many types of viral vectors. Furthermore, viral and non-viral gene delivery methods carry the risk of insertional mutagenesis and have been shown to increase the off-target activity of site-specific nucleases. We previously demonstrated that direct delivery of zinc-finger nuclease proteins enables highly efficient gene knockout in a variety of mammalian cell types with reduced off-target effects. Here we show that conjugation of cell-penetrating poly-Arg peptides to a surface-exposed Cys residue present on each TAL effector repeat imparted cell-penetrating activity to purified TALEN proteins. These modifications are reversible under reducing conditions and enabled TALEN-mediated gene knockout of the human CCR5 and BMPR1A genes at rates comparable to those achieved with transient transfection of TALEN expression vectors. These findings demonstrate that direct protein delivery, facilitated by conjugation of chemical functionalities onto the TALEN protein surface, is a promising alternative to current non-viral and viral-based methods for TALEN delivery into mammalian cells.
Collapse
Affiliation(s)
- Jia Liu
- The Departments of Chemistry and Cell and Molecular Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Thomas Gaj
- The Departments of Chemistry and Cell and Molecular Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - James T Patterson
- The Departments of Chemistry and Cell and Molecular Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Shannon J Sirk
- The Departments of Chemistry and Cell and Molecular Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Carlos F Barbas
- The Departments of Chemistry and Cell and Molecular Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
| |
Collapse
|
30
|
Zhang S, Wang W, Peng Y, Gu Q, Luo J, Zhou J, Wu J, Hou Y, Cao J. Amelioration of radiation-induced skin injury by HIV-TAT-mediated protein transduction of RP-1 from Rana pleurade. Int J Med Sci 2014; 11:44-51. [PMID: 24396285 PMCID: PMC3880990 DOI: 10.7150/ijms.7463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 12/11/2013] [Indexed: 11/20/2022] Open
Abstract
Radiation-induced reactive oxygen species (ROS) can damage DNA and most other biological macromolecules in skin and radiation-induced skin injury is a serious concern for radiation therapy. Skin possesses an extremely efficient antioxidant system, which is conferred by two systems: antioxidant enzymes and small molecules that can scavenge ROS by donating electrons. Amphibian skin is a multifunctional organ, which protects against dangers of various oxidative stresses. Recently, a small peptide called RP-1 was isolated from the skin secretions of Rana pleurade, which shows strong antioxidant activity. However, this RP-1 peptide is limited because its inability to across the cell membrane. Protein transduction domains (PTDs) have demonstrated high efficiency for facilitating the internalization of both homologous and heterogeneous proteins into cells. This study aims to elucidate the protective effects of a HIV-TAT (TAT) PTD-coupled RP-1 fusion protein (TAT-RP1) on radiation-induced skin injury in vitro and in vivo. The synthesized fusion TAT-RP1 peptide can be incorporated into human keratinocyte HaCaT cells in a dose- and time-dependent manner without cytotoxicity. We then evaluated the protective role of TAT-RP1 against ionizing radiation. TAT-RP1 supplementation increased anti-superoxide anion ability of HaCaT cells and decreased HaCaT cell radiosensitivity to irradiation. Moreover, TAT-RP1 was able to penetrate the skin of rats, entering epidermis as well as the dermis of the subcutaneous layer in skin tissue. Topical spread of TAT-RP1 promoted the amelioration of radiation-induced skin damage in rats. These results suggest that TAT-RP1 has potential as a protein therapy for radiation-induced skin injury.
Collapse
Affiliation(s)
- Shuyu Zhang
- 1. School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China
| | - Wenjie Wang
- 2. Cyrus Tang Hematology Center, Soochow University, Suzhou 215123, China
| | - Ying Peng
- 3. Jiangsu Institute of Nuclear Medicine and Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi 214063, China
| | - Qing Gu
- 1. School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China
| | - Judong Luo
- 1. School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China
| | - Jundong Zhou
- 4. Department of Radio-oncology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215001, China
| | - Jinchang Wu
- 4. Department of Radio-oncology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215001, China
| | - Yinglong Hou
- 5. Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014, China
| | - Jianping Cao
- 1. School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China
| |
Collapse
|
31
|
Gu Q, Feng T, Cao H, Tang Y, Ge X, Luo J, Xue J, Wu J, Yang H, Zhang S, Cao J. HIV-TAT mediated protein transduction of Cu/Zn-superoxide dismutase-1 (SOD1) protects skin cells from ionizing radiation. Radiat Oncol 2013; 8:253. [PMID: 24175971 PMCID: PMC3839640 DOI: 10.1186/1748-717x-8-253] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 10/30/2013] [Indexed: 02/03/2023] Open
Abstract
Background Radiation-induced skin injury remains a serious concern during radiotherapy. Cu/Zn-superoxide dismutase (Cu/Zn-SOD, SOD1) is a conserved enzyme for scavenging superoxide radical in cells. Because of the integrity of cell membranes, exogenous molecule is not able to be incorporated into cells, which limited the application of natural SOD1. The aim of this study was to evaluate the protective role of HIV-TAT protein transduction domain mediated protein transduction of SOD1 (TAT-SOD1) against ionizing radiation. Methods The recombinant TAT-SOD1 and SOD1 were obtained by prokaryotic–based protein expression system. The transduction effect and biological activity of TAT-SOD1 was measured by immunofluorescence and antioxidant capability assays in human keratinocyte HaCaT cells. Mito-Tracker staining, reactive oxygen species (ROS) generation assay, cell apoptosis analysis and malondialdehyde (MDA) assay were used to access the protective effect of TAT- SOD1. Results Uptake of TAT-SOD1 by HaCaT cells retained its biological activity. Compared with natural SOD1, the application of TAT-SOD1 significantly enhanced the viability and decreased the apoptosis induced by X-ray irradiation. Moreover, TAT-SOD1 reduced ROS and preserved mitochondrial integrity after radiation exposure in HaCaT cells. Radiation-induced γH2AX foci, which are representative of DNA double strand breaks, were decreased by pretreatment with TAT-SOD1. Furthermore, subcutaneous application of TAT-SOD1 resulted in a significant decrease in 45 Gy electron beam-induced ROS and MDA concentration in the skins of rats. Conclusions This study provides evidences for the protective role of TAT-SOD1 in alleviating radiation-induced damage in HaCaT cells and rat skins, which suggests a new therapeutic strategy for radiation-induced skin injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Shuyu Zhang
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | | |
Collapse
|
32
|
Pesce D, Wu Y, Kolbe A, Weil T, Herrmann A. Enhancing cellular uptake of GFP via unfolded supercharged protein tags. Biomaterials 2013; 34:4360-7. [PMID: 23478039 DOI: 10.1016/j.biomaterials.2013.02.038] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 02/13/2013] [Indexed: 01/15/2023]
Abstract
One of the barriers to the development of protein therapeutics is effective delivery to mammalian cells. The proteins must maintain a careful balance of polar moieties to enable administration and distribution and hydrophobic character to minimize cell toxicity. Numerous strategies have been applied to this end, from appending additional cationic peptides to supercharging the protein itself, sometimes with limited success. Here we present a strategy that combines these methods, by equipping a protein with supercharged elastin-like polypeptide (ELP) tags. We monitored cellular uptake and cell viability for GFP reporter proteins outfitted with a range of ELP tags and demonstrated enhanced uptake that correlates with the number of positive charges, while maintaining remarkably low cytotoxicity and resistance to degradation in the cell. GFP uptake proceeded mainly through caveolae-mediated endocytosis and we observed GFP emission inside the cells over extended time (up to 48 h). Low toxicity combined with high molecular weights of the tag opens the way to simultaneously optimize cell uptake and pharmacokinetic parameters. Thus, cationic supercharged ELP tags show great potential to improve the therapeutic profile of protein drugs leading to more efficient and safer biotherapeutics.
Collapse
Affiliation(s)
- Diego Pesce
- Department of Polymer Chemistry, Zernike Institute for Advanced Materials, University of Groningen, Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
33
|
Hu PF, Guan WJ, Li XC, Zhang WX, Li CL, Ma YH. Study on characteristics of in vitro culture and intracellular transduction of exogenous proteins in fibroblast cell line of Liaoning cashmere goat. Mol Biol Rep 2012; 40:327-36. [PMID: 23065271 DOI: 10.1007/s11033-012-2064-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 10/03/2012] [Indexed: 10/27/2022]
Abstract
Establishment of fibroblast cell lines of endangered goat breeds and research on the gene or protein functions based on the cells made a significant contribution to the conservation and utilization of genetic resources. In this study, a fibroblast cell line of Liaoning cashmere goat, frozen in 174 cryovials with 5 × 10(6) cells each, was successfully established from 60 goats ear marginal tissues using explant culture and cryopreservation techniques. Biological analysis of in vitro cultured cell line showed that, the cells were morphologically consistent with fibroblasts; the average viability of the cells was 94.9 % before freezing and 90.1 % after thawing; the growth process of cells was consisted of a lag phase, a logarithmic phase and a plateau phase; cell population doubling time was 65.5 h; more than 90 % of cells were diploid prior to the 6th generation; Neither microbial contamination nor cross-contamination was detected. To determine cell permeability, intracellular path and stability of exogenous proteins during the transduction, a TAT protein transduction domain was fused to the C-terminus of enhanced green fluorescent protein, the established fibroblast cell line was treated with the purified exogenous proteins at various concentrations by adding them to the cell culture media for 1-24 h and assayed cell morphology and protein presence, it was found that the purified exogenous proteins readily entered cells at a concentration of 0.1 mg/ml within 1.5 h and some of them could translocate into nucleus, moreover, the exogenous proteins appeared to be stable inside cells for up to 24 h.
Collapse
Affiliation(s)
- P F Hu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | | | | | | | | | | |
Collapse
|
34
|
Choi YJ, Lee JY, Chung CP, Park YJ. Cell-penetrating superoxide dismutase attenuates oxidative stress-induced senescence by regulating the p53-p21(Cip1) pathway and restores osteoblastic differentiation in human dental pulp stem cells. Int J Nanomedicine 2012; 7:5091-106. [PMID: 23049256 PMCID: PMC3459692 DOI: 10.2147/ijn.s31723] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Human dental pulp stem cells (DPSCs) have potential applications in tissue regeneration because of their convenient cell harvesting procedures and multipotent capacity. However, the tissue regenerative potential of DPSCs is known to be negatively regulated by aging in long-term culture and under oxidative stress. With an aim of reducing cellular senescence and oxidative stress in DPSCs, an intracellular delivery system for superoxide dismutase 1 (SOD1) was developed. We conjugated SOD1 with a cell-penetrating peptide known as low-molecular weight protamine (LMWP), and investigated the effect of LMWP-SOD1 conjugates on hydrogen peroxide-induced cellular senescence and osteoblastic differentiation. Results LMWP-SOD1 significantly attenuated enlarged and flattened cell morphology and increased senescence-associated β-galactosidase activity. Under the same conditions, LMWP-SOD1 abolished activation of the cell cycle regulator proteins, p53 and p21Cip1, induced by hydrogen peroxide. In addition, LMWP-SOD1 reversed the inhibition of osteoblastic differentiation and downregulation of osteogenic gene markers induced by hydrogen peroxide. However, LMWP-SOD1 could not reverse the decrease in odontogenesis caused by hydrogen peroxide. Conclusion Overall, cell-penetrating LMWP-SOD1 conjugates are effective for attenuation of cellular senescence and reversal of osteoblastic differentiation of DPSCs caused by oxidative stress inhibition. This result suggests potential application in the field of antiaging and tissue engineering to overcome the limitations of senescent stem cells.
Collapse
Affiliation(s)
- Yoon Jung Choi
- Craniomaxillofacial Reconstructive Sciences, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
35
|
Cell-penetrating properties of the transactivator of transcription and polyarginine (R9) peptides, their conjugative effect on nanoparticles and the prospect of conjugation with arsenic trioxide. Anticancer Drugs 2012; 23:471-82. [PMID: 22241171 DOI: 10.1097/cad.0b013e32835065ed] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cell-penetrating peptides (CPPs) are short chains of amino acids with the distinct ability to cross cell plasma membranes. They are usually between seven and 30 residues in length. The mechanism of action is still a highly debated subject among researchers; it seems that a commonality between all CPPs is the presence of positively charged residues within the amino acid chain. Polyarginine and the transactivator of transcription peptide are two widely used CPPs. One distinct application of these CPPs is the ability to further enhance the therapeutic properties of a range of different agents. One group of agents of particular importance are nanoparticles (NPs). Most NPs have no mechanism for cellular uptake. Hence, by conjugating CPPs to NPs, the amount of NPs taken up by cells can be increased, and therefore, the therapeutic benefits can be maximized. Some examples of this will be explored further in this review. In addition to CPPs, the concept of conjugation with the anticancer drug arsenic trioxide is reviewed and the prospect of transactivator of transcription-conjugated arsenic trioxide albumin microspheres is also discussed. Recent locked nucleic acid technology to stabilize nucleotides (RNA or DNA) aptamer complexes able to target cancer cells more specifically and selectively to kill tumour cells and spare normal body cells. NPs tagged with modified locked nucleic acid-aptamers have the potential to kill cancer cells more specifically and effectively while sparing normal cells.
Collapse
|
36
|
Giordano CR, Terlecky SR. Peroxisomes, cell senescence, and rates of aging. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1358-62. [PMID: 22497955 DOI: 10.1016/j.bbadis.2012.03.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 03/02/2012] [Accepted: 03/22/2012] [Indexed: 12/31/2022]
Abstract
The peroxisome is functionally integrated into an exquisitely complex network of communicating endomembranes which is only beginning to be appreciated. Despite great advances in identifying essential components and characterizing molecular mechanisms associated with the organelle's biogenesis and function, there is a large gap in our understanding of how peroxisomes are incorporated into metabolic pathways and subcellular communication networks, how they contribute to cellular aging, and where their influence is manifested on the initiation and progression of degenerative disease. In this review, we summarize recent evidence pointing to the organelle as an important regulator of cellular redox balance with potentially far-reaching effects on cell aging and the genesis of human disease. The roles of the organelle in lipid homeostasis, anaplerotic reactions, and other critical metabolic and biochemical processes are addressed elsewhere in this volume. This article is part of a Special Issue entitled: Metabolic Functions and Biogenesis of Peroxisomes in Health and Disease.
Collapse
Affiliation(s)
- Courtney R Giordano
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, USA
| | | |
Collapse
|
37
|
Tahara Y, Honda S, Kamiya N, Goto M. Transdermal delivery of insulin using a solid-in-oil nanodispersion enhanced by arginine-rich peptides. MEDCHEMCOMM 2012. [DOI: 10.1039/c2md20059g] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
38
|
Guo J, Chen Y, Yuan B, Liu S, Rao P. Effects of intracellular superoxide removal at acupoints with TAT-SOD on obesity. Free Radic Biol Med 2011; 51:2185-9. [PMID: 22019441 DOI: 10.1016/j.freeradbiomed.2011.09.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 08/11/2011] [Accepted: 09/29/2011] [Indexed: 01/30/2023]
Abstract
TAT-SOD is a recombinant protein of superoxide dismutase fused with TAT peptide. By pure accident, we discovered that topical application of TAT-SOD to acupoints could result in acupuncture-like action. This study aimed to validate the accidental discovery by investigating the effect on simple obesity of the topical application of TAT-SOD to acupoints in comparison with acupuncture. 90 subjects were divided into 3 groups for 12-week treatments. Regular hospital acupuncture treatment was given to Acupuncture Group 3 times a week. TAT-SOD Group were instructed first to locate acupoints and apply 0.1ml of 5000u SOD/ml TAT-SOD cream in an area of 1cm(2) to each of the same set of acupoints, which they then conducted at home three times daily. Placebo Group applied the vehicle cream the same manner as TAT-SOD Group. Both TAT-SOD and acupuncture treatments decreased adiposity with overall clinical effective rates of 60.0% and 76.7%, respectively. The placebo group showed no improvement. The results validate that the enzymatic removal of the intracellular superoxide at acupoints could generate acupuncture-like effects, and indicate a possibility of the new method as a simple substitute to acupuncture and an insight of superoxide modulation along meridians for acupuncture mechanism.
Collapse
Affiliation(s)
- Jingke Guo
- Institute of Biotechnology, Fuzhou University, 523 Gongye Road, Fuzhou, Fujian, China
| | | | | | | | | |
Collapse
|
39
|
Desai P, Patlolla RR, Singh M. Interaction of nanoparticles and cell-penetrating peptides with skin for transdermal drug delivery. Mol Membr Biol 2011; 27:247-59. [PMID: 21028936 DOI: 10.3109/09687688.2010.522203] [Citation(s) in RCA: 254] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Topical or transdermal drug delivery is challenging because the skin acts as a natural and protective barrier. Therefore, several methods have been examined to increase the permeation of therapeutic molecules into and through the skin. One approach is to use the nanoparticulate delivery system. Starting with liposomes and other vesicular systems, several other types of nanosized drug carriers have been developed such as solid lipid nanoparticles, nanostructured lipid carriers, polymer-based nanoparticles and magnetic nanoparticles for dermatological applications. This review article discusses how different particulate systems can interact and penetrate into the skin barrier. In this review, the effectiveness of nanoparticles, as well as possible mode of actions of nanoparticles, is presented. In addition to nanoparticles, cell-penetrating peptide (CPP)-mediated drug delivery into the skin and the possible mechanism of CPP-derived delivery into the skin is discussed. Lastly, the effectiveness and possible mechanism of CPP-modified nanocarriers into the skin are addressed.
Collapse
Affiliation(s)
- Pinaki Desai
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | | | | |
Collapse
|
40
|
Shuvaev VV, Muzykantov VR. Targeted modulation of reactive oxygen species in the vascular endothelium. J Control Release 2011; 153:56-63. [PMID: 21457736 DOI: 10.1016/j.jconrel.2011.03.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 03/21/2011] [Indexed: 01/28/2023]
Abstract
'Endothelial cells lining vascular luminal surface represent an important site of signaling and injurious effects of reactive oxygen species (ROS) produced by other cells and endothelium itself in ischemia, inflammation and other pathological conditions. Targeted delivery of ROS modulating enzymes conjugated with antibodies to endothelial surface molecules (vascular immunotargeting) provides site-specific interventions in the endothelial ROS, unattainable by other formulations including PEG-modified enzymes. Targeting of ROS generating enzymes (e.g., glucose oxidase) provides ROS- and site-specific models of endothelial oxidative stress, whereas targeting of antioxidant enzymes SOD and catalase offers site-specific quenching of superoxide anion and H(2)O(2). These targeted antioxidant interventions help to clarify specific role of endothelial ROS in vascular and pulmonary pathologies and provide basis for design of targeted therapeutics for treatment of these pathologies. In particular, antibody/catalase conjugates alleviate acute lung ischemia/reperfusion injury, whereas antibody/SOD conjugates inhibit ROS-mediated vasoconstriction and inflammatory endothelial signaling. Encapsulation in protease-resistant, ROS-permeable carriers targeted to endothelium prolongs protective effects of antioxidant enzymes, further diversifying the means for targeted modulation of endothelial ROS.
Collapse
Affiliation(s)
- Vladimir V Shuvaev
- Department of Pharmacology and Center for Translational Targeted Therapeutics and Nanomedicine, Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6068, USA
| | | |
Collapse
|
41
|
Shibagaki N, Okamoto T, Mitsui H, Inozume T, Kanzaki M, Shimada S. Novel immunotherapeutic approaches to skin cancer treatments using protein transduction technology. J Dermatol Sci 2011; 61:153-61. [DOI: 10.1016/j.jdermsci.2010.12.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Revised: 12/07/2010] [Accepted: 12/09/2010] [Indexed: 12/27/2022]
|
42
|
Lee JH, Lu H. Chimeric p53 as an alternative therapy for hypoxic tumors. Cancer Biol Ther 2011; 11:108-10. [PMID: 21189450 DOI: 10.4161/cbt.11.1.14526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Jun-Ho Lee
- Department of Biochemistry, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
43
|
Expression, purification, and refolding of active Nrf2 transcription factor fused to protein transduction TAT tag. Protein Expr Purif 2010; 74:280-8. [DOI: 10.1016/j.pep.2010.06.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Revised: 06/24/2010] [Accepted: 06/25/2010] [Indexed: 11/19/2022]
|
44
|
Ma WF, Chen HY, Du J, Tan Y, Cai SH. A novel recombinant protein TAT-GFP-KDEL with dual-function of penetrating cell membrane and locating at endoplasm reticulum. J Drug Target 2010; 17:329-33. [PMID: 19558358 DOI: 10.1080/10611860802582459] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Although the potential value of phenotypic/functional knockout technology with intrabody/kine in prevention and cure of some serious diseases, such as AIDS and cancer, is being regarded, there are still several technical difficulties. One of the the most critical problems is how to directly deliver the intrabody/kine proteins into endoplasm reticulum (ER). In this study, a novel recombinant protein, TAT-GFP-KDEL, was designed and constructed. In this recombinant protein, HIV-derived TAT (47-57) and an ER retention four-peptide sequence KDEL were fused at the N-terminal and C-terminal of GFP respectively. The results showed that TAT-GFP-KDEL had been successfully expressed in bacteria BL21 and its purity reached to 95%. Moreover, we observed that this recombinant protein was able to efficiently transduce into MOLT-4 cells and accurately locate at ER. This study may provide an available strategy to promote the transmembrane delivery and ER localization of protein-based intrabody/kine.
Collapse
Affiliation(s)
- Wei Feng Ma
- School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, PR China
| | | | | | | | | |
Collapse
|
45
|
Kim JE, Kim DW, Kwak SE, Ryu HJ, Yeo SI, Kwon OS, Choi SY, Kang TC. Pyridoxal-5'-phosphate phosphatase/chronophin inhibits long-term potentiation induction in the rat dentate gyrus. Hippocampus 2010; 19:1078-89. [PMID: 19253407 DOI: 10.1002/hipo.20568] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pyridoxal-5'-phosphate (PLP)-phosphatase/chronophin (PLPP/CIN) directly dephosphorylates actin-depolymerizing factor (ADF)/cofilin as well as PLP. Although PLPP/CIN plays a role in the regulation of F-actin and vitamin B(6) metabolism, there is no direct evidence to support a correlation between PLPP/CIN and F-actin polymerization during long-term potentiation (LTP) induction. In this study, we investigated whether the expression of PLPP/CIN is altered following LTP induction, and whether Tat-PLPP/CIN transduction affects LTP induction in the rat dentate gyrus (DG). PLPP/CIN immunoreactivity was markedly decreased in dentate granule cells after the induction of LTP. Tat-PLPP/CIN transduction (20 and 200 microg/kg) decreased the efficiency of high frequency stimulus-induced potentiation of populations spike amplitude as compared to saline or Tat-protein-treated animals. The PLPP/CIN protein level showed an inverse correlation with phosphorylated ADF/cofilin levels and F-actin content. These findings suggest that PLPP/CIN-mediated actin dynamics may play an important role in the changes of morphological properties (dendritic spine reorganization) of the hippocampus in LTP.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, Kangwon-Do, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Trabulo S, Cardoso AL, Mano M, De Lima MCP. Cell-Penetrating Peptides-Mechanisms of Cellular Uptake and Generation of Delivery Systems. Pharmaceuticals (Basel) 2010; 3:961-993. [PMID: 27713284 PMCID: PMC4034016 DOI: 10.3390/ph3040961] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 02/20/2010] [Accepted: 03/29/2010] [Indexed: 01/13/2023] Open
Abstract
The successful clinical application of nucleic acid-based therapeutic strategies has been limited by the poor delivery efficiency achieved by existing vectors. The development of alternative delivery systems for improved biological activity is, therefore, mandatory. Since the seminal observations two decades ago that the Tat protein, and derived peptides, can translocate across biological membranes, cell-penetrating peptides (CPPs) have been considered one of the most promising tools to improve non-invasive cellular delivery of therapeutic molecules. Despite extensive research on the use of CPPs for this purpose, the exact mechanisms underlying their cellular uptake and that of peptide conjugates remain controversial. Over the last years, our research group has been focused on the S413-PV cell-penetrating peptide, a prototype of this class of peptides that results from the combination of 13-amino-acid cell penetrating sequence derived from the Dermaseptin S4 peptide with the SV40 large T antigen nuclear localization signal. By performing an extensive biophysical and biochemical characterization of this peptide and its analogs, we have gained important insights into the mechanisms governing the interaction of CPPs with cells and their translocation across biological membranes. More recently, we have started to explore this peptide for the intracellular delivery of nucleic acids (plasmid DNA, siRNA and oligonucleotides). In this review we discuss the current knowledge of the mechanisms responsible for the cellular uptake of cell-penetrating peptides, including the S413-PV peptide, and the potential of peptide-based formulations to mediate nucleic acid delivery.
Collapse
Affiliation(s)
- Sara Trabulo
- Center for Neuroscience and Cell Biology of Coimbra, Department of Zoology, University of Coimbra, Portugal
- Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, Apartado 3126, 3001-401 Coimbra, Portugal
| | - Ana Luísa Cardoso
- Center for Neuroscience and Cell Biology of Coimbra, Department of Zoology, University of Coimbra, Portugal
- Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, Apartado 3126, 3001-401 Coimbra, Portugal
| | - Miguel Mano
- Center for Neuroscience and Cell Biology of Coimbra, Department of Zoology, University of Coimbra, Portugal
- Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, Apartado 3126, 3001-401 Coimbra, Portugal
| | - Maria C Pedroso De Lima
- Center for Neuroscience and Cell Biology of Coimbra, Department of Zoology, University of Coimbra, Portugal.
- Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, Apartado 3126, 3001-401 Coimbra, Portugal.
| |
Collapse
|
47
|
Wu Y, Ren C, Gao Y, Hou B, Chen T, Zhang C. A novel method for promoting heterologous protein expression in Escherichia coli by fusion with the HIV-1 TAT core domain. Amino Acids 2010; 39:811-20. [PMID: 20213440 DOI: 10.1007/s00726-010-0534-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Accepted: 02/12/2010] [Indexed: 11/29/2022]
Abstract
The human immunodeficiency virus type 1 (HIV-1) transactivator of transcription (TAT) protein, a member of the protein transduction domain (PTD) superfamily, can deliver heterologous proteins across most biomembranes without losing bioactivity. However, there is no report on whether the TAT core domain containing the sequence 'YGRKKRRQRRR' has other functions. As the TAT core domain is most basic (pI=12.8) and has biomembrane crossing ability, we hypothesized it might probably influence the protein expression level due to subcellular redistribution of target proteins in the cells. To address this issue, we constructed the prokaryotic expression vector pET28b-TAT-EGFP (using the vector pET28b-EGFP for control) containing the core domain coding region, and transformed the vector into E. coli BL21 (DE3) cells for expression of the enhanced green fluorescent protein (EGFP) with the inducer isopropyl-beta-D-thiogalactopyranoside (IPTG). Equal amount of the total proteins were fractionated using 15% SDS-PAGE and identified by western blot, and the plasmid copy number was assayed by Southern blot. In order to further study the subcellular localization of heterologous proteins in E. coli cells, the cytoplasmic and periplasmic components were extracted by chloroform and osmotic shock techniques. Interestingly, our data showed that the TAT core domain was not only able to promote the heterologous protein expression in E. coli, but also improve the yields and the solubility of heterologous proteins, while the plasmid copy number of TAT-containing clones and TAT-free clones was not affected by the TAT core domain. In addition, the TAT-tagged protein was mainly localized in the cytoplasm and also accumulated in the periplasmic space along with the time for protein expression, while in contrast, the TAT-free protein was mainly expressed in the periplasm and only a few in cytoplasm. A further examination on the distribution of the expressed proteins in cytoplasm and periplasm suggested that the TAT core domain might promote protein expression in the cytoplasm initially and then partially deliver them across the cytomembrane to the periplasmic space in a concentration-dependent manner. Taken together, our current data have provided a novel method for improving heterologous protein expression in prokaryotic cells by fusion with the TAT core domain, which will promote expression efficiency of bioactive proteins for protein engineering.
Collapse
Affiliation(s)
- Yonghong Wu
- State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Taiping Road 27, 100850, Beijing, China
| | | | | | | | | | | |
Collapse
|
48
|
Borysowski J, Górski A. Fusion to cell-penetrating peptides will enable lytic enzymes to kill intracellular bacteria. Med Hypotheses 2010; 74:164-6. [DOI: 10.1016/j.mehy.2009.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 07/03/2009] [Indexed: 12/26/2022]
|
49
|
Manipulation of cellular GSH biosynthetic capacity via TAT-mediated protein transduction of wild-type or a dominant-negative mutant of glutamate cysteine ligase alters cell sensitivity to oxidant-induced cytotoxicity. Toxicol Appl Pharmacol 2009; 243:35-45. [PMID: 19914271 DOI: 10.1016/j.taap.2009.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 11/04/2009] [Accepted: 11/06/2009] [Indexed: 02/07/2023]
Abstract
The glutathione (GSH) antioxidant defense system plays a central role in protecting mammalian cells against oxidative injury. Glutamate cysteine ligase (GCL) is the rate-limiting enzyme in GSH biosynthesis and is a heterodimeric holoenzyme composed of catalytic (GCLC) and modifier (GCLM) subunits. As a means of assessing the cytoprotective effects of enhanced GSH biosynthetic capacity, we have developed a protein transduction approach whereby recombinant GCL protein can be rapidly and directly transferred into cells when coupled to the HIV TAT protein transduction domain. Bacterial expression vectors encoding TAT fusion proteins of both GCL subunits were generated and recombinant fusion proteins were synthesized and purified to near homogeneity. The TAT-GCL fusion proteins were capable of heterodimerization and formation of functional GCL holoenzyme in vitro. Exposure of Hepa-1c1c7 cells to the TAT-GCL fusion proteins resulted in the time- and dose-dependent transduction of both GCL subunits and increased cellular GCL activity and GSH levels. A heterodimerization-competent, enzymatically deficient GCLC-TAT mutant was also generated in an attempt to create a dominant-negative suppressor of GCL. Transduction of cells with a catalytically inactive GCLC(E103A)-TAT mutant decreased cellular GCL activity in a dose-dependent manner. TAT-mediated manipulation of cellular GCL activity was also functionally relevant as transduction with wild-type GCLC(WT)-TAT or mutant GCLC(E103A)-TAT conferred protection or enhanced sensitivity to H(2)O(2)-induced cell death, respectively. These findings demonstrate that TAT-mediated transduction of wild-type or dominant-inhibitory mutants of the GCL subunits is a viable means of manipulating cellular GCL activity to assess the effects of altered GSH biosynthetic capacity.
Collapse
|
50
|
Ma WF, Du J, Fu LP, Fang R, Chen HY, Cai SH. Phenotypic knockout of CXCR4 by a novel recombinant protein TAT/54R/KDEL inhibits tumors metastasis. Mol Cancer Res 2009; 7:1613-21. [PMID: 19825996 DOI: 10.1158/1541-7786.mcr-09-0078] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The chemokine receptor, CXCR4, and its specific ligand, CXCL12, have been proven to regulate the directional trafficking and invasion of breast cancer cells to sites of metastases, and similar phenomena have also been identified in many malignant tumors that aberrantly overexpress CXCR4. Therefore, blocking the interaction between CXCR4 and CXCL12 is considered a possible approach to efficiently prevent cancer metastasis. Employing a cellular phenotypic knockout strategy based on intrakines, we developed a novel recombinant chimeric protein, TAT/54R/KDEL, which contains three distinct functional domains: CXCL12/54R, a mutant of CXCL12 with CXCR4 antagonism, as well as HIV-derived TAT (47-57) and an endoplasmic reticulum retention four-peptide sequence KDEL that links at its NH(2) and COOH termini, respectively. Using the MOLT-4 cell line, which expressed CXCR4 highly and stably in vitro, we determined that TAT/54R/KDEL was able to efficiently transfer into the endoplasmic reticulum of tumor cells, where it specifically binds to the newly synthesized CXCR4 and prevents the latter from reaching the surface. Chemotaxis assays showed that the cells treated with TAT/54R/KDEL failed to migrate toward CXCL12. Furthermore, we observed that the systemic treatment of TAT/54R/KDEL could impair lung metastasis in a highly metastatic mammary cancer cell line, 4T1 cells, with the decrease of CXCR4 on their membrane. Our results suggest that the phenotypic knockout strategy of CXCR4 using a novel recombinant protein TAT/54R/KDEL might be a possible approach for inhibiting relative tumor metastasis mediated by CXCR4/CXCL12 interaction.
Collapse
Affiliation(s)
- Wei-Feng Ma
- Department of Microbiology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, P.R. China
| | | | | | | | | | | |
Collapse
|