1
|
Nie Y, Fu G, Leng Y. Nuclear Delivery of Nanoparticle-Based Drug Delivery Systems by Nuclear Localization Signals. Cells 2023; 12:1637. [PMID: 37371107 DOI: 10.3390/cells12121637] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/31/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Nanomedicine 2.0 refers to the next generation of nanotechnology-based medical therapies and diagnostic tools. This field focuses on the development of more sophisticated and precise nanoparticles (NPs) for targeted drug delivery, imaging, and sensing. It has been established that the nuclear delivery of NP-loaded drugs can increase their therapeutic efficacy. To effectively direct the NPs to the nucleus, the attachment of nuclear localization signals (NLSs) to NPs has been employed in many applications. In this review, we will provide an overview of the structure of nuclear pore complexes (NPCs) and the classic nuclear import mechanism. Additionally, we will explore various nanoparticles, including their synthesis, functionalization, drug loading and release mechanisms, nuclear targeting strategies, and potential applications. Finally, we will highlight the challenges associated with developing nucleus-targeted nanoparticle-based drug delivery systems (NDDSs) and provide insights into the future of NDDSs.
Collapse
Affiliation(s)
- Yuhan Nie
- Innovation and Integration Center of New Laser Technology, Shanghai Institute of Optics and Fine Mechanics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Guo Fu
- Innovation and Integration Center of New Laser Technology, Shanghai Institute of Optics and Fine Mechanics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Yuxin Leng
- State Key Laboratory of High Field Laser Physics and CAS Center for Excellence in Ultra-Intense Laser Science, Shanghai Institute of Optics and Fine Mechanics, Chinese Academy of Sciences, Shanghai 201800, China
| |
Collapse
|
2
|
TAT-Conjugated NDUFS8 Can Be Transduced into Mitochondria in a Membrane-Potential-Independent Manner and Rescue Complex I Deficiency. Int J Mol Sci 2021; 22:ijms22126524. [PMID: 34204592 PMCID: PMC8234171 DOI: 10.3390/ijms22126524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/12/2021] [Accepted: 06/15/2021] [Indexed: 12/21/2022] Open
Abstract
NADH dehydrogenase (ubiquinone) Fe-S protein 8 (NDUFS8) is a nuclear-encoded core subunit of human mitochondrial complex I. Defects in NDUFS8 are associated with Leigh syndrome and encephalomyopathy. Cell-penetrating peptide derived from the HIV-1 transactivator of transcription protein (TAT) has been successfully applied as a carrier to bring fusion proteins into cells without compromising the biological function of the cargoes. In this study, we developed a TAT-mediated protein transduction system to rescue complex I deficiency caused by NDUFS8 defects. Two fusion proteins (TAT-NDUFS8 and NDUFS8-TAT) were exogenously expressed and purified from Escherichia coli for transduction of human cells. In addition, similar constructs were generated and used in transfection studies for comparison. The results showed that both exogenous TAT-NDUFS8 and NDUFS8-TAT were delivered into mitochondria and correctly processed. Interestingly, the mitochondrial import of TAT-containing NDUFS8 was independent of mitochondrial membrane potential. Treatment with TAT-NDUFS8 not only significantly improved the assembly of complex I in an NDUFS8-deficient cell line, but also partially rescued complex I functions both in the in-gel activity assay and the oxygen consumption assay. Our current findings suggest the considerable potential of applying the TAT-mediated protein transduction system for treatment of complex I deficiency.
Collapse
|
3
|
Jubran R, Saar-Ray M, Wawruszak A, Ziporen L, Donin N, Bairey O, Fishelson Z. Mortalin peptides exert antitumor activities and act as adjuvants to antibody-mediated complement-dependent cytotoxicity. Int J Oncol 2020; 57:1013-1026. [PMID: 32700755 DOI: 10.3892/ijo.2020.5101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/09/2020] [Indexed: 11/06/2022] Open
Abstract
Cancer cells have developed numerous strategies to maintain their proliferative capacity and to withstand different kinds of stress. The mitochondrial stress‑70 protein named glucose regulated protein 75 (GRP75), also known as mortalin, is an intriguing cancer pro‑survival factor. It is constitutively expressed in normal tissues but is upregulated in many tumors, and was shown to be a cancer prognostic biomarker. Mortalin is an inhibitor of complement‑dependent cytotoxicity (CDC) and may therefore protect cells from antibody‑based immunotherapy. To target mortalin for cancer therapy, our laboratory designed several mortalin mimetic peptides with sequences predicted to be involved in mortalin binding to its client proteins. The peptides were synthesized with a C‑terminal transactivator of transcription sequence. By using cell death methodologies, the mechanism of action of the mortalin mimetic peptides on cancer cells was studied. Two peptides in particular, Mot‑P2 and Mot‑P7, were found to be highly toxic to lymphoma and ovarian, breast and prostate carcinoma cells. The analysis of their mode of action revealed that they may induce, within minutes, plasma membrane perturbations and mitochondrial stress. Furthermore, Mot‑P2 and Mot‑P7 activated necrotic cell death, leading to plasma membrane perforation, mitochondrial inner membrane depolarization and decrease in ATP level. In addition, Mot‑P7, but not Mot‑P2, required extracellular calcium ions to fully mediate cell death and was partially inhibited by plasma membrane cholesterol. At sub‑toxic concentrations, the two peptides moderately inhibited cancer cell proliferation and blocked cell cycle at G2/M. Both peptides may bind intracellularly to mortalin and/or a mortalin‑binding protein, hence knocking down mortalin expression reduced cell death. Combining treatment with Mot‑P2 or Mot‑P7 and CDC resulted in increased cell death. This study identified highly cytotoxic mortalin mimetic peptides that may be used as monotherapy or combined with complement‑activating antibody therapy to target mortalin for precision cancer therapy.
Collapse
Affiliation(s)
- Ritta Jubran
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Moran Saar-Ray
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin 20-093, Poland
| | - Lea Ziporen
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Natalie Donin
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Osnat Bairey
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center - Beilinson Hospital, Petach Tikva 49100, and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Zvi Fishelson
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
4
|
Kang YC, Son M, Kang S, Im S, Piao Y, Lim KS, Song MY, Park KS, Kim YH, Pak YK. Cell-penetrating artificial mitochondria-targeting peptide-conjugated metallothionein 1A alleviates mitochondrial damage in Parkinson's disease models. Exp Mol Med 2018; 50:1-13. [PMID: 30120245 PMCID: PMC6098059 DOI: 10.1038/s12276-018-0124-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/10/2018] [Accepted: 05/08/2018] [Indexed: 12/14/2022] Open
Abstract
An excess of reactive oxygen species (ROS) relative to the antioxidant capacity causes oxidative stress, which plays a role in the development of Parkinson’s disease (PD). Because mitochondria are both sites of ROS generation and targets of ROS damage, the delivery of antioxidants to mitochondria might prevent or alleviate PD. To transduce the antioxidant protein human metallothionein 1A (hMT1A) into mitochondria, we computationally designed a cell-penetrating artificial mitochondria-targeting peptide (CAMP). The recombinant CAMP-conjugated hMT1A fusion protein (CAMP-hMT1A) successfully localized to the mitochondria. Treating a cell culture model of PD with CAMP-hMT1A restored tyrosine hydroxylase expression and mitochondrial activity and reduced ROS production. Furthermore, injection of CAMP-hMT1A into the brain of a mouse model of PD rescued movement impairment and dopaminergic neuronal degeneration. CAMP-hMT1A delivery into mitochondria might be therapeutic against PD by alleviating mitochondrial damage, and we predict that CAMP could be used to deliver other cargo proteins to the mitochondria. A peptide targeting mitochondria can help deliver an antioxidant protein to mitigate the effects of Parkinson’s disease in cellular and mouse models. Youngmi Pak from Kyung Hee University, Seoul, South Korea, and co-workers engineered bacteria to express the human version of an antioxidant protein called metallothionein 1A fused to a short peptide sequence so that it localizes to mitochondria, the cellular power plants. Once inside the mitochondria, the peptide is removed, leaving the mature antioxidant protein to mop up damaging free radicals, a common problem seen in the cells of patients with Parkinson’s disease, and restore mitochondria to a healthier state. The protein improved mitochondrial function in both a human cell line and in the neurons of mice with a Parkinson’s-like disease, suggesting it might also help patients with this devastating neurological condition.
Collapse
Affiliation(s)
- Young Cheol Kang
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, 02447, Korea
| | - Minuk Son
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, 02447, Korea
| | - Sora Kang
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, 02447, Korea
| | - Suyeol Im
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, 02447, Korea
| | - Ying Piao
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, 02447, Korea.,Department of Emergency, Yanbian University Hospital, Yanji City, Jilin Province, China
| | - Kwang Suk Lim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, 133-791, Korea
| | - Min-Young Song
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, 02447, Korea.,Biomedical Omics Group, Korea Basic Science Institute, Cheongju-si, Chungbuk, South Korea
| | - Kang-Sik Park
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, 02447, Korea
| | - Yong-Hee Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, 133-791, Korea
| | - Youngmi Kim Pak
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, 02447, Korea. .,Department of Physiology, College of Medicine, Kyung Hee University, Seoul, 02447, Korea.
| |
Collapse
|
5
|
Verechshagina NA, Konstantinov YM, Kamenski PA, Mazunin IO. Import of Proteins and Nucleic Acids into Mitochondria. BIOCHEMISTRY (MOSCOW) 2018; 83:643-661. [DOI: 10.1134/s0006297918060032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
6
|
Verechshagina N, Nikitchina N, Yamada Y, Harashima Н, Tanaka M, Orishchenko K, Mazunin I. Future of human mitochondrial DNA editing technologies. Mitochondrial DNA A DNA Mapp Seq Anal 2018; 30:214-221. [PMID: 29764251 DOI: 10.1080/24701394.2018.1472773] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
ATP and other metabolites, which are necessary for the development, maintenance, and functioning of bodily cells are all synthesized in the mitochondria. Multiple copies of the genome, present within the mitochondria, together with its maternal inheritance, determine the clinical manifestation and spreading of mutations in mitochondrial DNA (mtDNA). The main obstacle in the way of thorough understanding of mitochondrial biology and the development of gene therapy methods for mitochondrial diseases is the absence of systems that allow to directly change mtDNA sequence. Here, we discuss existing methods of manipulating the level of mtDNA heteroplasmy, as well as the latest systems, that could be used in the future as tools for human mitochondrial genome editing.
Collapse
Affiliation(s)
- N Verechshagina
- a Laboratory of Molecular Genetics Technologies , Immanuel Kant Baltic Federal University , Kaliningrad , Russia
| | - N Nikitchina
- a Laboratory of Molecular Genetics Technologies , Immanuel Kant Baltic Federal University , Kaliningrad , Russia
| | - Y Yamada
- b Faculty of Pharmaceutical Sciences, Laboratory for Molecular Design of Pharmaceutics , Hokkaido University , Sapporo , Japan
| | - Н Harashima
- b Faculty of Pharmaceutical Sciences, Laboratory for Molecular Design of Pharmaceutics , Hokkaido University , Sapporo , Japan
| | - M Tanaka
- c Department for Health and Longevity Research , National Institutes of Biomedical Innovation, Health and Nutrition , Ibaraki City, Osaka , Japan.,d Department of Neurology , Juntendo University Graduate School of Medicine , Tokyo , Japan
| | - K Orishchenko
- a Laboratory of Molecular Genetics Technologies , Immanuel Kant Baltic Federal University , Kaliningrad , Russia.,e Laboratory of Cell Technologies , Institute of Cytology and Genetics SB RAS , Novosibirsk , Russia
| | - I Mazunin
- a Laboratory of Molecular Genetics Technologies , Immanuel Kant Baltic Federal University , Kaliningrad , Russia
| |
Collapse
|
7
|
Erlich‐Hadad T, Hadad R, Feldman A, Greif H, Lictenstein M, Lorberboum‐Galski H. TAT-MTS-MCM fusion proteins reduce MMA levels and improve mitochondrial activity and liver function in MCM-deficient cells. J Cell Mol Med 2018; 22:1601-1613. [PMID: 29265583 PMCID: PMC5824393 DOI: 10.1111/jcmm.13435] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 09/25/2017] [Indexed: 12/14/2022] Open
Abstract
Methylmalonic aciduria (MMA) is a disorder of organic acid metabolism resulting from a functional defect of the mitochondrial enzyme, methylmalonyl-CoA mutase (MCM). The main treatments for MMA patients are dietary restriction of propiogenic amino acids and carnitine supplementation. Liver or combined liver/kidney transplantation has been used to treat those with the most severe clinical manifestations. Thus, therapies are necessary to help improve quality of life and prevent liver, renal and neurological complications. Previously, we successfully used the TAT-MTS-Protein approach for replacing a number of mitochondrial-mutated proteins. In this targeted system, TAT, an 11 a.a peptide, which rapidly and efficiently can cross biological membranes, is fused to a mitochondrial targeting sequence (MTS), followed by the mitochondrial mature protein which sends the protein into the mitochondria. In the mitochondria, the TAT-MTS is cleaved off and the native protein integrates into its natural complexes and is fully functional. In this study, we used heterologous MTSs of human, nuclear-encoded mitochondrial proteins, to target the human MCM protein into the mitochondria. All fusion proteins reached the mitochondria and successfully underwent processing. Treatment of MMA patient fibroblasts with these fusion proteins restored mitochondrial activity such as ATP production, mitochondrial membrane potential and oxygen consumption, indicating the importance of mitochondrial function in this disease. Treatment with the fusion proteins enhanced cell viability and most importantly reduced MMA levels. Treatment also enhanced albumin and urea secretion in a CRISPR/Cas9-engineered HepG2 MUT (-/-) liver cell line. Therefore, we suggest using this TAT-MTS-Protein approach for the treatment of MMA.
Collapse
Affiliation(s)
- Tal Erlich‐Hadad
- Department of Biochemistry and Molecular BiologyInstitute for Medical Research Israel‐Canada (IMRIC)Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
| | - Rita Hadad
- Department of Biochemistry and Molecular BiologyInstitute for Medical Research Israel‐Canada (IMRIC)Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
| | | | | | - Michal Lictenstein
- Department of Biochemistry and Molecular BiologyInstitute for Medical Research Israel‐Canada (IMRIC)Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
| | - Haya Lorberboum‐Galski
- Department of Biochemistry and Molecular BiologyInstitute for Medical Research Israel‐Canada (IMRIC)Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
| |
Collapse
|
8
|
Zielonka J, Sikora A, Hardy M, Ouari O, Vasquez-Vivar J, Cheng G, Lopez M, Kalyanaraman B. Mitochondria-Targeted Triphenylphosphonium-Based Compounds: Syntheses, Mechanisms of Action, and Therapeutic and Diagnostic Applications. Chem Rev 2017; 117:10043-10120. [PMID: 28654243 PMCID: PMC5611849 DOI: 10.1021/acs.chemrev.7b00042] [Citation(s) in RCA: 1041] [Impact Index Per Article: 130.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mitochondria are recognized as one of the most important targets for new drug design in cancer, cardiovascular, and neurological diseases. Currently, the most effective way to deliver drugs specifically to mitochondria is by covalent linking a lipophilic cation such as an alkyltriphenylphosphonium moiety to a pharmacophore of interest. Other delocalized lipophilic cations, such as rhodamine, natural and synthetic mitochondria-targeting peptides, and nanoparticle vehicles, have also been used for mitochondrial delivery of small molecules. Depending on the approach used, and the cell and mitochondrial membrane potentials, more than 1000-fold higher mitochondrial concentration can be achieved. Mitochondrial targeting has been developed to study mitochondrial physiology and dysfunction and the interaction between mitochondria and other subcellular organelles and for treatment of a variety of diseases such as neurodegeneration and cancer. In this Review, we discuss efforts to target small-molecule compounds to mitochondria for probing mitochondria function, as diagnostic tools and potential therapeutics. We describe the physicochemical basis for mitochondrial accumulation of lipophilic cations, synthetic chemistry strategies to target compounds to mitochondria, mitochondrial probes, and sensors, and examples of mitochondrial targeting of bioactive compounds. Finally, we review published attempts to apply mitochondria-targeted agents for the treatment of cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
- Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
- Cancer Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Adam Sikora
- Institute of Applied Radiation Chemistry, Lodz University of Technology, ul. Wroblewskiego 15, 93-590 Lodz, Poland
| | - Micael Hardy
- Aix Marseille Univ, CNRS, ICR, UMR 7273, 13013 Marseille, France
| | - Olivier Ouari
- Aix Marseille Univ, CNRS, ICR, UMR 7273, 13013 Marseille, France
| | - Jeannette Vasquez-Vivar
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
- Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Gang Cheng
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
- Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Marcos Lopez
- Translational Biomedical Research Group, Biotechnology Laboratories, Cardiovascular Foundation of Colombia, Carrera 5a No. 6-33, Floridablanca, Santander, Colombia, 681003
- Graduate Program of Biomedical Sciences, Faculty of Health, Universidad del Valle, Calle 4B No. 36-00, Cali, Colombia, 760032
| | - Balaraman Kalyanaraman
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
- Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
- Cancer Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| |
Collapse
|
9
|
Alta RYP, Vitorino HA, Goswami D, Liria CW, Wisnovsky SP, Kelley SO, Machini MT, Espósito BP. Mitochondria-penetrating peptides conjugated to desferrioxamine as chelators for mitochondrial labile iron. PLoS One 2017; 12:e0171729. [PMID: 28178347 PMCID: PMC5298241 DOI: 10.1371/journal.pone.0171729] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/24/2017] [Indexed: 12/16/2022] Open
Abstract
Desferrioxamine (DFO) is a bacterial siderophore with a high affinity for iron, but low cell penetration. As part of our ongoing project focused on DFO-conjugates, we synthesized, purified, characterized and studied new mtDFOs (DFO conjugated to the Mitochondria Penetrating Peptides TAT49-57, 1A, SS02 and SS20) using a succinic linker. These new conjugates retained their strong iron binding ability and antioxidant capacity. They were relatively non toxic to A2780 cells (IC50 40–100 μM) and had good mitochondrial localization (Rr +0.45 –+0.68) as observed when labeled with carboxy-tetramethylrhodamine (TAMRA) In general, mtDFO caused only modest levels of mitochondrial DNA (mtDNA) damage. DFO-SS02 retained the antioxidant ability of the parent peptide, shown by the inhibition of mitochondrial superoxide formation. None of the compounds displayed cell cycle arrest or enhanced apoptosis. Taken together, these results indicate that mtDFO could be promising compounds for amelioration of the disease symptoms of iron overload in mitochondria.
Collapse
Affiliation(s)
- Roxana Y. P. Alta
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Hector A. Vitorino
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Cleber W. Liria
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Simon P. Wisnovsky
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Shana O. Kelley
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - M. Terêsa Machini
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail: (MTM); (BPE)
| | - Breno P. Espósito
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail: (MTM); (BPE)
| |
Collapse
|
10
|
Marcus D, Lichtenstein M, Cohen N, Hadad R, Erlich-Hadad T, Greif H, Lorberboum-Galski H. Heterologous mitochondrial targeting sequences can deliver functional proteins into mitochondria. Int J Biochem Cell Biol 2016; 81:48-56. [PMID: 27771440 DOI: 10.1016/j.biocel.2016.10.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 10/13/2016] [Accepted: 10/18/2016] [Indexed: 01/23/2023]
Abstract
Mitochondrial Targeting Sequences (MTSs) are responsible for trafficking nuclear-encoded proteins into mitochondria. Once entering the mitochondria, the MTS is recognized and cleaved off. Some MTSs are long and undergo two-step processing, as in the case of the human frataxin (FXN) protein (80aa), implicated in Friedreich's ataxia (FA). Therefore, we chose the FXN protein to examine whether nuclear-encoded mitochondrial proteins can efficiently be targeted via a heterologous MTS (hMTS) and deliver a functional protein into mitochondria. We examined three hMTSs; that of citrate synthase (cs), lipoamide deydrogenase (LAD) and C6ORF66 (ORF), as classically MTS sequences, known to be removed by one-step processing, to deliver FXN into mitochondria, in the form of fusion proteins. We demonstrate that using hMTSs for delivering FXN results in the production of 4-5-fold larger amounts of the fusion proteins, and at 4-5-fold higher concentrations. Moreover, hMTSs delivered a functional FXN protein into the mitochondria even more efficiently than the native MTSfxn, as evidenced by the rescue of FA patients' cells from oxidative stress; demonstrating a 18%-54% increase in cell survival; and a 13%-33% increase in ATP levels, as compared to the fusion protein carrying the native MTS. One fusion protein with MTScs increased aconitase activity within patients' cells, by 400-fold. The implications form our studies are of vast importance for both basic and translational research of mitochondrial proteins as any mitochondrial protein can be delivered efficiently by an hMTS. Moreover, effective targeting of functional proteins is important for restoration of mitochondrial function and treatment of related disorders.
Collapse
Affiliation(s)
- Dana Marcus
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Michal Lichtenstein
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Natali Cohen
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Rita Hadad
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Tal Erlich-Hadad
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | | | - Haya Lorberboum-Galski
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel.
| |
Collapse
|
11
|
Jain A, Chugh A. Mitochondrial transit peptide exhibits cell penetration ability and efficiently delivers macromolecules to mitochondria. FEBS Lett 2016; 590:2896-905. [PMID: 27461847 DOI: 10.1002/1873-3468.12329] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/15/2016] [Accepted: 07/19/2016] [Indexed: 01/09/2023]
Abstract
Mitochondrial malfunction under various circumstances can lead to a variety of disorders. Effective targeting of macromolecules (drugs) is important for restoration of mitochondrial function and treatment of related disorders. We have designed a novel cell-penetrating mitochondrial transit peptide (CpMTP) for delivery of macromolecules to mitochondria. Comparison between properties of cell-penetrating peptides (CPPs) and mitochondrial signal sequences enabled prediction of peptides with dual ability for cellular translocation and mitochondrial localization. Among the predicted peptides, CpMTP translocates across HeLa cells and shows successful delivery of noncovalently conjugated cargo molecules to mitochondria. CpMTP may have applications in transduction and transfection of mitochondria for therapeutics.
Collapse
Affiliation(s)
- Aastha Jain
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, India
| | - Archana Chugh
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, India
| |
Collapse
|
12
|
Towards the development of an enzyme replacement therapy for the metabolic disorder propionic acidemia. Mol Genet Metab Rep 2016; 8:51-60. [PMID: 27504265 PMCID: PMC4968140 DOI: 10.1016/j.ymgmr.2016.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/30/2016] [Accepted: 06/30/2016] [Indexed: 12/11/2022] Open
Abstract
Propionic acidemia (PA) is a life-threatening disease caused by the deficiency of a mitochondrial biotin-dependent enzyme known as propionyl coenzyme-A carboxylase (PCC). This enzyme is responsible for degrading the metabolic intermediate, propionyl coenzyme-A (PP-CoA), derived from multiple metabolic pathways. Currently, except for drastic surgical and dietary intervention that can only provide partial symptomatic relief, no other form of therapeutic option is available for this genetic disorder. Here, we examine a novel approach in protein delivery by specifically targeting and localizing our protein candidate of interest into the mitochondrial matrix of the cells. In order to test this concept of delivery, we have utilized cell penetrating peptides (CPPs) and mitochondria targeting sequences (MTS) to form specific fusion PCC protein, capable of translocating and localizing across cell membranes. In vitro delivery of our candidate fusion proteins, evaluated by confocal images and enzymatic activity assay, indicated effectiveness of this strategy. Therefore, it holds immense potential in creating a new paradigm in site-specific protein delivery and enzyme replacement therapeutic for PA.
Collapse
Key Words
- CPPs, cell penetrating peptides
- CoA, coenzyme-A
- ERT, enzyme replacement therapy
- Enzyme replacement therapy
- His-tag, six histidines tag
- LAD, lipoamine dehydrogenase
- MPP, mitochondrial processing peptidase
- MTS, mitochondria targeting sequences
- Mitochondrial targeting sequences
- PA, propionic acidemia
- PCC, propionyl coenzyme-A carboxylase
- PCCA, PCCα subunit
- PCCB, PCCβ subunit
- PP-CoA, propionyl coenzyme-A
- Propionic acidemia
- Propionyl coenzyme-A carboxylase
- Protein transduction domains
- UPLC-MS/MS, ultra performance liquid chromatography tandem mass spectrometry
Collapse
|
13
|
Chiu LS, Anderton RS, Knuckey NW, Meloni BP. The neuroprotective potential of arginine-rich peptides for the acute treatment of traumatic brain injury. Expert Rev Neurother 2016; 16:361-3. [PMID: 26840929 DOI: 10.1586/14737175.2016.1150180] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Li Shan Chiu
- a Western Australian Neuroscience Research Institute , Nedlands , Australia.,b Centre for Neuromuscular and Neurological Disorders , The University of Western Australia , Nedlands , Australia
| | - Ryan S Anderton
- a Western Australian Neuroscience Research Institute , Nedlands , Australia.,b Centre for Neuromuscular and Neurological Disorders , The University of Western Australia , Nedlands , Australia.,c School of Heath Sciences , The University Notre Dame Australia , Fremantle , Western Australia , Australia
| | - Neville W Knuckey
- a Western Australian Neuroscience Research Institute , Nedlands , Australia.,b Centre for Neuromuscular and Neurological Disorders , The University of Western Australia , Nedlands , Australia.,d Department of Neurosurgery , Sir Charles Gairdner Hospital, QEII Medical Centre , Nedlands , Western Australia , Australia
| | - Bruno P Meloni
- a Western Australian Neuroscience Research Institute , Nedlands , Australia.,b Centre for Neuromuscular and Neurological Disorders , The University of Western Australia , Nedlands , Australia.,d Department of Neurosurgery , Sir Charles Gairdner Hospital, QEII Medical Centre , Nedlands , Western Australia , Australia
| |
Collapse
|
14
|
Tischner C, Wenz T. Keep the fire burning: Current avenues in the quest of treating mitochondrial disorders. Mitochondrion 2015; 24:32-49. [DOI: 10.1016/j.mito.2015.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 06/18/2015] [Accepted: 06/24/2015] [Indexed: 12/18/2022]
|
15
|
Sato Y, Nakamura T, Yamada Y, Akita H, Harashima H. Multifunctional enveloped nanodevices (MENDs). ADVANCES IN GENETICS 2015; 88:139-204. [PMID: 25409606 DOI: 10.1016/b978-0-12-800148-6.00006-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
It is anticipated that nucleic acid medicines will be in widespread use in the future, since they have the potential to cure diseases based on molecular mechanisms at the level of gene expression. However, intelligent delivery systems are required to achieve nucleic acid therapy, since they can perform their function only when they reach the intracellular site of action. We have been developing a multifunctional envelope-type nanodevice abbreviated as MEND, which consists of functional nucleic acids as a core and lipid envelope, and can control not only biodistribution but also the intracellular trafficking of nucleic acids. In this chapter, we review the development and evolution of the MEND by providing several successful examples, including the R8-MEND, the KALA-MEND, the MITO-Porter, the YSK-MEND, and the PALM.
Collapse
Affiliation(s)
- Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo City, Hokkaido, Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo City, Hokkaido, Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo City, Hokkaido, Japan
| | - Hidetaka Akita
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo City, Hokkaido, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo City, Hokkaido, Japan
| |
Collapse
|
16
|
Lin R, Zhang P, Cheetham AG, Walston J, Abadir P, Cui H. Dual peptide conjugation strategy for improved cellular uptake and mitochondria targeting. Bioconjug Chem 2014; 26:71-7. [PMID: 25547808 PMCID: PMC4306504 DOI: 10.1021/bc500408p] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mitochondria are critical regulators of cellular function and survival. Delivery of therapeutic and diagnostic agents into mitochondria is a challenging task in modern pharmacology because the molecule to be delivered needs to first overcome the cell membrane barrier and then be able to actively target the intracellular organelle. Current strategy of conjugating either a cell penetrating peptide (CPP) or a subcellular targeting sequence to the molecule of interest only has limited success. We report here a dual peptide conjugation strategy to achieve effective delivery of a non-membrane-penetrating dye 5-carboxyfluorescein (5-FAM) into mitochondria through the incorporation of both a mitochondrial targeting sequence (MTS) and a CPP into one conjugated molecule. Notably, circular dichroism studies reveal that the combined use of α-helix and PPII-like secondary structures has an unexpected, synergistic contribution to the internalization of the conjugate. Our results suggest that although the use of positively charged MTS peptide allows for improved targeting of mitochondria, with MTS alone it showed poor cellular uptake. With further covalent linkage of the MTS-5-FAM conjugate to a CPP sequence (R8), the dually conjugated molecule was found to show both improved cellular uptake and effective mitochondria targeting. We believe these results offer important insight into the rational design of peptide conjugates for intracellular delivery.
Collapse
Affiliation(s)
- Ran Lin
- Department of Chemical and Biomolecular Engineering, ‡Institute for NanoBioTechnology, §Division of Geriatrics Medicine and Gerontology, and ⊥Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University , Baltimore, Maryland 21218, United States
| | | | | | | | | | | |
Collapse
|
17
|
Pepe S, Mentzer RM, Gottlieb RA. Cell-permeable protein therapy for complex I dysfunction. J Bioenerg Biomembr 2014; 46:337-45. [PMID: 25005682 DOI: 10.1007/s10863-014-9559-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 06/18/2014] [Indexed: 01/09/2023]
Abstract
Complex I deficiency is difficult to treat because of the size and complexity of the multi-subunit enzyme complex. Mutations or deletions in the mitochondrial genome are not amenable to gene therapy. However, animal studies have shown that yeast-derived internal NADH quinone oxidoreductase (Ndi1) can be delivered as a cell-permeable recombinant protein (Tat-Ndi1) that can functionally replace complex I damaged by ischemia/reperfusion. Current and future treatment of disorders affecting complex I are discussed, including the use of Tat-Ndi1.
Collapse
Affiliation(s)
- Salvatore Pepe
- Heart Research, Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, Australia
| | | | | |
Collapse
|
18
|
killerFLIP: a novel lytic peptide specifically inducing cancer cell death. Cell Death Dis 2013; 4:e894. [PMID: 24176852 PMCID: PMC3920952 DOI: 10.1038/cddis.2013.401] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 09/03/2013] [Accepted: 09/05/2013] [Indexed: 02/07/2023]
Abstract
One of the objectives in the development of effective cancer therapy is induction of tumor-selective cell death. Toward this end, we have identified a small peptide that, when introduced into cells via a TAT cell-delivery system, shows a remarkably potent cytoxicity in a variety of cancer cell lines and inhibits tumor growth in vivo, whereas sparing normal cells and tissues. This fusion peptide was named killerFLIP as its sequence was derived from the C-terminal domain of c-FLIP, an anti-apoptotic protein. Using structure activity analysis, we determined the minimal bioactive core of killerFLIP, namely killerFLIP-E. Structural analysis of cells using electron microscopy demonstrated that killerFLIP-E triggers cell death accompanied by rapid (within minutes) plasma membrane permeabilization. Studies of the structure of the active core of killerFLIP (-E) indicated that it possesses amphiphilic properties and self-assembles into micellar structures in aqueous solution. The biochemical properties of killerFLIP are comparable to those of cationic lytic peptides, which participate in defense against pathogens and have also demonstrated anticancer properties. We show that the pro-cell death effects of killerFLIP are independent of its sequence similarity with c-FLIPL as killerFLIP-induced cell death was largely apoptosis and necroptosis independent. A killerFLIP-E variant containing a scrambled c-FLIPL motif indeed induced similar cell death, suggesting the importance of the c-FLIPL residues but not of their sequence. Thus, we report the discovery of a promising synthetic peptide with novel anticancer activity in vitro and in vivo.
Collapse
|
19
|
Dual-mode enhancement of metallothionein protein with cell transduction and retention peptide fusion. J Control Release 2013; 171:193-200. [DOI: 10.1016/j.jconrel.2013.07.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 06/14/2013] [Accepted: 07/02/2013] [Indexed: 02/04/2023]
|
20
|
Datta A, Akatsu H, Heese K, Sze SK. Quantitative clinical proteomic study of autopsied human infarcted brain specimens to elucidate the deregulated pathways in ischemic stroke pathology. J Proteomics 2013; 91:556-68. [PMID: 24007662 DOI: 10.1016/j.jprot.2013.08.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 08/15/2013] [Accepted: 08/20/2013] [Indexed: 11/26/2022]
Abstract
UNLABELLED Ischemic stroke, still lacking an effective neuroprotective therapy is the third leading cause of global mortality and morbidity. Here, we have applied an 8-plex iTRAQ-based 2D-LC-MS/MS strategy to study the commonly regulated infarct proteome from three different brain regions (putamen, thalamus and the parietal lobe) of female Japanese patients. Infarcts were compared with age-, post-mortem interval- and location-matched control specimens. The iTRAQ experiment confidently identified 1520 proteins with 0.1% false discovery rate. Bioinformatics data mining and immunochemical validation of pivotal perturbed proteins revealed a global failure of the cellular energy metabolism in the infarcted tissues as seen by the parallel down-regulation of proteins related to glycolysis, pyruvate dehydrogenase complex, TCA cycle and oxidative phosphorylation. The concomitant down-regulation of all participating proteins (SLC25A11, SLC25A12, GOT2 and MDH2) of malate-aspartate shuttle might be responsible for the metabolic in-coordination between the cytosol and mitochondria resulting in the failure of energy metabolism. The levels of proteins related to reactive gliosis (VIM, GFAP) and anti-inflammatory response (ANXA1, ANXA2) showed an increasing trend. The elevation of ferritin (FTL, FTH1) may indicate an iron-mediated oxidative imbalance aggravating the mitochondrial failure and neurotoxicity. The deregulated proteins could be useful as potential therapeutic targets or biomarkers for ischemic stroke. BIOLOGICAL SIGNIFICANCE Clinical proteomics of stroke has been lagging behind other areas of clinical proteomics like Alzheimer's disease or schizophrenia. Our study is the first quantitative clinical proteomics study where iTRAQ-2D-LC-MS/MS has been utilized in the area of ischemic stroke to obtain a comparative profile of human ischemic infarcts and age-, sex-, location- and post-mortem interval-matched control brain specimens. Different pathological attributes of ischemic stroke well-known through basic and pre-clinical research such as failure of cellular energy metabolism, reactive gliosis, activation of anti-inflammatory response and aberrant iron metabolism have been observed at the bedside. Our dataset could act as a reference for similar studies done in the future using ischemic brain samples from various brain banks across the world. A meta-analysis of these studies could help to map the pathological proteome specific to ischemic stroke that will guide the scientific community to better evaluate the pros and cons of the pre-clinical models for efficacy and mechanistic studies. Infarct being the core of injury should have the most intense regulation for several key proteins involved in the pathophysiology of ischemic stroke. Hence, a part of the up-regulated proteome could leak into the general circulation that may offer candidates of interest as potential biomarkers. In support of our proposed hypothesis, we report ferritin in the current study as one of the most elevated proteins in the infarct, which has been documented as a biomarker in the context of ischemic stroke by an independent study. Overall, our approach has the potential to identify probable therapeutic targets and biomarkers in the area of ischemic stroke.
Collapse
Affiliation(s)
- Arnab Datta
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | | | | | | |
Collapse
|
21
|
Marcus D, Lichtenstein M, Saada A, Lorberboum-Galski H. Replacement of the C6ORF66 assembly factor (NDUFAF4) restores complex I activity in patient cells. Mol Med 2013; 19:124-34. [PMID: 23670274 DOI: 10.2119/molmed.2012.00343] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 05/07/2013] [Indexed: 12/21/2022] Open
Abstract
Disorders of the oxidative phosphorylation (OXPHOS) system frequently result in a severe multisystem disease with the consequence of early childhood death. Among these disorders, isolated complex I deficiency is the most frequently diagnosed, accounting for one-third of all cases of respiratory chain deficiency. We chose to focus on complex I deficiency, caused by mutation in the assembly factor chromosome 6, open reading frame 66 (C6ORF66; NADH dehydrogenase [ubiquinone] complex I assembly factor 4 [NDUFAF4]) protein. We used the approach of cell- and organelle-directed protein/enzyme replacement therapy, with the transactivator of transcription (TAT) peptide as the moiety delivery system. This step will enable us to deliver the wild-type assembly factor C6ORF66 into patient cells and their mitochondria, leading to the proper assembly and function of complex I and, as a result, to a functional OXPHOS system. We designed and constructed the TAT-ORF fusion protein by gene fusion techniques, expressed the protein in an Escherichia coli expression system and highly purified it. Our results indicate that TAT-ORF enters patients' cells and their mitochondria rapidly and efficiently. TAT-ORF is biologically active and led to an increase in complex I activity. TAT-ORF also increased the number of patient cells and improved the activity of their mitochondria. Moreover, we observed an increase in ATP production, a decrease in the content of mitochondria and a decrease in the level of reactive oxygen species. Our results suggest that this approach of protein replacement therapy for the treatment of mitochondrial disorders is a promising one.
Collapse
Affiliation(s)
- Dana Marcus
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | | | | |
Collapse
|
22
|
Vinsant S, Mansfield C, Jimenez-Moreno R, Del Gaizo Moore V, Yoshikawa M, Hampton TG, Prevette D, Caress J, Oppenheim RW, Milligan C. Characterization of early pathogenesis in the SOD1(G93A) mouse model of ALS: part I, background and methods. Brain Behav 2013; 3:335-50. [PMID: 24381807 PMCID: PMC3869677 DOI: 10.1002/brb3.143] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 03/21/2013] [Accepted: 03/22/2013] [Indexed: 11/10/2022] Open
Abstract
Charcot first described amyotrophic lateral sclerosis (ALS) in 1869; however, its causes remain largely unknown and effective, long-term treatment strategies are not available. The first mouse model of ALS was developed after the identification of mutations in the superoxide dismutase 1 (SOD1) gene in 1993, and accordingly most of our knowledge of the etiology and pathogenesis of the disease comes from studies carried out using this animal model. Although numerous preclinical trials have been conducted in the mutant SOD1 mouse models, the results have been disappointing because they did not positively translate to clinical trials. One explanation may be that current understanding of when and where pathogenesis begins is insufficient to accurately guide preclinical trials. Further characterization of these early events may provide insight into disease onset, help in the discovery of presymptomatic diagnostic disease markers, and identify novel therapeutic targets. Here, we describe the rationale, approach, and methods for our extensive analysis of early changes that included an ultrastructural examination of central and peripheral components of the neuromuscular system in the SOD1(G93A) mouse and correlated these alterations with early muscle denervation, motor dysfunction, and motoneuron death. We also provide a discussion of published work to review what is known regarding early pathology in the SOD1 mouse model of ALS. The significance of this work is that we have examined early pathology simultaneously in both the spinal cord and peripheral neuromuscular system, and the results are presented in the companion paper (Part II, Results and Discussion). Our results provide evidence as to why a thorough characterization of animal models throughout the life span is critical for a strong foundation to design preclinical trials that may produce meaningful results.
Collapse
Affiliation(s)
- Sharon Vinsant
- Department of Neurobiology and Anatomy, The Neuroscience Program and The ALS Center Winston-Salem, North Carolina
| | - Carol Mansfield
- Department of Neurobiology and Anatomy, The Neuroscience Program and The ALS Center Winston-Salem, North Carolina
| | - Ramon Jimenez-Moreno
- Department of Neurobiology and Anatomy, The Neuroscience Program and The ALS Center Winston-Salem, North Carolina
| | | | - Masaaki Yoshikawa
- Department of Neurobiology and Anatomy, The Neuroscience Program and The ALS Center Winston-Salem, North Carolina
| | | | - David Prevette
- Department of Neurobiology and Anatomy, The Neuroscience Program and The ALS Center Winston-Salem, North Carolina
| | - James Caress
- Department of Neurology and the ALS Center, Wake Forest University School of Medicine Winston-Salem, North Carolina
| | - Ronald W Oppenheim
- Department of Neurobiology and Anatomy, The Neuroscience Program and The ALS Center Winston-Salem, North Carolina
| | - Carol Milligan
- Department of Neurobiology and Anatomy, The Neuroscience Program and The ALS Center Winston-Salem, North Carolina
| |
Collapse
|
23
|
Vyas PM, Tomamichel WJ, Pride PM, Babbey CM, Wang Q, Mercier J, Martin EM, Payne RM. A TAT-frataxin fusion protein increases lifespan and cardiac function in a conditional Friedreich's ataxia mouse model. Hum Mol Genet 2012; 21:1230-47. [PMID: 22113996 PMCID: PMC3284115 DOI: 10.1093/hmg/ddr554] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 11/21/2011] [Indexed: 11/14/2022] Open
Abstract
Friedreich's ataxia (FRDA) is the most common inherited human ataxia and results from a deficiency of the mitochondrial protein, frataxin (FXN), which is encoded in the nucleus. This deficiency is associated with an iron-sulfur (Fe-S) cluster enzyme deficit leading to progressive ataxia and a frequently fatal cardiomyopathy. There is no cure. To determine whether exogenous replacement of the missing FXN protein in mitochondria would repair the defect, we used the transactivator of transcription (TAT) protein transduction domain to deliver human FXN protein to mitochondria in both cultured patient cells and a severe mouse model of FRDA. A TAT-FXN fusion protein bound iron in vitro, transduced into mitochondria of FRDA deficient fibroblasts and reduced caspase-3 activation in response to an exogenous iron-oxidant stress. Injection of TAT-FXN protein into mice with a conditional loss of FXN increased their growth velocity and mean lifespan by 53% increased their mean heart rate and cardiac output, increased activity of aconitase and reversed abnormal mitochondrial proliferation and ultrastructure in heart. These results show that a cell-penetrant peptide is capable of delivering a functional mitochondrial protein in vivo to rescue a very severe disease phenotype, and present the possibility of TAT-FXN as a protein replacement therapy.
Collapse
Affiliation(s)
- Piyush M. Vyas
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Wendy J. Tomamichel
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - P. Melanie Pride
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Clifford M. Babbey
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Qiujuan Wang
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jennifer Mercier
- Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Elizabeth M. Martin
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - R. Mark Payne
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
24
|
Papadopoulou LC, Tsiftsoglou AS. Transduction of human recombinant proteins into mitochondria as a protein therapeutic approach for mitochondrial disorders. Pharm Res 2011; 28:2639-56. [PMID: 21874377 DOI: 10.1007/s11095-011-0546-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 07/21/2011] [Indexed: 01/10/2023]
Abstract
Protein therapy is considered an alternative approach to gene therapy for treatment of genetic-metabolic disorders. Human protein therapeutics (PTs), developed via recombinant DNA technology and used for the treatment of these illnesses, act upon membrane-bound receptors to achieve their pharmacological response. On the contrary, proteins that normally act inside the cells cannot be developed as PTs in the conventional way, since they are not able to "cross" the plasma membrane. Furthermore, in mitochondrial disorders, attributed either to depleted or malfunctioned mitochondrial proteins, PTs should also have to reach the subcellular mitochondria to exert their therapeutic potential. Nowadays, there is no effective therapy for mitochondrial disorders. The development of PTs, however, via the Protein Transduction Domain (PTD) technology offered new opportunities for the deliberate delivery of human recombinant proteins inside eukaryotic subcellular organelles. To this end, mitochondrial disorders could be clinically encountered with the delivery of human mitochondrial proteins (engineered via recombinant DNA and PTD technologies) at specific intramitochondrial sites to exert their function. Overall, PTD-mediated Protein Replacement Therapy emerges as a suitable model system for the therapeutic approach for mitochondrial disorders.
Collapse
Affiliation(s)
- Lefkothea C Papadopoulou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, Thessaloniki, GR54124, Macedonia, Greece.
| | | |
Collapse
|
25
|
Marmolino D. Friedreich's ataxia: past, present and future. BRAIN RESEARCH REVIEWS 2011; 67:311-30. [PMID: 21550666 DOI: 10.1016/j.brainresrev.2011.04.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 03/29/2011] [Accepted: 04/12/2011] [Indexed: 01/26/2023]
Abstract
Friedreich's ataxia (FRDA) is an autosomal recessive inherited disorder characterized by progressive gait and limb ataxia, dysarthria, areflexia, loss of vibratory and position sense, and a progressive motor weakness of central origin. Additional features include hypertrophic cardiomyopathy and diabetes. Large GAA repeat expansions in the first intron of the FXN gene are the most common mutation underlying FRDA. Patients show severely reduced levels of a FXN-encoded mitochondrial protein called frataxin. Frataxin deficiency is associated with abnormalities of iron metabolism: decreased iron-sulfur cluster (ISC) biogenesis, accumulation of iron in mitochondria and depletion in the cytosol, enhanced cellular iron uptake. Some models have also shown reduced heme synthesis. Evidence for oxidative stress has been reported. Respiratory chain dysfunction aggravates oxidative stress by increasing leakage of electrons and the formation of superoxide. In vitro studies have demonstrated that Frataxin deficient cells not only generate more free radicals, but also show a reduced capacity to mobilize antioxidant defenses. The search for experimental drugs increasing the amount of frataxin is a very active and timely area of investigation. In cellular and in animal model systems, the replacement of frataxin function seems to alleviate the symptoms or even completely reverse the phenotype. Therefore, drugs increasing the amount of frataxin are attractive candidates for novel therapies. This review will discuss recent findings on FRDA pathogenesis, frataxin function, new treatments, as well as recent animal and cellular models. Controversial aspects are also discussed.
Collapse
Affiliation(s)
- Daniele Marmolino
- Laboratoire de Neurologie experimentale, Universite Libre de Bruxeles, Route de Lennik 808, Campus Erasme, 1070 Bruxelles, Belgium.
| |
Collapse
|
26
|
Rayapureddi JP, Tomamichel WJ, Walton ST, Payne RM. TAT fusion protein transduction into isolated mitochondria is accelerated by sodium channel inhibitors. Biochemistry 2011; 49:9470-9. [PMID: 20925426 DOI: 10.1021/bi101057v] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stringent control of ion and protein transport across the mitochondrial membranes is required to maintain mitochondrial function and biogenesis. In particular, the inner mitochondrial membrane is generally impermeable to proteins entering the matrix except via tightly regulated protein import mechanisms. Recently, cell penetrant peptides have been shown to move across the inner mitochondrial membrane in a manner suggesting an independent mechanism. HIV-1 transactivator of transcription (TAT) is an arginine-rich cell penetrant peptide, 47YGRKKRRQRRR57, which can transduce full-length proteins not only across the cell membrane but also into intracellular organelles. In this study, we investigated the ability of a TAT-containing protein to move into the mitochondrial matrix. Using a novel FACS assay for isolated, purified mitochondria, we show that TAT can deliver a modified fluorescent protein, mMDH-GFP, to the matrix of mitochondria and it is subsequently processed by the matrix peptidases. In addition, transduction of TAT-mMDH-GFP into mitochondria is independent of canonical protein import pathways as well as mitochondrial membrane potential. In direct contrast to published reports regarding the cell membrane where the sodium channel inhibitor, amiloride, blocks endocytosis and inhibits TAT transduction, TAT transduction into mitochondria is markedly increased by this same sodium channel inhibitor. These results confirm that the cell penetrant peptide, TAT, can readily transduce a protein cargo into the mitochondrial matrix. These results also demonstrate a novel role for mitochondrial sodium channels in mediating TAT transduction into mitochondria that is independent of endocytotic mechanisms. The mechanism of TAT transduction into mitochondria therefore is distinctly different from transduction across the cell membrane.
Collapse
|
27
|
Abstract
Friedreich's Ataxia is the most common inherited ataxia in man. It is a mitochondrial disease caused by severely reduced expression of the iron binding protein, frataxin. A large GAA triplet expansion in the human FRDA gene encoding this protein inhibits expression of this gene. It is inherited in an autosomal recessive pattern and typically diagnosed in childhood. The primary symptoms include severe and progressive neuropathy, and a hypertrophic cardiomyopathy that may cause death. The cardiomyopathy is difficult to treat and is frequently associated with arrhythmias, heart failure, and intolerance of cardiovascular stress, such as surgeries. Innovative approaches to therapy, such as histone deacetylase inhibitors, and enzyme replacement with cell penetrant peptide fusion proteins, hold promise for this and other similar mitochondrial disorders. This review will focus on the basic findings of this disease, and the cardiomyopathy associated with its diagnosis.
Collapse
Affiliation(s)
- R Mark Payne
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202
| |
Collapse
|
28
|
Payne RM, Pride PM, Babbey CM. Cardiomyopathy of Friedreich's ataxia: use of mouse models to understand human disease and guide therapeutic development. Pediatr Cardiol 2011; 32:366-78. [PMID: 21360265 PMCID: PMC3097037 DOI: 10.1007/s00246-011-9943-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 02/11/2011] [Indexed: 01/02/2023]
Abstract
Friedreich's ataxia is a multisystem disorder of mitochondrial function affecting primarily the heart and brain. Patients experience a severe cardiomyopathy that can progress to heart failure and death. Although the gene defect is known, the precise function of the deficient mitochondrial protein, frataxin, is not known and limits therapeutic development. Animal models have been valuable for understanding the basic events of this disease. A significant need exists to focus greater attention on the heart disease in Friedreich's ataxia, to understand its long-term outcome, and to develop new therapeutic strategies using existing medications and approaches. This review discusses some key features of the cardiomyopathy in Friedreich's ataxia and potential therapeutic developments.
Collapse
Affiliation(s)
- R Mark Payne
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut, R4302, Indianapolis, IN 46202, USA.
| | | | | |
Collapse
|
29
|
Wenz T, Williams SL, Bacman SR, Moraes CT. Emerging therapeutic approaches to mitochondrial diseases. ACTA ACUST UNITED AC 2011; 16:219-29. [PMID: 20818736 DOI: 10.1002/ddrr.109] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Mitochondrial diseases are very heterogeneous and can affect different tissues and organs. Moreover, they can be caused by genetic defects in either nuclear or mitochondrial DNA as well as by environmental factors. All of these factors have made the development of therapies difficult. In this review article, we will discuss emerging approaches to the therapy of mitochondrial disorders, some of which are targeted to specific conditions whereas others may be applicable to a more diverse group of patients.
Collapse
Affiliation(s)
- Tina Wenz
- Department of Neurology, University of Miami School of Medicine, 1095 NW 14th Terrace, Miami, FL 33136, USA
| | | | | | | |
Collapse
|
30
|
Rapoport M, Salman L, Sabag O, Patel MS, Lorberboum-Galski H. Successful TAT-mediated enzyme replacement therapy in a mouse model of mitochondrial E3 deficiency. J Mol Med (Berl) 2010; 89:161-70. [PMID: 21079907 DOI: 10.1007/s00109-010-0693-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 10/21/2010] [Accepted: 10/26/2010] [Indexed: 11/25/2022]
Abstract
Medicine today offers no cure for patients suffering from mitochondrial disorders, such as lipoamide dehydrogenase (LAD; also known as E3) deficiency, and treatment is limited to symptomatic care. LAD is one of the components of the α-ketoacid dehydrogenase complexes, which are mitochondrial multienzyme complexes crucial for the metabolism of carbohydrates and amino acids. Recently, we tested the therapeutic approach for treating mitochondrial disorders whereby the activity of multicomponent complexes in the mitochondria is restored by TAT-mediated enzyme replacement therapy (ERT). The LAD deficiency disease was used before as a proof-of-principle in vitro, in patients' cells, utilizing the TAT-LAD fusion protein. In this report, we present successful TAT-mediated ERT in an in vivo mouse model using E3-deficient mice. We demonstrate the delivery of TAT-LAD into E3-deficient mice tissues and that a single administration of TAT-LAD results in a significant increase in the enzymatic activity of the mitochondrial multienzyme complex pyruvate dehydrogenase complex within the liver, heart and, most importantly, the brain of TAT-LAD-treated E3-deficient mice. We believe that this TAT-mediated ERT approach could change the management of mitochondrial disorders and of other metabolic diseases in modern medicine.
Collapse
Affiliation(s)
- Matan Rapoport
- Department of Biochemistry and Molecular Biology, Institute for Medical Research-Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, 91120, Israel
| | | | | | | | | |
Collapse
|
31
|
Shokolenko IN, Alexeyev MF, LeDoux SP, Wilson GL. The approaches for manipulating mitochondrial proteome. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:451-461. [PMID: 20544885 PMCID: PMC3249350 DOI: 10.1002/em.20570] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Over the past decade a large volume of research data has accumulated which has established a fundamental role for mitochondria in normal cellular functioning, as well as in various pathologies. Mitochondria play a pivotal role in metabolism and energy production, and are one of the key players involved in programmed cell death. On the other hand, mitochondrial dysfunction is implicated, directly or indirectly in numerous pathological conditions including inherited mitochondrial disorders, diabetes, cardiovascular and neurodegenerative diseases, and a variety of malignancies. The ability to modulate mitochondrial function by altering the diverse protein component of this organelle may be of great value for developing future therapeutic interventions. This review will discuss approaches used to introduce proteins into mitochondria. One group of methods utilizes strategies aimed at expressing proteins from genes in the nucleus. These include overexpression of nuclear-encoded mitochondrial proteins, allotopic expression, which is the re-coding and relocation of mitochondrial genes to the nucleus for expression and subsequent delivery of their gene products to mitochondria, and xenotopic expression, which is the nuclear expression of genes coding electron transport chain components from distant species, for delivery of their products to mammalian mitochondria. Additionally, antigenomic and progenomic strategies which focus on expression of mitochondrially targeted nuclear proteins involved in the maintenance of mtDNA will be discussed. The second group of methods considered will focus on attempts to use purified proteins for mitochondrial delivery. Special consideration has been given to the complexities involved in targeting exogenous proteins to mitochondria.
Collapse
|
32
|
Foltopoulou PF, Tsiftsoglou AS, Bonovolias ID, Ingendoh AT, Papadopoulou LC. Intracellular delivery of full length recombinant human mitochondrial L-Sco2 protein into the mitochondria of permanent cell lines and SCO2 deficient patient's primary cells. Biochim Biophys Acta Mol Basis Dis 2010; 1802:497-508. [PMID: 20193760 DOI: 10.1016/j.bbadis.2010.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Revised: 01/18/2010] [Accepted: 02/22/2010] [Indexed: 10/19/2022]
Abstract
Mutations in human SCO2 gene, encoding the mitochondrial inner membrane Sco2 protein, have been found to be responsible for fatal infantile cardioencephalomyopathy and cytochrome c oxidase (COX) deficiency. One potentially fruitful therapeutic approach for this mitochondrial disorder should be considered the production of human recombinant full length L-Sco2 protein and its deliberate transduction into the mitochondria. Recombinant L-Sco2 protein, fused with TAT, a Protein Transduction Domain (PTD), was produced in bacteria and purified from inclusion bodies (IBs). Following solubilisation with l-arginine, this fusion L-Sco2 protein was transduced in cultured mammalian cells of different origin (U-87 MG, T24, K-562, and patient's primary fibroblasts) and assessed for stability, transduction into mitochondria, processing and impact on recovery of COX activity. Our results indicate that: a) l-Arg solution was effective in solubilising recombinant fusion L-Sco2 protein, derived from IBs; b) fusion L-Sco2 protein was delivered successfully via a time- and concentration-dependent process into the mitochondria of human U-87 MG and T24 cells; c) fusion L-Sco2 protein was also transduced in human K-562 cells, transiently depleted of SCO2 transcripts and thus COX deficient; transduction of this fusion protein led to partial recovery of COX activity in such cells; d) [(35)S]Methionine-labelled fusion L-Sco2 protein, produced in a cell free transcription/translation system and incubated with intact isolated mitochondria derived from K-562 cells, was efficiently processed to yield the corresponding mature Sco2 protein, thus justifying the potential of the transduced fusion L-Sco2 protein to successfully activate COX holoenzyme; and finally, e) recombinant fusion L-Sco2 protein was successfully transduced into the mitochondria of primary fibroblasts derived from SCO2/COX deficient patient and facilitated recovery of COX activity. These findings provide the rationale of delivering recombinant proteins via PTD technology as a model for therapeutic approach of mitochondrial disorders.
Collapse
Affiliation(s)
- Parthena F Foltopoulou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Macedonia, Greece
| | | | | | | | | |
Collapse
|
33
|
Marmolino D, Acquaviva F. Friedreich's Ataxia: from the (GAA)n repeat mediated silencing to new promising molecules for therapy. CEREBELLUM (LONDON, ENGLAND) 2009; 8:245-59. [PMID: 19165552 DOI: 10.1007/s12311-008-0084-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Accepted: 11/14/2008] [Indexed: 10/25/2022]
Abstract
Friedreich's ataxia (FRDA) is a neurodegenerative disease due to a pathological expansion of a GAA triplet repeat in the first intron of the FXN gene encoding for the mitochondrial protein frataxin. The expansion is responsible for most cases of FRDA through the formation of a nonusual B-DNA structure and heterochromatin conformation that determine a direct transcriptional silencing and the subsequent reduction in frataxin expression. Among other functions, frataxin is an iron chaperone central for the assembly of iron-sulfur clusters in mitochondria; its reduction is associated with iron accumulation in mitochondria, increased cellular sensitivity to oxidative stress and cell damage. There is, nowadays, no effective therapy for FRDA and current therapeutic strategies mainly act to slow down the consequences of frataxin deficiency. Therefore, drugs that are able to increase the amount of frataxin are excellent candidates for a rational approach to FRDA therapy. Recently, several drugs have been assessed for their ability to increase the amount of cellular frataxin, including human recombinant erythropoietin, histone deacetylase inhibitors, and the PPAR-gamma agonists.
Collapse
Affiliation(s)
- Daniele Marmolino
- Laboratoire de Neurologie Expérimentale, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium,
| | | |
Collapse
|
34
|
Rapoport M, Lorberboum-Galski H. TAT-based drug delivery system--new directions in protein delivery for new hopes? Expert Opin Drug Deliv 2009; 6:453-63. [PMID: 19413454 DOI: 10.1517/17425240902887029] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
There has been great progress in the use of TAT-based drug delivery systems for the delivery of different macromolecules into cells in vitro and in vivo, thus circumventing the bioavailability barrier that is a problem for so many drugs. There are many advantages to using this system, such as the ability to deliver these cargoes into all types of cells in culture and into all organs in vivo. This system can even deliver cargoes into the brain across the blood-brain barrier. In addition, the ability to target specific intracellular sub-localizations such as the nuclei, the mitochondria and lysosomes further expands the possibilities of this drug delivery system to the development of sub-cellular organelle-targeted therapy. The therapeutic applications seem almost unlimited, and the use of the TAT-based delivery system has extended from proteins to a large variety of cargoes such as oligonucleotides, imaging agents, low molecular mass drugs, nanoparticles, micelles and liposomes. In this review the most recent advances in the use of the TAT-based drug delivery system will be described, mainly discussing TAT-mediated protein delivery and the use of the TAT system for enzyme replacement therapy.
Collapse
Affiliation(s)
- Matan Rapoport
- Faculty of Medicine Hebrew University, Department of Cellular Biochemistry and Human Genetics, Jerusalem, Israel
| | | |
Collapse
|
35
|
Tao F, Johns RA. Tat-Mediated Peptide Intervention in Analgesia and Anesthesia. Drug Dev Res 2009; 71:99-105. [PMID: 20711510 DOI: 10.1002/ddr.20331] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Membrane-permeable peptide carriers are attractive drug delivery tools. Among such carriers, the protein transduction domain (PTD) of the human immunodeficiency virus-type 1 Tat protein is most frequently used and has been successfully shown to deliver a large variety of cargoes. The Tat PTD can facilitate the uptake of large, biologically active molecules into mammalian cells, and recent studies have shown that it can mediate the delivery of different cargoes into tissues throughout a living organism. Given that the Tat PTD-mediated delivery is size-independent, this technology could make previously non-applicable large molecules usable to modulate biological function in vivo and treat human diseases. It is likely that the peptide carrier-mediated intracellular delivery process encompasses multiple mechanisms, but endocytic pathways are the predominant internalization routes. Tat PTD has been successfully used in preclinical models for the study of cancer, ischemia, inflammation, analgesia, and anesthesia. Our recent studies have shown that intraperitoneally injected fusion Tat peptide Tat-PSD-95 PDZ2 can be delivered into the spinal cord to dose-dependently disrupt protein-protein interactions between PSD-95 and NMDA receptors. This peptide significantly inhibits chronic inflammatory pain and reduces the threshold for halothane anesthesia. The ability of the Tat PTD to target any cell is advantageous in some respects. However, the drug delivery system will be more attractive if we can modify the Tat PTD to deliver cargo only into desired organs to avoid possible side effects.
Collapse
Affiliation(s)
- Feng Tao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
36
|
HIV-1 Tat activates neuronal ryanodine receptors with rapid induction of the unfolded protein response and mitochondrial hyperpolarization. PLoS One 2008; 3:e3731. [PMID: 19009018 PMCID: PMC2579580 DOI: 10.1371/journal.pone.0003731] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Accepted: 10/23/2008] [Indexed: 01/12/2023] Open
Abstract
Neurologic disease caused by human immunodeficiency virus type 1 (HIV-1) is ultimately refractory to highly active antiretroviral therapy (HAART) because of failure of complete virus eradication in the central nervous system (CNS), and disruption of normal neural signaling events by virally induced chronic neuroinflammation. We have previously reported that HIV-1 Tat can induce mitochondrial hyperpolarization in cortical neurons, thus compromising the ability of the neuron to buffer calcium and sustain energy production for normal synaptic communication. In this report, we demonstrate that Tat induces rapid loss of ER calcium mediated by the ryanodine receptor (RyR), followed by the unfolded protein response (UPR) and pathologic dilatation of the ER in cortical neurons in vitro. RyR antagonism attenuated both Tat-mediated mitochondrial hyperpolarization and UPR induction. Delivery of Tat to murine CNS in vivo also leads to long-lasting pathologic ER dilatation and mitochondrial morphologic abnormalities. Finally, we performed ultrastructural studies that demonstrated mitochondria with abnormal morphology and dilated endoplasmic reticulum (ER) in brain tissue of patients with HIV-1 inflammation and neurodegeneration. Collectively, these data suggest that abnormal RyR signaling mediates the neuronal UPR with failure of mitochondrial energy metabolism, and is a critical locus for the neuropathogenesis of HIV-1 in the CNS.
Collapse
|
37
|
Rector RS, Payne RM, Ibdah JA. Mitochondrial trifunctional protein defects: clinical implications and therapeutic approaches. Adv Drug Deliv Rev 2008; 60:1488-96. [PMID: 18652860 DOI: 10.1016/j.addr.2008.04.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2008] [Accepted: 04/21/2008] [Indexed: 02/09/2023]
Abstract
The mitochondrial trifunctional protein (MTP) is a heterotrimeric protein that consists of four alpha-subunits and four beta-subunits and catalyzes three of the four chain-shortening reactions in the mitochondrial beta-oxidation of long-chain fatty acids. Families with recessively inherited MTP defects display a spectrum of maternal and fetal phenotypes. Current management of patients with MTP defects include long-term dietary therapy of fasting avoidance, low-fat/high-carbohydrate diet with restriction of long-chain fatty acid intake and substitution with medium-chain fatty acids. These dietary approaches appear promising in the short-term, but the long-term outcome of patients treated with dietary intervention is largely unknown. Potential therapeutic approaches targeted at correcting the metabolic defect will be discussed. We will discuss the potential use of protein transduction domains that cross the mitochondrial membranes for the treatment of mitochondrial disorders. In addition, we discuss the phenotypes of MTP in a heterozygous state and potential ways to intervene to increase hepatic fatty acid oxidative capacity.
Collapse
|
38
|
Yamada Y, Harashima H. Mitochondrial drug delivery systems for macromolecule and their therapeutic application to mitochondrial diseases. Adv Drug Deliv Rev 2008; 60:1439-62. [PMID: 18655816 DOI: 10.1016/j.addr.2008.04.016] [Citation(s) in RCA: 190] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Accepted: 04/21/2008] [Indexed: 11/30/2022]
Abstract
Mitochondrial dysfunction has been implicated in a variety of human disorders--the so-called mitochondrial diseases. Therefore, the organelle is a promising therapeutic drug target. In this review, we describe the key role of mitochondria in living cells, a number of mitochondrial drug delivery systems and mitochondria-targeted therapeutic strategies. In particular, we discuss mitochondrial delivery of macromolecules, such as proteins and nucleic acids. The discussion of protein delivery is limited primarily to the mitochondrial import machinery. In the section on mitochondrial gene delivery and therapy, we discuss mitochondrial diseases caused by mutations in mitochondrial DNA, several gene delivery strategies and approaches to mitochondrial gene therapy. This review also summarizes our current efforts regarding liposome-based delivery system including use of a multifunctional envelope-type nano-device (MEND) and mitochondrial liposome-based delivery as anti-cancer therapies. Furthermore, we introduce the novel MITO-Porter--a liposome-based mitochondrial delivery system that functions using a membrane-fusion mechanism.
Collapse
Affiliation(s)
- Yuma Yamada
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | | |
Collapse
|
39
|
|
40
|
TAT-mediated Delivery of LAD Restores Pyruvate Dehydrogenase Complex Activity in the Mitochondria of Patients with LAD Deficiency. Mol Ther 2008; 16:691-7. [DOI: 10.1038/mt.2008.4] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
41
|
Multifunctional envelope-type nano device (MEND) as a non-viral gene delivery system. Adv Drug Deliv Rev 2008; 60:559-71. [PMID: 18037529 DOI: 10.1016/j.addr.2007.10.007] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Accepted: 10/09/2007] [Indexed: 11/24/2022]
Abstract
In this review, we describe a key role of octaarginine (R8) in developing our new concept of "Programmed Packaging", by which we succeeded in creating a multifunctional envelope-type nano device (MEND) as a non-viral gene-delivery system. This concept can be applied not only to nuclear targeting of plasmid DNA (pDNA) but also to cytosolic delivery of functional nucleic acids such as oligonucleotides or siRNA. This concept has been extended to other organelles such as mitochondria as a foundation for innovative nanomedicine. Finally, we discuss the rate-limiting step in gene delivery by comparing non-viral and viral gene delivery systems, which clearly indicates the importance of nuclear disposition of pDNA for efficient transfection.
Collapse
|
42
|
Rapoport M, Saada A, Elpeleg O, Lorberboum-Galski H. TAT-mediated Delivery of LAD Restores Pyruvate Dehydrogenase Complex Activity in the Mitochondria of Patients with LAD Deficiency. Mol Ther 2008. [DOI: 10.1038/sj.mt.6300410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
43
|
Breunig M, Bauer S, Goepferich A. Polymers and nanoparticles: Intelligent tools for intracellular targeting? Eur J Pharm Biopharm 2008; 68:112-28. [PMID: 17804211 DOI: 10.1016/j.ejpb.2007.06.010] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2007] [Revised: 05/31/2007] [Accepted: 06/06/2007] [Indexed: 01/17/2023]
Abstract
In recent years, a new generation of drugs has entered the pharmaceutical market. Some are more potent, but some are also more toxic and thus, therapeutical efficacy may be hindered, and severe side effects may be observed, unless they are delivered to their assigned place of effect. Those targets are not only certain cell types, moreover, in cancer therapy for example, some drugs even have to be targeted to a specific cell organelle. Those targets in eukaryotic cells include among others endo- and lysosomes, mitochondria, the so-called power plants of the cells, and the biggest compartment with almost all the genetic information, the nucleus. In this review, we describe how the drugs can be directed to specific subcellular organelles and focus especially on synthetic polymers and nanoparticles as their carriers. Furthermore, we portray the progress that has been accomplished in recent years in the field of designing the carriers for efficient delivery into these target structures. Yet, we do not fail to mention the obstacles that still exist and are preventing polymeric and nanoparticular drug carrier systems from their broad application in humans.
Collapse
Affiliation(s)
- M Breunig
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetstrasse 31, Regensburg, Germany
| | | | | |
Collapse
|
44
|
Babady NE, Carelle N, Wells RD, Rouault TA, Hirano M, Lynch DR, Delatycki MB, Wilson RB, Isaya G, Puccio H. Advancements in the pathophysiology of Friedreich's Ataxia and new prospects for treatments. Mol Genet Metab 2007; 92:23-35. [PMID: 17596984 PMCID: PMC3965197 DOI: 10.1016/j.ymgme.2007.05.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2007] [Revised: 05/17/2007] [Accepted: 05/17/2007] [Indexed: 01/26/2023]
Abstract
On November 9-12, 2006, the Friedreich's Ataxia Research Alliance (FARA) and the National Institutes of Health (NIH) hosted the Third International Friedreich's Ataxia (FRDA) Scientific Conference at the NIH in Bethesda, Maryland, highlighting the exciting research leading now to a variety of clinical trials that show promise of effective treatments for this devastating disorder. Nearly 150 leading FRDA scientists from around the world discussed their new insights and findings. The presence of six pharmaceutical and biotechnology companies underscored the importance of the public-private partnership that has grown in the past years. Some of these companies are already involved in advancing promising drug compounds into clinical trials, while others are eager to help take newer discoveries through drug development and into subsequent clinical trials. National Institute of Neurological Disorders and Stroke (NINDS) Director Dr. Story Landis noted in her opening remarks for the conference that there was a "palpable sense of energy, excitement, and enthusiasm" over the scientific progress made since the FRDA gene was discovered over 10 years ago.
Collapse
Affiliation(s)
- Ngolela E. Babady
- Departments of Pediatric & Adolescent Medicine and Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Nadege Carelle
- Institut de Genetique et de Biologie Moleculaire et Cellulaire, CNRS/INSERM, Universite Louis Pasteur, Strasbourg, France
| | - Robert D. Wells
- Institute of Biosciences and Technology, Texas A&M University System HSC, Houston, TX 77030, USA
| | - Tracey A. Rouault
- Cell Biology and Metabolism Branch, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Michio Hirano
- Department of Neurology, Columbia University Medical Center, New York, New York 10032, USA
| | - David R. Lynch
- Department of Neurology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Martin B. Delatycki
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Childrens Research Institute, Royal Children’s Hospital, Melbourne, Australia
| | - Robert B. Wilson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19106, USA
| | - Grazia Isaya
- Departments of Pediatric & Adolescent Medicine and Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
- Corresponding authors: Tel: +1-507-266-0110 (G. Isaya) and + 33-3-8865-3416 (H. Puccio); Fax: +1 5072669315 (G. Isaya) and +33-3-8865-3264 (H. Puccio); (G. Isaya) and (H. Puccio)
| | - Hélène Puccio
- Institut de Genetique et de Biologie Moleculaire et Cellulaire, CNRS/INSERM, Universite Louis Pasteur, Strasbourg, France
- Corresponding authors: Tel: +1-507-266-0110 (G. Isaya) and + 33-3-8865-3416 (H. Puccio); Fax: +1 5072669315 (G. Isaya) and +33-3-8865-3264 (H. Puccio); (G. Isaya) and (H. Puccio)
| |
Collapse
|
45
|
van Vlerken LE, Vyas TK, Amiji MM. Poly(ethylene glycol)-modified nanocarriers for tumor-targeted and intracellular delivery. Pharm Res 2007; 24:1405-14. [PMID: 17393074 DOI: 10.1007/s11095-007-9284-6] [Citation(s) in RCA: 409] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2006] [Accepted: 02/27/2007] [Indexed: 01/29/2023]
Abstract
The success of anti-cancer therapies largely depends on the ability of the therapeutics to reach their designated cellular and intracellular target sites, while minimizing accumulation and action at non-specific sites. Surface modification of nanoparticulate carriers with poly(ethylene glycol) (PEG)/poly(ethylene oxide) (PEO) has emerged as a strategy to enhance solubility of hydrophobic drugs, prolong circulation time, minimize non-specific uptake, and allow for specific tumor-targeting through the enhanced permeability and retention effect. Furthermore, PEG/PEO modification has emerged as a platform for incorporation of active targeting ligands, thereby providing the drug and gene carriers with specific tumor-targeting properties through a flexible tether. This review focuses on the recent developments surrounding such PEG/PEO-surface modification of polymeric nanocarriers to promote tumor-targeting capabilities, thereby enhancing efficacy of anti-cancer therapeutic strategies.
Collapse
Affiliation(s)
- Lilian E van Vlerken
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, 110 Mugar Life Sciences Building, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
46
|
Norman JP, Perry SW, Kasischke KA, Volsky DJ, Gelbard HA. HIV-1 trans activator of transcription protein elicits mitochondrial hyperpolarization and respiratory deficit, with dysregulation of complex IV and nicotinamide adenine dinucleotide homeostasis in cortical neurons. THE JOURNAL OF IMMUNOLOGY 2007; 178:869-76. [PMID: 17202348 DOI: 10.4049/jimmunol.178.2.869] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
HIV-1 causes a common, progressive neurological disorder known as HIV-associated dementia (HAD). The prevalence of this disorder has increased despite the use of highly active antiretroviral therapy, and its underlying pathogenesis remains poorly understood. However, evidence suggests that some aspects of HAD may be reversible. To model the reversible aspects of HAD, we have used the HIV-1 neurotoxin trans activator of transcription protein (Tat) to investigate nonlethal changes in cultured neurons. Exposure of rodent cortical neurons to sublethal concentrations of Tat elicits mitochondrial hyperpolarization. In this study, we used the cationic lipophilic dye rhodamine 123 to confirm this observation, and then performed follow-up studies to examine the mechanism involved. In intact neurons, we found Tat elicited a rapid drop in internal mitochondrial pH, and addition of Tat to purified mitochondrial extracts inhibited complex IV of the electron transport chain. To correlate enzyme activity in mitochondrial extracts with results in intact cells, we measured neuronal respiration following Tat exposure. Cortical neurons demonstrated decreased respiration upon Tat treatment, consistent with inhibition of complex IV. We examined mitochondrial Ca(2+) homeostasis using a mitochondrial targeted enhanced yellow fluorescent protein-calmodulin construct. We detected a decrease in mitochondrial calcium concentration following exposure to Tat. Finally, we measured the energy intermediate NAD(P)H after Tat treatment, and found a 20% decrease in the autofluorescence. Based on these findings, we suggest that decreased NADPH and calcium concentration contribute to subsequent respiratory decline after exposure to Tat, with detrimental effects on neuronal signaling.
Collapse
Affiliation(s)
- John P Norman
- Department of Neurology (Child Neurology Division), Center for Aging and Developmental Biology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| | | | | | | | | |
Collapse
|
47
|
Yamada Y, Akita H, Kogure K, Kamiya H, Harashima H. Mitochondrial drug delivery and mitochondrial disease therapy--an approach to liposome-based delivery targeted to mitochondria. Mitochondrion 2006; 7:63-71. [PMID: 17296332 DOI: 10.1016/j.mito.2006.12.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2006] [Accepted: 10/06/2006] [Indexed: 01/09/2023]
Abstract
Recent progress in genetics and molecular biology has provided useful information regarding the molecular mechanisms associated with the mitochondrial diseases. Genetic approaches were initiated in the late 1980s to clarify the gene responsible for various mitochondrial diseases, and information concerning genetic mutations is currently used in the diagnosis of mitochondrial diseases. Moreover, it was also revealed that mitochondria play a central role in apoptosis, or programmed cell death, which is closely related to the loss of physiological functions of tissues. Therefore, drug therapies targeted to the mitochondria would be highly desirable. In spite of the huge amount of mechanism-based studies of mitochondrial diseases, effective therapies have not yet been established mainly because of the lack of an adequate delivery system. To date, numerous investigators have attempted to establish a mitochondrial drug delivery system. However, many problems remain to be overcome before a clinical application can be achieved. To fulfill a drug delivery targeted to mitochondria, we first need to establish a method to encapsulate various drugs, proteins, peptides, and genes into a drug carrier depending on their physical characteristics. Second, we need to target it to a specific cell. Finally, multi-processes of intracellular trafficking should be sophisticatedly regulated so as to release a drug carrier from the endosome to the cytosol, and thereafter to deliver to the mitochondria. In this review, we describe the current state of the development of mitochondrial drug delivery systems, and discuss the advantage and disadvantage of each system. Our current efforts to develop an efficient method for the packaging of macromolecules and regulating intracellular trafficking are also summarized. Furthermore, novel concept of "Regulation of intramitochondrial trafficking" is proposed herein as a future challenge to the development of a mitochondrial drug delivery system.
Collapse
Affiliation(s)
- Yuma Yamada
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Sapporo 060-0812, Japan
| | | | | | | | | |
Collapse
|
48
|
Abstract
Intracellular delivery of various drugs, including DNA, and drug carriers can sharply increase the efficiency of various treatment protocols. However, the receptor-mediated endocytosis of drugs, drug carriers, and DNA results in their lysosomal delivery and significant degradation. The problem can be solved and therapy efficacy still further increased if the approaches for direct intracytoplasmic delivery that bypass the endocytic pathway are developed. This is especially important for many anticancer drugs (proapoptotic drugs whose primary action site is the mitochondrial membrane) and gene therapy (nuclear or mitochondrial genomes should be targeted). This review considers several current approaches for intracellular drug delivery: the use of pH-sensitive liposomes, the use of cell-penetrating proteins and peptides, and the use of immunoliposomes targeting intracellular antigens. Among intracellular targets, nuclei (gene therapy), mitochondria (proapoptotic cancer therapy and targeting of the mitochondrial genome), and lysosomes (lysosomal targeting of enzymes for the therapy of the lysosomal storage diseases) are considered. Examples of successful intracellular and organelle-specific delivery of biologically active molecules, including DNA, are presented; unanswered questions, challenges, and future trends are also discussed.
Collapse
Affiliation(s)
- Vladimir P Torchilin
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115, USA.
| |
Collapse
|
49
|
Khan SM, Smigrodzki RM, Swerdlow RH. Cell and animal models of mtDNA biology: progress and prospects. Am J Physiol Cell Physiol 2006; 292:C658-69. [PMID: 16899549 DOI: 10.1152/ajpcell.00224.2006] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The past two decades have witnessed an evolving understanding of the mitochondrial genome's (mtDNA) role in basic biology and disease. From the recognition that mutations in mtDNA can be responsible for human disease to recent efforts showing that mtDNA mutations accumulate over time and may be responsible for some phenotypes of aging, the field of mitochondrial genetics has greatly benefited from the creation of cell and animal models of mtDNA mutation. In this review, we critically discuss the past two decades of efforts and insights gained from cell and animal models of mtDNA mutation. We attempt to reconcile the varied and at times contradictory findings by highlighting the various methodologies employed and using human mtDNA disease as a guide to better understanding of cell and animal mtDNA models. We end with a discussion of scientific and therapeutic challenges and prospects for the future of mtDNA transfection and gene therapy.
Collapse
Affiliation(s)
- Shaharyar M Khan
- Gencia Corp., 706 B Forrest St., Charlottesville, VA 22903, USA.
| | | | | |
Collapse
|
50
|
Zheng N, Huang B, Xu J, Huang S, Chen J, Hu X, Ying K, Yu X. Enzymatic and physico-chemical characteristics of recombinant cMDH and mMDH of Clonorchis sinensis. Parasitol Res 2006; 99:174-80. [PMID: 16541263 DOI: 10.1007/s00436-005-0121-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2005] [Accepted: 12/19/2005] [Indexed: 11/29/2022]
Abstract
The cytosol and mitochondrial malate dehydrogenases (MDHs, EC 1.1.1.37) of Clonorchis sinensis were expressed in Escherichia coli as a fusion protein with a 6xHis and GST tag, respectively. The cytosol MDH of Clonorchis sinensis (Cs-cMDH) has higher resistibility to acid than mitochondrial MDH (Cs-mMDH). The Cs-cMDH also has higher heat resistibility and thermal stability than Cs-mMDH. Although there is only 22.8% identity between the amino acid sequences of Cs-cMDH and Cs-mMDH, they share several conserved residues. There are some differences between the circular dichroism spectra of Cs-cMDH and Cs-mMDH, but they have approximate percentages of helix. 4,4'-Bisdimethylamino diphenylcarbinol can decrease the Cs-mMDH activity but not the Cs-cMDH activity. Paraziquantel, metronidazole and albendazole did not inhibit the enzymes' activity, but adenosine 5'-monophosphate showed competitive inhibition to enzyme, with the Ki for Cs-cMDH and Cs-mMDH being 2.81 and 0.49 mM, respectively.
Collapse
Affiliation(s)
- Nancai Zheng
- Center for Disease Control and Prevention of Jiangmen City, Jiangmen, 529020, Guangdong Province, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|