1
|
Maji D, Miguela V, Cameron AD, Campbell DA, Sasset L, Yao X, Thompson AT, Sussman C, Yang D, Miller R, Drozdz MM, Liberatore RA. Enhancing In Vivo Electroporation Efficiency through Hyaluronidase: Insights into Plasmid Distribution and Optimization Strategies. Pharmaceutics 2024; 16:547. [PMID: 38675208 PMCID: PMC11053992 DOI: 10.3390/pharmaceutics16040547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Electroporation (EP) stands out as a promising non-viral plasmid delivery strategy, although achieving optimal transfection efficiency in vivo remains a challenge. A noteworthy advancement in the field of in vivo EP is the application of hyaluronidase, an enzyme with the capacity to degrade hyaluronic acid in the extracellular matrix, which thereby enhances DNA transfer efficiency by 2- to 3-fold. This paper focuses on elucidating the mechanism of hyaluronidase's impact on transfection efficiency. We demonstrate that hyaluronidase promotes a more uniform distribution of plasmid DNA (pDNA) within skeletal muscle. Additionally, our study investigates the effect of the timing of hyaluronidase pretreatment on EP efficiency by including time intervals of 0, 5, and 30 min between hyaluronidase treatment and the application of pulses. Serum levels of the pDNA-encoded transgene reveal a minimal influence of the hyaluronidase pretreatment time on the final serum protein levels following delivery in both mice and rabbit models. Leveraging bioimpedance measurements, we capture morphological changes in muscle induced by hyaluronidase treatment, which result in a varied pDNA distribution. Subsequently, these findings are employed to optimize EP electrical parameters following hyaluronidase treatment in animal models. This paper offers novel insights into the potential of hyaluronidase in enhancing the effectiveness of in vivo EP, as well as guides optimized electroporation strategies following hyaluronidase use.
Collapse
Affiliation(s)
- Debnath Maji
- RenBio Inc., Long Island City, New York, NY 11101, USA
| | - Verónica Miguela
- RenBio Inc., Long Island City, New York, NY 11101, USA
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas—Universidad Miguel Hernández de Elche, Sant Joan d’Alacant, 03550 Alicante, Spain
| | | | | | - Linda Sasset
- RenBio Inc., Long Island City, New York, NY 11101, USA
| | - Xin Yao
- RenBio Inc., Long Island City, New York, NY 11101, USA
| | | | | | - David Yang
- RenBio Inc., Long Island City, New York, NY 11101, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Robert Miller
- RenBio Inc., Long Island City, New York, NY 11101, USA
| | | | | |
Collapse
|
2
|
Spugnini EP, Condello M, Crispi S, Baldi A. Electroporation in Translational Medicine: From Veterinary Experience to Human Oncology. Cancers (Basel) 2024; 16:1067. [PMID: 38473422 DOI: 10.3390/cancers16051067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/20/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
Electroporation (EP) is a broadly accepted procedure that, through the application of electric pulses with appropriate amplitudes and waveforms, promotes the delivery of anticancer molecules in various oncology therapies. EP considerably boosts the absorptivity of targeted cells to anticancer molecules of different natures, thus upgrading their effectiveness. Its use in veterinary oncology has been widely explored, and some applications, such as electrochemotherapy (ECT), are currently approved as first-line treatments for several neoplastic conditions. Other applications include irreversible electroporation and EP-based cancer vaccines. In human oncology, EP is still mostly restricted to therapies for cutaneous tumors and the palliation of cutaneous and visceral metastases of malignant tumors. Fields where veterinary experience could help smooth the clinical transition to humans include intraoperative EP, interventional medicine and cancer vaccines. This article recapitulates the state of the art of EP in veterinary and human oncology, recounting the most relevant results to date.
Collapse
Affiliation(s)
| | | | - Stefania Crispi
- Institute of Biosciences and BioResources-UOS Naples CNR, Via P. Castellino 111, 80131 Naples, Italy
| | - Alfonso Baldi
- Biopulse Srl, 00144 Rome, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Campania University "Luigi Vanvitelli", 81100 Caserta, Italy
| |
Collapse
|
3
|
Pertinez H, Kaushik A, Curley P, Arshad U, El-Khateeb E, Li SY, Tasneen R, Sharp J, Kijak E, Herriott J, Neary M, Noë M, Flexner C, Nuermberger E, Owen A, Ammerman NC. Hyaluronidase impacts exposures of long-acting injectable paliperidone palmitate in rodent models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.03.583160. [PMID: 38496644 PMCID: PMC10942359 DOI: 10.1101/2024.03.03.583160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
A significant challenge in the development of long-acting injectable drug formulations, especially for anti-infective agents, is delivering an efficacious dose within a tolerable injection volume. Co-administration of the extracellular matrix-degrading enzyme hyaluronidase can increase maximum tolerable injection volumes but is untested for this benefit with long-acting injectable formulations. One concern is that hyaluronidase could potentially alter the tissue response surrounding an injection depot, a response known to be important for drug release kinetics of long-acting injectable formulations. The objective of this pilot study was to evaluate the impact of co-administration of hyaluronidase on the drug release kinetics, pharmacokinetic profiles, and injection site histopathology of the long-acting injectable paliperidone palmitate for up to four weeks following intramuscular injection in mouse and rat models. In both species, co-administration of hyaluronidase increased paliperidone plasma exposures the first week after injection but did not negate the overall long-acting release nature of the formulation. Hyaluronidase-associated modification of the injection site depot was observed in mice but not in rats. These findings suggest that further investigation of hyaluronidase with long-acting injectable agents is warranted.
Collapse
Affiliation(s)
- Henry Pertinez
- Center of Excellence for Long-Acting Technologies (CELT), William Henry Duncan Building, University of Liverpool, Crown Street, Liverpool L7 8TX, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, William Henry Duncan Building, Crown Street, Liverpool L69 7BE, UK
| | - Amit Kaushik
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD, 21287, USA
| | - Paul Curley
- Center of Excellence for Long-Acting Technologies (CELT), William Henry Duncan Building, University of Liverpool, Crown Street, Liverpool L7 8TX, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, William Henry Duncan Building, Crown Street, Liverpool L69 7BE, UK
| | - Usman Arshad
- Center of Excellence for Long-Acting Technologies (CELT), William Henry Duncan Building, University of Liverpool, Crown Street, Liverpool L7 8TX, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, William Henry Duncan Building, Crown Street, Liverpool L69 7BE, UK
| | - Eman El-Khateeb
- Center of Excellence for Long-Acting Technologies (CELT), William Henry Duncan Building, University of Liverpool, Crown Street, Liverpool L7 8TX, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, William Henry Duncan Building, Crown Street, Liverpool L69 7BE, UK
| | - Si-Yang Li
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD, 21287, USA
| | - Rokeya Tasneen
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD, 21287, USA
| | - Joanne Sharp
- Center of Excellence for Long-Acting Technologies (CELT), William Henry Duncan Building, University of Liverpool, Crown Street, Liverpool L7 8TX, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, William Henry Duncan Building, Crown Street, Liverpool L69 7BE, UK
| | - Edyta Kijak
- Center of Excellence for Long-Acting Technologies (CELT), William Henry Duncan Building, University of Liverpool, Crown Street, Liverpool L7 8TX, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, William Henry Duncan Building, Crown Street, Liverpool L69 7BE, UK
| | - Joanne Herriott
- Center of Excellence for Long-Acting Technologies (CELT), William Henry Duncan Building, University of Liverpool, Crown Street, Liverpool L7 8TX, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, William Henry Duncan Building, Crown Street, Liverpool L69 7BE, UK
| | - Megan Neary
- Center of Excellence for Long-Acting Technologies (CELT), William Henry Duncan Building, University of Liverpool, Crown Street, Liverpool L7 8TX, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, William Henry Duncan Building, Crown Street, Liverpool L69 7BE, UK
| | - Michaël Noë
- Department of Pathology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands
| | - Charles Flexner
- Departments of Medicine and Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Eric Nuermberger
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD, 21287, USA
| | - Andrew Owen
- Center of Excellence for Long-Acting Technologies (CELT), William Henry Duncan Building, University of Liverpool, Crown Street, Liverpool L7 8TX, UK
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, William Henry Duncan Building, Crown Street, Liverpool L69 7BE, UK
| | - Nicole C. Ammerman
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD, 21287, USA
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD Rotterdam, Netherlands
| |
Collapse
|
4
|
Ueno S, Seino Y, Hidaka S, Nakatani M, Hitachi K, Murao N, Maeda Y, Fujisawa H, Shibata M, Takayanagi T, Iizuka K, Yabe D, Sugimura Y, Tsuchida K, Hayashi Y, Suzuki A. Blockade of glucagon increases muscle mass and alters fiber type composition in mice deficient in proglucagon-derived peptides. J Diabetes Investig 2023; 14:1045-1055. [PMID: 37300240 PMCID: PMC10445200 DOI: 10.1111/jdi.14032] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023] Open
Abstract
AIMS/INTRODUCTION Glucagon is secreted from pancreatic α-cells and plays an important role in amino acid metabolism in liver. Various animal models deficient in glucagon action show hyper-amino acidemia and α-cell hyperplasia, indicating that glucagon contributes to feedback regulation between the liver and the α-cells. In addition, both insulin and various amino acids, including branched-chain amino acids and alanine, participate in protein synthesis in skeletal muscle. However, the effect of hyperaminoacidemia on skeletal muscle has not been investigated. In the present study, we examined the effect of blockade of glucagon action on skeletal muscle using mice deficient in proglucagon-derived peptides (GCGKO mice). MATERIALS AND METHODS Muscles isolated from GCGKO and control mice were analyzed for their morphology, gene expression and metabolites. RESULTS GCGKO mice showed muscle fiber hypertrophy, and a decreased ratio of type IIA and an increased ratio of type IIB fibers in the tibialis anterior. The expression levels of myosin heavy chain (Myh) 7, 2, 1 and myoglobin messenger ribonucleic acid were significantly lower in GCGKO mice than those in control mice in the tibialis anterior. GCGKO mice showed a significantly higher concentration of arginine, asparagine, serine and threonine in the quadriceps femoris muscles, and also alanine, aspartic acid, cysteine, glutamine, glycine and lysine, as well as four amino acids in gastrocnemius muscles. CONCLUSIONS These results show that hyperaminoacidemia induced by blockade of glucagon action in mice increases skeletal muscle weight and stimulates slow-to-fast transition in type II fibers of skeletal muscle, mimicking the phenotype of a high-protein diet.
Collapse
Affiliation(s)
- Shinji Ueno
- Departments of Endocrinology, Diabetes and MetabolismFujita Health University School of MedicineToyoakeAichiJapan
| | - Yusuke Seino
- Departments of Endocrinology, Diabetes and MetabolismFujita Health University School of MedicineToyoakeAichiJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoKyotoJapan
| | - Shihomi Hidaka
- Departments of Endocrinology, Diabetes and MetabolismFujita Health University School of MedicineToyoakeAichiJapan
| | - Masashi Nakatani
- Faculty of RehabilitationSeijoh UniversityTokaiAichiJapan
- Institute for Comprehensive Medical ScienceFujita Health UniversityToyoakeAichiJapan
| | - Keisuke Hitachi
- Institute for Comprehensive Medical ScienceFujita Health UniversityToyoakeAichiJapan
| | - Naoya Murao
- Departments of Endocrinology, Diabetes and MetabolismFujita Health University School of MedicineToyoakeAichiJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoKyotoJapan
| | - Yasuhiro Maeda
- Open Facility CenterFujita Health UniversityToyoakeAichiJapan
| | - Haruki Fujisawa
- Departments of Endocrinology, Diabetes and MetabolismFujita Health University School of MedicineToyoakeAichiJapan
| | - Megumi Shibata
- Departments of Endocrinology, Diabetes and MetabolismFujita Health University School of MedicineToyoakeAichiJapan
| | - Takeshi Takayanagi
- Departments of Endocrinology, Diabetes and MetabolismFujita Health University School of MedicineToyoakeAichiJapan
| | - Katsumi Iizuka
- Department of Clinical NutritionFujita Health UniversityToyoakeAichiJapan
| | - Daisuke Yabe
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoKyotoJapan
- Department of Diabetes, Endocrinology and MetabolismGifu University Graduate School of MedicineGifuGifuJapan
- Department of Rheumatology and Clinical ImmunologyGifu University Graduate School of MedicineGifuGifuJapan
- Center for One Medicine Innovative Translational ResearchGifu University Graduate School of MedicineGifuGifuJapan
- Center for Healthcare Information TechnologyTokai National Higher Education and Research SystemNagoyaAichiJapan
- Division of Molecular and Metabolic MedicineKobe University Graduate School of MedicineKobeHyogoJapan
| | - Yoshihisa Sugimura
- Departments of Endocrinology, Diabetes and MetabolismFujita Health University School of MedicineToyoakeAichiJapan
| | - Kunihiro Tsuchida
- Institute for Comprehensive Medical ScienceFujita Health UniversityToyoakeAichiJapan
| | - Yoshitaka Hayashi
- Department of Endocrinology, Research Institute of Environmental MedicineNagoya UniversityNagoyaAichiJapan
- Department of EndocrinologyNagoya University Graduate School of MedicineNagoyaAichiJapan
| | - Atsushi Suzuki
- Departments of Endocrinology, Diabetes and MetabolismFujita Health University School of MedicineToyoakeAichiJapan
| |
Collapse
|
5
|
Kubat GB, Bouhamida E, Ulger O, Turkel I, Pedriali G, Ramaccini D, Ekinci O, Ozerklig B, Atalay O, Patergnani S, Nur Sahin B, Morciano G, Tuncer M, Tremoli E, Pinton P. Mitochondrial dysfunction and skeletal muscle atrophy: Causes, mechanisms, and treatment strategies. Mitochondrion 2023; 72:33-58. [PMID: 37451353 DOI: 10.1016/j.mito.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Skeletal muscle, which accounts for approximately 40% of total body weight, is one of the most dynamic and plastic tissues in the human body and plays a vital role in movement, posture and force production. More than just a component of the locomotor system, skeletal muscle functions as an endocrine organ capable of producing and secreting hundreds of bioactive molecules. Therefore, maintaining healthy skeletal muscles is crucial for supporting overall body health. Various pathological conditions, such as prolonged immobilization, cachexia, aging, drug-induced toxicity, and cardiovascular diseases (CVDs), can disrupt the balance between muscle protein synthesis and degradation, leading to skeletal muscle atrophy. Mitochondrial dysfunction is a major contributing mechanism to skeletal muscle atrophy, as it plays crucial roles in various biological processes, including energy production, metabolic flexibility, maintenance of redox homeostasis, and regulation of apoptosis. In this review, we critically examine recent knowledge regarding the causes of muscle atrophy (disuse, cachexia, aging, etc.) and its contribution to CVDs. Additionally, we highlight the mitochondrial signaling pathways involvement to skeletal muscle atrophy, such as the ubiquitin-proteasome system, autophagy and mitophagy, mitochondrial fission-fusion, and mitochondrial biogenesis. Furthermore, we discuss current strategies, including exercise, mitochondria-targeted antioxidants, in vivo transfection of PGC-1α, and the potential use of mitochondrial transplantation as a possible therapeutic approach.
Collapse
Affiliation(s)
- Gokhan Burcin Kubat
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey.
| | - Esmaa Bouhamida
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Gaia Pedriali
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Daniela Ramaccini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Ozgur Ekinci
- Department of Pathology, Gazi University, 06500 Ankara, Turkey
| | - Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Ozbeyen Atalay
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Simone Patergnani
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Beyza Nur Sahin
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Giampaolo Morciano
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Elena Tremoli
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Paolo Pinton
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
6
|
Hughes DC, Hardee JP, Waddell DS, Goodman CA. CORP: Gene delivery into murine skeletal muscle using in vivo electroporation. J Appl Physiol (1985) 2022; 133:41-59. [PMID: 35511722 DOI: 10.1152/japplphysiol.00088.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The strategy of gene delivery into skeletal muscles has provided exciting avenues in identifying new potential therapeutics towards muscular disorders and addressing basic research questions in muscle physiology through overexpression and knockdown studies. In vivo electroporation methodology offers a simple, rapidly effective technique for the delivery of plasmid DNA into post-mitotic skeletal muscle fibers and the ability to easily explore the molecular mechanisms of skeletal muscle plasticity. The purpose of this review is to describe how to robustly electroporate plasmid DNA into different hindlimb muscles of rodent models. Further, key parameters (e.g., voltage, hyaluronidase, plasmid concentration) which contribute to the successful introduction of plasmid DNA into skeletal muscle fibers will be discussed. In addition, details on processing tissue for immunohistochemistry and fiber cross-sectional area (CSA) analysis will be outlined. The overall goal of this review is to provide the basic and necessary information needed for successful implementation of in vivo electroporation of plasmid DNA and thus open new avenues of discovery research in skeletal muscle physiology.
Collapse
Affiliation(s)
- David C Hughes
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Justin P Hardee
- Centre for Muscle Research (CMR), Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - David S Waddell
- Department of Biology, University of North Florida, Jacksonville, FL, United States
| | - Craig A Goodman
- Centre for Muscle Research (CMR), Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| |
Collapse
|
7
|
Boye C, Arpag S, Francis M, DeClemente S, West A, Heller R, Bulysheva A. Reduction of plasmid vector backbone length enhances reporter gene expression. Bioelectrochemistry 2022; 144:107981. [PMID: 34847374 DOI: 10.1016/j.bioelechem.2021.107981] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 09/18/2021] [Accepted: 10/07/2021] [Indexed: 11/21/2022]
Abstract
Gene therapy has a wide range of applications for various types of pathologies. Viral methods of gene delivery provide high levels of gene expression but have various safety concerns. Non-viral methods are largely known to provide lower levels of expression. We aim to address this issue by using plasmid DNA with smaller backbones to increase gene expression levels when delivered using non-viral methods. In this study we compare gene expression levels between two vectors with firefly luciferase encoding gene insert using liposome complexes and gene electrotransfer as delivery methods. A 2-fold reduction in plasmid vector backbone size, disproportionately enhanced gene expression levels more than 10-fold in rat tenocytes in vitro, and rat myocardium in vivo, while improvements in delivery to the skin were more moderate.
Collapse
Affiliation(s)
- Carly Boye
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Sezgi Arpag
- LifeNet Health, Virginia Beach, VA, United States
| | | | - Scott DeClemente
- Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, United States
| | - Aislin West
- Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, United States
| | - Richard Heller
- Department of Medical Engineering, University of South Florida, Tampa, FL, United States
| | - Anna Bulysheva
- Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, United States
| |
Collapse
|
8
|
Holloway GP, Nickerson JG, Lally JSV, Petrick HL, Dennis KMJH, Jain SS, Alkhateeb H, Bonen A. Co-overexpression of CD36 and FABPpm increases fatty acid transport additively, not synergistically, within muscle. Am J Physiol Cell Physiol 2022; 322:C546-C553. [PMID: 35138177 DOI: 10.1152/ajpcell.00435.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We aimed to determine the combined effects of over-expressing FABPpm and CD36 on skeletal muscle fatty acid transport to establish if these transport proteins function collaboratively. Electrotransfection with either FABPpm or CD36 increased their protein content at the plasma membrane (+75% and +64%), increased fatty acid transport rates +24% for FABPpm and +62% for CD36, resulting in a calculated transport efficiency of ~0.019 and ~0.053 per unit protein change for FABPpm and CD36, respectively. We subsequently used these data to determine if increasing both proteins additively or synergistically increased fatty acid transport. Co-transfection of FABPpm and CD36 simultaneously increased protein content in whole muscle (FABPpm, +46%; CD36, +45%) and at the sarcolemma (FABPpm, +41% and CD36, +42%), as well as fatty acid transport rates (+50%). Since the relative effects of changing FABPpm and CD36 content had been independently determined, we were able to a predict a change in fatty acid transport based on the overexpression of plasmalemmal transporters in the co-transfection experiments. This prediction yielded an increase in fatty acid transport of +0.984 and +1.722 pmol/mg prot/15sec for FABPpm and CD36, respectively, for a total increase of +2.96 pmol/mg prot/15sec. This calculated determination was remarkably consistent with the measured change in transport, namely +2.89 pmol/mg prot/15sec. Altogether, these data indicate that increasing CD36 and FABPpm alters fatty acid transport rates additively, but not synergistically, suggesting an independent mechanism-of-action within muscle for each transporter. This conclusion was further supported by the observation that plasmalemmal CD36 and FABPpm did not co-immunoprecipitate.
Collapse
Affiliation(s)
- Graham P Holloway
- Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canad
| | | | - James S V Lally
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Heather L Petrick
- Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canad
| | - Kaitlyn M J H Dennis
- Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canad
| | - Swati S Jain
- Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canad
| | | | - Arend Bonen
- Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canad
| |
Collapse
|
9
|
Pagant S, Liberatore RA. In Vivo Electroporation of Plasmid DNA: A Promising Strategy for Rapid, Inexpensive, and Flexible Delivery of Anti-Viral Monoclonal Antibodies. Pharmaceutics 2021; 13:1882. [PMID: 34834297 PMCID: PMC8618954 DOI: 10.3390/pharmaceutics13111882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/23/2022] Open
Abstract
Since the first approval of monoclonal antibodies by the United States Food and Drug Administration (FDA) in 1986, therapeutic antibodies have become one of the predominant classes of drugs in oncology and immunology. Despite their natural function in contributing to antiviral immunity, antibodies as drugs have only more recently been thought of as tools for combating infectious diseases. Passive immunization, or the delivery of the products of an immune response, offers near-immediate protection, unlike the active immune processes triggered by traditional vaccines, which rely on the time it takes for the host's immune system to develop an effective defense. This rapid onset of protection is particularly well suited to containing outbreaks of emerging viral diseases. Despite these positive attributes, the high cost associated with antibody manufacture and the need for a cold chain for storage and transport limit their deployment on a global scale, especially in areas with limited resources. The in vivo transfer of nucleic acid-based technologies encoding optimized therapeutic antibodies transform the body into a bioreactor for rapid and sustained production of biologics and hold great promise for circumventing the obstacles faced by the traditional delivery of antibodies. In this review, we provide an overview of the different antibody delivery strategies that are currently being developed, with particular emphasis on in vivo transfection of naked plasmid DNA facilitated by electroporation.
Collapse
|
10
|
Boye C, Arpag S, Burcus N, Lundberg C, DeClemente S, Heller R, Francis M, Bulysheva A. Cardioporation enhances myocardial gene expression in rat heart. Bioelectrochemistry 2021; 142:107892. [PMID: 34371349 DOI: 10.1016/j.bioelechem.2021.107892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 11/19/2022]
Abstract
Damage from myocardial infarction (MI) and subsequent heart failure are serious public health concerns. Current clinical treatments and therapies to treat MI damage largely do not address the regeneration of cardiomyocytes. In a previous study, we established that it is possible to promote regeneration of cardiac muscle with vascular endothelial growth factor B gene delivery directly to the ischemic myocardium. In the current study we aim to optimize cardioporation parameters to increase expression efficiency by varying electrode configuration, applied voltage, pulse length, and plasmid vector size. By using a surface monopolar electrode, optimized pulsing conditions and reducing vector size, we were able to prevent ventricular fibrillation, increase survival, reduce tissue damage, and significantly increase gene expression levels.
Collapse
Affiliation(s)
- Carly Boye
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Sezgi Arpag
- LifeNet Health, Virginia Beach, VA, United States
| | - Nina Burcus
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States
| | - Cathryn Lundberg
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States
| | - Scott DeClemente
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States
| | - Richard Heller
- Department of Medical Engineering, University of South Florida, Tampa, FL, United States
| | | | - Anna Bulysheva
- Department of Electrical and Computer Engineering, Old Dominion University, United States
| |
Collapse
|
11
|
Definition of a Novel Plasmid-Based Gene Transfection Protocol of Mammalian Skeletal Muscles by Means of In Vivo Electroporation. Int J Mol Sci 2020; 21:ijms21186494. [PMID: 32899477 PMCID: PMC7555604 DOI: 10.3390/ijms21186494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 11/17/2022] Open
Abstract
We describe an original electroporation protocol for in vivo plasmid DNA transfection. The right hind limbs of C57 mice are exposed to a specifically designed train of permeabilizing electric pulses by transcutaneous application of tailored needle electrodes, immediately after the injection of pEGFP-C1 plasmid encoding GFP (Green Fluorescente Protein). The electroporated rodents show a greater GFP expression than the controls at three different time points (4, 10, and 15 days). The electroporated muscles display only mild interstitial myositis, with a significant increase in inflammatory cell infiltrates. Finally, mild gait abnormalities are registered in electroporated mice only in the first 48 h after the treatment. This protocol has proven to be highly efficient in terms of expression levels of the construct, is easy to apply since it does not require surgical exposure of the muscle and is well tolerated by the animals because it does not cause evident morphological and functional damage to the electroporated muscle.
Collapse
|
12
|
Russell SJ, Schneider MF. Alternative signaling pathways from IGF1 or insulin to AKT activation and FOXO1 nuclear efflux in adult skeletal muscle fibers. J Biol Chem 2020; 295:15292-15306. [PMID: 32868454 DOI: 10.1074/jbc.ra120.013634] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/20/2020] [Indexed: 12/25/2022] Open
Abstract
Muscle atrophy is regulated by the balance between protein degradation and synthesis. FOXO1, a transcription factor, helps to determine this balance by activating pro-atrophic gene transcription when present in muscle fiber nuclei. Foxo1 nuclear efflux is promoted by AKT-mediated Foxo1 phosphorylation, eliminating FOXO1's atrophy-promoting effect. AKT activation can be promoted by insulin-like growth factor 1 (IGF1) or insulin via a pathway including IGF1 or insulin, phosphatidylinositol 3-kinase, and AKT. We used confocal fluorescence time-lapse imaging of FOXO1-GFP in adult isolated living muscle fibers maintained in culture to explore the effects of IGF1 and insulin on FOXO1-GFP nuclear efflux with and without pharmacological inhibitors. We observed that although AKT inhibitor blocks the IGF1- or insulin-induced effect on FOXO1 nuclear efflux, phosphatidylinositol 3-kinase inhibitors, which we show to be effective in these fibers, do not. We also found that inhibition of the protein kinase ACK1 or ATM contributes to the suppression of FOXO1 nuclear efflux after IGF1. These results indicate a novel pathway that has been unexplored in the IGF1- or insulin-induced regulation of FOXO1 and present information useful both for therapeutic interventions for muscle atrophy and for further investigative areas into insulin insensitivity and type 2 diabetes.
Collapse
Affiliation(s)
- Sarah J Russell
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Martin F Schneider
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
13
|
Andrews CD, Huang Y, Ho DD, Liberatore RA. In vivo expressed biologics for infectious disease prophylaxis: rapid delivery of DNA-based antiviral antibodies. Emerg Microbes Infect 2020; 9:1523-1533. [PMID: 32579067 PMCID: PMC7473320 DOI: 10.1080/22221751.2020.1787108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
With increasing frequency, humans are facing outbreaks of emerging infectious diseases (EIDs) with the potential to cause significant morbidity and mortality. In the most extreme instances, such outbreaks can become pandemics, as we are now witnessing with COVID-19. According to the World Health Organization, this new disease, caused by the novel coronavirus SARS-CoV-2, has already infected more than 10 million people worldwide and led to 499,913 deaths as of 29 June, 2020. How high these numbers will eventually go depends on many factors, including policies on travel and movement, availability of medical support, and, because there is no vaccine or highly effective treatment, the pace of biomedical research. Other than an approved antiviral drug that can be repurposed, monoclonal antibodies (mAbs) hold the most promise for providing a stopgap measure to lessen the impact of an outbreak while vaccines are in development. Technical advances in mAb identification, combined with the flexibility and clinical experience of mAbs in general, make them ideal candidates for rapid deployment. Furthermore, the development of mAb cocktails can provide a faster route to developing a robust medical intervention than searching for a single, outstanding mAb. In addition, mAbs are well-suited for integration into platform technologies for delivery, in which minimal components need to be changed in order to be redirected against a novel pathogen. In particular, utilizing the manufacturing and logistical benefits of DNA-based platform technologies in order to deliver one or more antiviral mAbs has the potential to revolutionize EID responses.
Collapse
Affiliation(s)
| | - Yaoxing Huang
- Aaron Diamond AIDS Research Center, New York, NY, USA.,Columbia University Vagelos College of Physicans and Surgeons, New York, NY, USA
| | - David D Ho
- Aaron Diamond AIDS Research Center, New York, NY, USA.,Columbia University Vagelos College of Physicans and Surgeons, New York, NY, USA
| | | |
Collapse
|
14
|
Induction of a local muscular dystrophy using electroporation in vivo: an easy tool for screening therapeutics. Sci Rep 2020; 10:11301. [PMID: 32647247 PMCID: PMC7347864 DOI: 10.1038/s41598-020-68135-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 06/09/2020] [Indexed: 01/19/2023] Open
Abstract
Intramuscular injection and electroporation of naked plasmid DNA (IMEP) has emerged as a potential alternative to viral vector injection for transgene expression into skeletal muscles. In this study, IMEP was used to express the DUX4 gene into mouse tibialis anterior muscle. DUX4 is normally expressed in germ cells and early embryo, and silenced in adult muscle cells where its pathological reactivation leads to Facioscapulohumeral muscular dystrophy. DUX4 encodes a potent transcription factor causing a large deregulation cascade. Its high toxicity but sporadic expression constitutes major issues for testing emerging therapeutics. The IMEP method appeared as a convenient technique to locally express DUX4 in mouse muscles. Histological analyses revealed well delineated muscle lesions 1-week after DUX4 IMEP. We have therefore developed a convenient outcome measure by quantification of the damaged muscle area using color thresholding. This method was used to characterize lesion distribution and to assess plasmid recirculation and dose–response. DUX4 expression and activity were confirmed at the mRNA and protein levels and through a quantification of target gene expression. Finally, this study gives a proof of concept of IMEP model usefulness for the rapid screening of therapeutic strategies, as demonstrated using antisense oligonucleotides against DUX4 mRNA.
Collapse
|
15
|
Bozo IY, Deev RV, Smirnov IV, Fedotov AY, Popov VK, Mironov AV, Mironova OA, Gerasimenko AY, Komlev VS. 3D Printed Gene-activated Octacalcium Phosphate Implants for Large Bone Defects Engineering. Int J Bioprint 2020; 6:275. [PMID: 33088987 PMCID: PMC7557339 DOI: 10.18063/ijb.v6i3.275] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/14/2020] [Indexed: 01/27/2023] Open
Abstract
The aim of the study was the development of three-dimensional (3D) printed gene-activated implants based on octacalcium phosphate (OCP) and plasmid DNA encoding VEGFA. The first objective of the present work involved design and fabrication of gene-activated bone substitutes based on the OCP and plasmid DNA with VEGFA gene using 3D printing approach of ceramic constructs, providing the control of its architectonics compliance to the initial digital models. X-ray diffraction, scanning electron microscopy (SEM), Fourier transform infrared spectroscopy, and compressive strength analyses were applied to investigate the chemical composition, microstructure, and mechanical properties of the experimental samples. The biodegradation rate and the efficacy of plasmid DNA delivery in vivo were assessed during standard tests with subcutaneous implantation to rodents in the next stage. The final part of the study involved substitution of segmental tibia and mandibular defects in adult pigs with 3D printed gene-activated implants. Biodegradation, osteointegration, and effectiveness of a reparative osteogenesis were evaluated with computerized tomography, SEM, and a histological examination. The combination of gene therapy and 3D printed implants manifested the significant clinical potential for effective bone regeneration in large/critical size defect cases.
Collapse
Affiliation(s)
- Ilya Y Bozo
- Department of Maxillofacial Surgery, A.I. Burnazyan Federal Medical Biophysical Center, FMBA of Russia, Moscow, Russia.,Research and Development Department, Human Stem Cells Institute, Moscow, Russia
| | - Roman V Deev
- Research and Development Department, Human Stem Cells Institute, Moscow, Russia.,Department of Pathology, I.I. Mechnikov North-Western State Medical University, Saint-Petersburg, Russia
| | - Igor V Smirnov
- A.A. Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Moscow, Russia
| | - Alexander Yu Fedotov
- A.A. Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Moscow, Russia
| | - Vladimir K Popov
- Institute of Photon Technologies of Federal Scientific Research Centre "Crystallography and Photonics," Russian Academy of Sciences, Moscow, Russia
| | - Anton V Mironov
- Institute of Photon Technologies of Federal Scientific Research Centre "Crystallography and Photonics," Russian Academy of Sciences, Moscow, Russia
| | - Olga A Mironova
- Institute of Photon Technologies of Federal Scientific Research Centre "Crystallography and Photonics," Russian Academy of Sciences, Moscow, Russia
| | - Alexander Yu Gerasimenko
- Institute for Bionic Technologies and Engineering, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Institute of Biomedical Systems, National Research University of Electronic Technology, Moscow, Russia
| | - Vladimir S Komlev
- A.A. Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Moscow, Russia.,Institute of Photon Technologies of Federal Scientific Research Centre "Crystallography and Photonics," Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
16
|
Peri D, Deville M, Poignard C, Signori E, Natalini R. Numerical optimization of plasmid DNA delivery combined with hyaluronidase injection for electroporation protocol. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2020; 186:105204. [PMID: 31760303 DOI: 10.1016/j.cmpb.2019.105204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/06/2019] [Accepted: 11/11/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND AND OBJECTIVE The paper focuses on the numerical strategies to optimize a plasmid DNA delivery protocol, which combines hyaluronidase and electroporation. METHODS A well-defined continuum mechanics model of muscle porosity and advanced numerical optimization strategies have been used, to propose a substantial improvement of a pre-existing experimental protocol of DNA transfer in mice. Our work suggests that a computational model might help in the definition of innovative therapeutic procedures, thanks to the fine tuning of all the involved experimental steps. This approach is particularly interesting in optimizing complex and costly protocols, to make in vivo DNA therapeutic protocols more effective. RESULTS Our preliminary work suggests that computational model might help in the definition of innovative therapeutic protocol, thanks to the fine tuning of all the involved operations. CONCLUSIONS This approach is particularly interesting in optimizing complex and costly protocols for which the number of degrees of freedom prevents a experimental test of the possible configuration.
Collapse
Affiliation(s)
- Daniele Peri
- CNR-IAC - National Research Council, Istituto per le Applicazioni del Calcolo "Mauro Picone" Via dei Taurini 19, Rome 00185, Italy.
| | - Manon Deville
- Team MONC, INRIA Bordeaux-Sud-Ouest, Institut de Mathématiques de Bordeaux, CNRS UMR 5251 & Université de Bordeaux, 351 cours de la Libération, Talence Cedex 33405, France
| | - Clair Poignard
- Team MONC, INRIA Bordeaux-Sud-Ouest, Institut de Mathématiques de Bordeaux, CNRS UMR 5251 & Université de Bordeaux, 351 cours de la Libération, Talence Cedex 33405, France
| | - Emanuela Signori
- CNR-IFT - National Research Council - Istituto di Farmacologia Traslazionale, Via Fosso del Cavaliere 100, Rome 00133, Italy.
| | - Roberto Natalini
- CNR-IAC - National Research Council, Istituto per le Applicazioni del Calcolo "Mauro Picone" Via dei Taurini 19, Rome 00185, Italy
| |
Collapse
|
17
|
A Laboratory IGBT-Based High-voltage Pulsed Electric Field Generator for Effective Water Diffusivity Enhancement in Chicken Meat. FOOD BIOPROCESS TECH 2019. [DOI: 10.1007/s11947-019-02360-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
18
|
Del Campo A, Contreras-Hernández I, Castro-Sepúlveda M, Campos CA, Figueroa R, Tevy MF, Eisner V, Casas M, Jaimovich E. Muscle function decline and mitochondria changes in middle age precede sarcopenia in mice. Aging (Albany NY) 2019; 10:34-55. [PMID: 29302020 PMCID: PMC5811241 DOI: 10.18632/aging.101358] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/30/2017] [Indexed: 01/02/2023]
Abstract
Sarcopenia is the degenerative loss of muscle mass and strength with aging. Although a role of mitochondrial metabolism in muscle function and in the development of many diseases has been described, the role of mitochondrial topology and dynamics in the process of muscle aging is not fully understood. This work shows a time line of changes in both mitochondrial distribution and skeletal muscle function during mice lifespan. We isolated muscle fibers from flexor digitorum brevis of mice of different ages. A fusion-like phenotype of mitochondria, together with a change in orientation perpendicular to the fiber axis was evident in the Adult group compared to Juvenile and Older groups. Moreover, an increase in the contact area between sarcoplasmic reticulum and mitochondria was evident in the same group. Together with the morphological changes, mitochondrial Ca2+ resting levels were reduced at age 10-14 months and significantly increased in the Older group. This was consistent with a reduced number of mitochondria-to-jSR pairs in the Older group compared to the Juvenile. Our results support the idea of several age-dependent changes in mitochondria that are accentuated in midlife prior to a complete sarcopenic phenotype.
Collapse
Affiliation(s)
- Andrea Del Campo
- Center for Exercise, Metabolism and Cancer, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ignacio Contreras-Hernández
- Center for Exercise, Metabolism and Cancer, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Mauricio Castro-Sepúlveda
- Department of Cellular and Molecular Biology, School of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.,Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian A Campos
- Center for Exercise, Metabolism and Cancer, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Reinaldo Figueroa
- Center for Genomics and Bioinformatics, Universidad Mayor de Chile, Santiago, Chile
| | - María Florencia Tevy
- Center for Genomics and Bioinformatics, Universidad Mayor de Chile, Santiago, Chile
| | - Verónica Eisner
- Department of Cellular and Molecular Biology, School of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariana Casas
- Center for Exercise, Metabolism and Cancer, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Physiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Enrique Jaimovich
- Center for Exercise, Metabolism and Cancer, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
19
|
BDNF is a mediator of glycolytic fiber-type specification in mouse skeletal muscle. Proc Natl Acad Sci U S A 2019; 116:16111-16120. [PMID: 31320589 DOI: 10.1073/pnas.1900544116] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) influences the differentiation, plasticity, and survival of central neurons and likewise, affects the development of the neuromuscular system. Besides its neuronal origin, BDNF is also a member of the myokine family. However, the role of skeletal muscle-derived BDNF in regulating neuromuscular physiology in vivo remains unclear. Using gain- and loss-of-function animal models, we show that muscle-specific ablation of BDNF shifts the proportion of muscle fibers from type IIB to IIX, concomitant with elevated slow muscle-type gene expression. Furthermore, BDNF deletion reduces motor end plate volume without affecting neuromuscular junction (NMJ) integrity. These morphological changes are associated with slow muscle function and a greater resistance to contraction-induced fatigue. Conversely, BDNF overexpression promotes a fast muscle-type gene program and elevates glycolytic fiber number. These findings indicate that BDNF is required for fiber-type specification and provide insights into its potential modulation as a therapeutic target in muscle diseases.
Collapse
|
20
|
Sokołowska E, Błachnio-Zabielska AU. A Critical Review of Electroporation as A Plasmid Delivery System in Mouse Skeletal Muscle. Int J Mol Sci 2019; 20:ijms20112776. [PMID: 31174257 PMCID: PMC6600476 DOI: 10.3390/ijms20112776] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/13/2022] Open
Abstract
The gene delivery to skeletal muscles is a promising strategy for the treatment of both muscular disorders (by silencing or overexpression of specific gene) and systemic secretion of therapeutic proteins. The use of a physical method like electroporation with plate or needle electrodes facilitates long-lasting gene silencing in situ. It has been reported that electroporation enhances the expression of the naked DNA gene in the skeletal muscle up to 100 times and decreases the changeability of the intramuscular expression. Coelectransfer of reporter genes such as green fluorescent protein (GFP), luciferase or beta-galactosidase allows the observation of correctly performed silencing in the muscles. Appropriate selection of plasmid injection volume and concentration, as well as electrotransfer parameters, such as the voltage, the length and the number of electrical pulses do not cause long-term damage to myocytes. In this review, we summarized the electroporation methodology as well as the procedure of electrotransfer to the gastrocnemius, tibialis, soleus and foot muscles and compare their advantages and disadvantages.
Collapse
Affiliation(s)
- Emilia Sokołowska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, 15-222 Bialystok, Poland.
| | | |
Collapse
|
21
|
Hojman P, Brolin C, Nørgaard-Christensen N, Dethlefsen C, Lauenborg B, Olsen CK, Åbom MM, Krag T, Gehl J, Pedersen BK. IL-6 release from muscles during exercise is stimulated by lactate-dependent protease activity. Am J Physiol Endocrinol Metab 2019; 316:E940-E947. [PMID: 30779630 DOI: 10.1152/ajpendo.00414.2018] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IL-6 is secreted from muscles to the circulation during high-intensity and long-duration exercise, where muscle-derived IL-6 works as an energy sensor to increase release of energy substrates from liver and adipose tissues. We investigated the mechanism involved in the exercise-mediated surge in IL-6 during exercise. Using interval-based cycling in healthy young men, swimming exercise in mice, and electrical stimulation of primary human muscle cells, we explored the role of lactate production in muscular IL-6 release during exercise. First, we observed a tight correlation between lactate production and IL-6 release during both strenuous bicycling and electrically stimulated muscle cell cultures. In mice, intramuscular injection of lactate mimicked the exercise-dependent release of IL-6, and pH buffering of lactate production during exercise attenuated IL-6 secretion. Next, we used in vivo bioimaging to demonstrate that intrinsic intramuscular proteases were activated in mice during swimming, and that blockade of protease activity blunted swimming-induced IL-6 release in mice. Last, intramuscular injection of the protease hyaluronidase resulted in dramatic increases in serum IL-6 in mice, and immunohistochemical analyses showed that intramuscular lactate and hyaluronidase injections led to release of IL-6-containing intramyocellular vesicles. We identified a pool of IL-6 located within vesicles of skeletal muscle fibers, which could be readily secreted upon protease activity. This protease-dependent release of IL-6 was initiated by lactate production, linking training intensity and lactate production to IL-6 release during strenuous exercise.
Collapse
Affiliation(s)
- Pernille Hojman
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Copenhagen University Hospital, University of Copenhagen , Copenhagen , Denmark
| | - Camilla Brolin
- Department of Cellular and Molecular Medicine, University of Copenhagen , Copenhagen , Denmark
| | - Nynne Nørgaard-Christensen
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Copenhagen University Hospital, University of Copenhagen , Copenhagen , Denmark
| | - Christine Dethlefsen
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Copenhagen University Hospital, University of Copenhagen , Copenhagen , Denmark
| | - Britt Lauenborg
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Copenhagen University Hospital, University of Copenhagen , Copenhagen , Denmark
| | - Cecilie Køllner Olsen
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Copenhagen University Hospital, University of Copenhagen , Copenhagen , Denmark
| | - Mette Marie Åbom
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Copenhagen University Hospital, University of Copenhagen , Copenhagen , Denmark
| | - Thomas Krag
- Copenhagen Neuromuscular Center, Copenhagen University Hospital, University of Copenhagen , Copenhagen , Denmark
| | - Julie Gehl
- Center for Experimental Drug and Gene Electrotransfer, Department of Oncology and Palliative Care, Zealand University Hospital , Roskilde , Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Bente Klarlund Pedersen
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Copenhagen University Hospital, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
22
|
Long Non-Coding RNA Myoparr Regulates GDF5 Expression in Denervated Mouse Skeletal Muscle. Noncoding RNA 2019; 5:ncrna5020033. [PMID: 30965639 PMCID: PMC6631233 DOI: 10.3390/ncrna5020033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle is a highly plastic tissue and decreased skeletal muscle mass (muscle atrophy) results in deteriorated motor function and perturbed body homeostasis. Myogenin promoter-associated long non-coding RNA (lncRNA) Myoparr promotes skeletal muscle atrophy caused by surgical denervation; however, the precise molecular mechanism remains unclear. Here, we examined the downstream genes of Myoparr during muscle atrophy following denervation of tibialis anterior (TA) muscles in C57BL/6J mice. Myoparr knockdown affected the expression of 848 genes. Sixty-five of the genes differentially regulated by Myoparr knockdown coded secretory proteins. Among these 65 genes identified in Myoparr-depleted skeletal muscles after denervation, we focused on the increased expression of growth/differentiation factor 5 (GDF5), an inhibitor of muscle atrophy. Myoparr knockdown led to activated bone morphogenetic protein (BMP) signaling in denervated muscles, as indicated by the increased levels of phosphorylated Smad1/5/8. Our detailed evaluation of downstream genes of Myoparr also revealed that Myoparr regulated differential gene expression between myogenic differentiation and muscle atrophy. This is the first report demonstrating the in vivo role of Myoparr in regulating BMP signaling in denervated muscles. Therefore, lncRNAs that have inhibitory activity on BMP signaling may be putative therapeutic targets for skeletal muscle atrophy.
Collapse
|
23
|
Knudsen JR, Henriquez-Olguin C, Li Z, Jensen TE. Electroporated GLUT4-7myc-GFP detects in vivo glucose transporter 4 translocation in skeletal muscle without discernible changes in GFP patterns. Exp Physiol 2019; 104:704-714. [PMID: 30710396 DOI: 10.1113/ep087545] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/01/2019] [Indexed: 12/22/2022]
Abstract
NEW FINDINGS What is the central question of this study? Resolving the mechanism(s) leading to glucose transporter 4 (GLUT4) translocation to the muscle surface membrane has great therapeutic potential. However, the measurement of GLUT4 translocation is technically challenging. Here, we asked whether electroporation of GLUT4-7myc-GFP into skeletal muscle could be used as a tool to study GLUT4 translocation in vivo. What is the main finding and its importance? By acutely inducing GLUT4-7myc-GFP expression in skeletal muscle, we verified that in vivo exercise and AICAR stimulation increased the GLUT4 presence in the sarcolemma measured as myc signal. Importantly, the increased myc signal in the sarcolemma was not accompanied by major visual changes in the distribution of the GFP signal. ABSTRACT Insulin and exercise lead to translocation of the glucose transporter 4 (GLUT4) to the surface membrane of skeletal muscle fibres. This process is pivotal for facilitating glucose uptake into skeletal muscle. To study this, a robust assay is needed to measure the translocation of GLUT4 in adult skeletal muscle directly. Here, we aimed to validate a simple GLUT4 translocation assay using a genetically encoded biosensor in mouse skeletal muscle. We transfected GLUT4-7myc-GFP into mouse muscle to study live GLUT4 movement and to evaluate GLUT4 insertion in the muscle surface membrane after in vivo running exercise and pharmacological activation of AMP-activated protein kinase (AMPK). Transfection led to expression of GLUT4-7myc-GFP that was dynamic in live flexor digitorum brevis fibres and which, upon insulin stimulation, exposed the myc epitope extracellularly. Running exercise, in addition to AMPK activation by 5-aminoimidazole-4-carboxamide ribonucleotide, induced ∼125 and ∼100% increase, respectively, in extracellularly exposure of GLUT4 in the surface membrane of tibialis anterior muscle. Interestingly, the clear increase in surface-exposed GLUT4 content induced by insulin, exercise or AMPK activation was not accompanied by any discernible reorganization of the GLUT4-GFP signal. In conclusion, we provide a detailed description of an easy-to-use translocation assay to study GLUT4 accumulation at the surface membrane induced by exercise and exercise-mimicking stimuli. Notably, our analyses revealed that increased GLUT4 surface membrane accumulation was not accompanied by a discernible change in the GLUT4 localization pattern.
Collapse
Affiliation(s)
- Jonas Roland Knudsen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Carlos Henriquez-Olguin
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Zhencheng Li
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Elbenhardt Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
24
|
Hodge BA, Zhang X, Gutierrez-Monreal MA, Cao Y, Hammers DW, Yao Z, Wolff CA, Du P, Kemler D, Judge AR, Esser KA. MYOD1 functions as a clock amplifier as well as a critical co-factor for downstream circadian gene expression in muscle. eLife 2019; 8:e43017. [PMID: 30789342 PMCID: PMC6398978 DOI: 10.7554/elife.43017] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/20/2019] [Indexed: 01/13/2023] Open
Abstract
In the present study we show that the master myogenic regulatory factor, MYOD1, is a positive modulator of molecular clock amplitude and functions with the core clock factors for expression of clock-controlled genes in skeletal muscle. We demonstrate that MYOD1 directly regulates the expression and circadian amplitude of the positive core clock factor Bmal1. We identify a non-canonical E-box element in Bmal1 and demonstrate that is required for full MYOD1-responsiveness. Bimolecular fluorescence complementation assays demonstrate that MYOD1 colocalizes with both BMAL1 and CLOCK throughout myonuclei. We demonstrate that MYOD1 and BMAL1:CLOCK work in a synergistic fashion through a tandem E-box to regulate the expression and amplitude of the muscle specific clock-controlled gene, Titin-cap (Tcap). In conclusion, these findings reveal mechanistic roles for the muscle specific transcription factor MYOD1 in the regulation of molecular clock amplitude as well as synergistic regulation of clock-controlled genes in skeletal muscle.
Collapse
Affiliation(s)
- Brian A Hodge
- Department of Physiology and Functional GenomicsUniversity of FloridaGainesvilleUnited States
| | - Xiping Zhang
- Department of Physiology and Functional GenomicsUniversity of FloridaGainesvilleUnited States
| | | | - Yi Cao
- Department of Bioinformatics and Computational BiologyGenentech IncSouth San FranciscoUnited States
| | - David W Hammers
- Department of Pharmacology and TherapeuticsUniversity of Florida Health Science CenterGainesvilleUnited States
| | - Zizhen Yao
- Allen Institute for Brain ScienceSeattleUnited States
| | - Christopher A Wolff
- Department of Physiology and Functional GenomicsUniversity of FloridaGainesvilleUnited States
| | - Ping Du
- Department of Physiology and Functional GenomicsUniversity of FloridaGainesvilleUnited States
| | - Denise Kemler
- Department of Physiology and Functional GenomicsUniversity of FloridaGainesvilleUnited States
| | - Andrew R Judge
- Department of Physical TherapyUniversity of Florida Health Science CenterGainesvilleUnited States
| | - Karyn A Esser
- Department of Physiology and Functional GenomicsUniversity of FloridaGainesvilleUnited States
| |
Collapse
|
25
|
Yeo D, Kang C, Ji LL. PGC-1α Overexpression via Local In Vivo Transfection in Mouse Skeletal Muscle. Methods Mol Biol 2019; 1966:151-161. [PMID: 31041745 DOI: 10.1007/978-1-4939-9195-2_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The overexpression of a specific protein is a common method for investigating the specific biological function of the substance and the mechanism of action. In vivo electrotransfer has been confirmed to be one of the most reliable, efficient and cost-effective way to overexpress a protein in a select biological tissue. Typically, this technique involves a physical injection of plasmid DNA followed by electric pulses across the injection site. Here, we introduce this method that we used to transfect green fluorescent protein (GFP)-tagged PGC-1α plasmid DNA into mouse tibialis anterior (TA) muscle, which attained high transfection efficiency with no muscle damage. To quantify the transfection efficiency, we also demonstrate the visualization of plasmid DNA transfected fibers via immunohistochemical staining on muscle cross sections.
Collapse
Affiliation(s)
- Dongwook Yeo
- Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Chounghun Kang
- The Department of Physical Education, Inha University, Incheon, South Korea
| | - Li Li Ji
- Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota Twin Cities, Minneapolis, MN, USA.
| |
Collapse
|
26
|
A Continuum Mechanics Model of Enzyme-Based Tissue Degradation in Cancer Therapies. Bull Math Biol 2018; 80:3184-3226. [DOI: 10.1007/s11538-018-0515-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 09/24/2018] [Indexed: 12/29/2022]
|
27
|
Jourdain M, Melly S, Summermatter S, Hatakeyama S. Mouse models of cancer-induced cachexia: Hind limb muscle mass and evoked force as readouts. Biochem Biophys Res Commun 2018; 503:2415-2420. [PMID: 29969629 DOI: 10.1016/j.bbrc.2018.06.170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 06/29/2018] [Indexed: 12/29/2022]
Abstract
The majority of patients with advanced cancer suffer from cachexia, a systemic wasting syndrome, which subsequently impacts the tolerance to anti-cancer treatments, response to therapy, quality of life, and eventually, survival. Despite a high unmet medical need, there is currently no specific remedy available for an effective treatment of cachexia and its sequelae. A key feature of cachexia is the inexorable loss of skeletal muscle mass, which constitutes a main contributor to body weight loss and progressive functional impairments. Therefore, it's crucial to identify early readouts to detect and monitor the loss of muscle mass and function to initiate appropriate treatments timely. Here, we describe experimental cancer models using mouse (syngeneic) or human (xenograft) cancer cell lines with a rapid onset of tumor growth and cachexia. These models are easier to establish, monitor and reproduce compared to the genetically engineered mouse models currently available. Moreover, we establish readouts such as hind limb muscle mass and volume, as well as evoked force and food intake measurements, to allow the evaluation of potential therapeutic agents for the early treatment of cachexia and associated impairments.
Collapse
Affiliation(s)
- Marie Jourdain
- MusculoSkeletal Diseases, Novartis Institutes for Biomedical Research, Novartis Pharma AG, CH-4002, Basel, Switzerland.
| | - Stefan Melly
- MusculoSkeletal Diseases, Novartis Institutes for Biomedical Research, Novartis Pharma AG, CH-4002, Basel, Switzerland.
| | - Serge Summermatter
- MusculoSkeletal Diseases, Novartis Institutes for Biomedical Research, Novartis Pharma AG, CH-4002, Basel, Switzerland.
| | - Shinji Hatakeyama
- MusculoSkeletal Diseases, Novartis Institutes for Biomedical Research, Novartis Pharma AG, CH-4002, Basel, Switzerland.
| |
Collapse
|
28
|
Andrews CD, Luo Y, Sun M, Yu J, Goff AJ, Glass PJ, Padte NN, Huang Y, Ho DD. In Vivo Production of Monoclonal Antibodies by Gene Transfer via Electroporation Protects against Lethal Influenza and Ebola Infections. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:74-82. [PMID: 29034261 PMCID: PMC5633264 DOI: 10.1016/j.omtm.2017.09.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/13/2017] [Indexed: 11/19/2022]
Abstract
Monoclonal antibodies (mAbs) have wide clinical utility, but global access is limited by high costs and impracticalities associated with repeated passive administration. Here, we describe an optimized electroporation-based DNA gene transfer platform technology that can be utilized for production of functional mAbs in vivo, with the potential to reduce costs and administration burdens. We demonstrate that multiple mAbs can be simultaneously expressed at protective concentrations for a protracted period of time using DNA doses and electroporation conditions that are feasible clinically. The expressed mAbs could also protect mice against lethal influenza or Ebola virus challenges. Our findings suggest that this DNA gene transfer platform technology could be a game-changing advance that expands access to effective mAb therapeutics globally.
Collapse
Affiliation(s)
- Chasity D. Andrews
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
| | - Yang Luo
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
| | - Ming Sun
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
| | - Jian Yu
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
| | - Arthur J. Goff
- US Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA
| | - Pamela J. Glass
- US Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA
| | - Neal N. Padte
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
| | - Yaoxing Huang
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
| | - David D. Ho
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
- Corresponding author: David D. Ho, Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA.
| |
Collapse
|
29
|
Holechek SA, McAfee MS, Nieves LM, Guzman VP, Manhas K, Fouts T, Bagley K, Blattman JN. Retinaldehyde dehydrogenase 2 as a molecular adjuvant for enhancement of mucosal immunity during DNA vaccination. Vaccine 2016; 34:5629-5635. [PMID: 27670072 DOI: 10.1016/j.vaccine.2016.09.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/28/2016] [Accepted: 09/14/2016] [Indexed: 12/12/2022]
Abstract
In order for vaccines to induce efficacious immune responses against mucosally transmitted pathogens, such as HIV-1, activated lymphocytes must efficiently migrate to and enter targeted mucosal sites. We have previously shown that all-trans retinoic acid (ATRA) can be used as a vaccine adjuvant to enhance mucosal CD8+ T cell responses during vaccination and improve protection against mucosal viral challenge. However, the ATRA formulation is incompatible with most recombinant vaccines, and the teratogenic potential of ATRA at high doses limits its usage in many clinical settings. We hypothesized that increasing in vivo production of retinoic acid (RA) during vaccination with a DNA vector expressing retinaldehyde dehydrogenase 2 (RALDH2), the rate-limiting enzyme in RA biosynthesis, could similarly provide enhanced programming of mucosal homing to T cell responses while avoiding teratogenic effects. Administration of a RALDH2- expressing plasmid during immunization with a HIVgag DNA vaccine resulted in increased systemic and mucosal CD8+ T cell numbers with an increase in both effector and central memory T cells. Moreover, mice that received RALDH2 plasmid during DNA vaccination were more resistant to intravaginal challenge with a recombinant vaccinia virus expressing the same HIVgag antigen (VACVgag). Thus, RALDH2 can be used as an alternative adjuvant to ATRA during DNA vaccination leading to an increase in both systemic and mucosal T cell immunity and better protection from viral infection at mucosal sites.
Collapse
Affiliation(s)
- Susan A Holechek
- Biodesign Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, United States; School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, United States; Simon A. Levin Mathematical, Computational and Modeling Sciences Center, Arizona State University, Tempe, AZ 85287-3901, United States
| | - Megan S McAfee
- Biodesign Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, United States; School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, United States
| | - Lizbeth M Nieves
- Biodesign Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, United States; School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, United States
| | - Vanessa P Guzman
- Biodesign Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, United States; School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, United States
| | - Kavita Manhas
- Biodesign Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, United States; School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, United States
| | - Timothy Fouts
- Profectus BioSciences, Inc., Baltimore, MD 21224, United States
| | - Kenneth Bagley
- Profectus BioSciences, Inc., Baltimore, MD 21224, United States
| | - Joseph N Blattman
- Biodesign Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, United States; School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, United States.
| |
Collapse
|
30
|
Akerstrom T, Vedel K, Needham J, Hojman P, Kontou E, Hellsten Y, Wojtaszewski JF. Optimizing hyaluronidase dose and plasmid DNA delivery greatly improves gene electrotransfer efficiency in rat skeletal muscle. Biochem Biophys Rep 2015; 4:342-350. [PMID: 29124223 PMCID: PMC5669402 DOI: 10.1016/j.bbrep.2015.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 10/12/2015] [Accepted: 10/20/2015] [Indexed: 11/18/2022] Open
Abstract
Transfection of rat skeletal muscle in vivo is a widely used research model. However, gene electrotransfer protocols have been developed for mice and yield variable results in rats. We investigated whether changes in hyaluronidase pre-treatment and plasmid DNA delivery can improve transfection efficiency in rat skeletal muscle. We found that pre-treating the muscle with a hyaluronidase dose suitable for rats (0.56 U/g b.w.) prior to plasmid DNA injection increased transfection efficiency by >200% whereas timing of the pre-treatment did not affect efficiency. Uniformly distributing plasmid DNA delivery across the muscle by increasing the number of plasmid DNA injections further enhanced transfection efficiency whereas increasing plasmid dose from 0.2 to 1.6 µg/g b.w. or vehicle volume had no effect. The optimized protocol resulted in ~80% (CI95%: 79–84%) transfected muscle fibers with a homogenous distribution. We also show that transfection was stable over five weeks of regular exercise or inactivity. Our findings show that species-specific plasmid DNA delivery and hyaluronidase pre-treatment greatly improves transfection efficiency in rat skeletal muscle. Parameters for effective in vivo skeletal muscle transfection are species specific. Pre-treatment with a rat-specific hyaluronidase dose greatly improves transfection efficiency. Delivering plasmid DNA more uniformly enhances transfection efficiency in rat skeletal muscle. Transfection efficiency is not improved by increasing plasmid DNA dose. Exercise training does not affect transfection stability.
Collapse
Affiliation(s)
- Thorbjorn Akerstrom
- The August Krogh Centre, Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Correspondence to: The August Krogh Centre, Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Universitetsparken 13, DK-2100 Copenhagen, Denmark. Fax: +4535320870.The August Krogh Centre, Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of CopenhagenUniversitetsparken 13CopenhagenDK-2100Denmark
| | - Kenneth Vedel
- The August Krogh Centre, Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Josefine Needham
- The August Krogh Centre, Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Pernille Hojman
- Centre of Inflammation and Metabolism, Centre for Physical Activity Research, Copenhagen University Hospital, Copenhagen, Denmark
| | - Eftychia Kontou
- The August Krogh Centre, Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Ylva Hellsten
- The August Krogh Centre, Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen F.P. Wojtaszewski
- The August Krogh Centre, Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
31
|
Zhang HH, Qin GJ, Li XL, Zhang YH, Du PJ, Zhang PY, Zhao YY, Wu J. SIRT1 overexpression in skeletal muscle in vivo induces increased insulin sensitivity and enhanced complex I but not complex II-V functions in individual subsarcolemmal and intermyofibrillar mitochondria. J Physiol Biochem 2015; 71:177-90. [PMID: 25782776 DOI: 10.1007/s13105-015-0396-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 02/20/2015] [Indexed: 01/04/2023]
Abstract
SIRT1 is known to improve insulin resistance (IR), but whether this effect is direct or not is still unclear, and this question has not been addressed in vivo in the skeletal muscle. Therefore, we sought to test if acute overexpression of SIRT1 in skeletal muscle of high-fat diet (HFD) rats in vivo would affect subsarcolemmal (SS) and intermyofibrillar (IMF) mitochondrial complexes I-V activities and antioxidant enzymes thereby improving insulin action. In vivo electrotransfer was used to overexpress SIRT1 in the skeletal muscle of rats fed HFD for 12 weeks. Skeletal muscle insulin sensitivity and downstream effects of SIRT1 on AMPK, SIRT3, and mitochondrial biogenesis were studied. Citrate synthase (CS), complexes I-V, oxidative stress, and antioxidant levels were assessed in SS and IMF mitochondria. HFD rats showed skeletal muscle IR as well as decreased SIRT1 and SIRT3 expressions, mitochondrial DNA (mtDNA), and mitochondrial biogenesis (p < 0.05). SS and IMF mitochondria displayed lower CS, complexes I-V, and antioxidant enzyme activities (p < 0.05). By contrast, moderate (~2.5 folds) SIRT1 overexpression attenuated HFD-induced skeletal muscle IR. This improvement was associated with increased AMPK, PGC-1α, SIRT3, and mtDNA expressions as well as SS and IMF mitochondrial CS and complexes I-V activities. Importantly, SIRT1 overexpression largely restored antioxidant enzyme activities and enhanced complex I but not complexes II-V functions in individual SS and IMF mitochondria. This study suggests that SIRT1 overexpression improved IR at least partly by targeting complex I functions of SS and IMF mitochondria through the activation of SIRT1 and SIRT3.
Collapse
Affiliation(s)
- Hao-Hao Zhang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, 40 Daxue Road, 450052, Zhengzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Liu S, Ma L, Tan R, Lu Q, Geng Y, Wang G, Gu Z. Safe and efficient local gene delivery into skeletal muscle via a combination of Pluronic L64 and modified electrotransfer. Gene Ther 2014; 21:558-65. [PMID: 24694536 DOI: 10.1038/gt.2014.27] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/25/2013] [Accepted: 01/24/2014] [Indexed: 11/09/2022]
Abstract
Efficient DNA electrotransfer into muscles can be achieved by combining two types of electronic pulses sequentially: short high-voltage (HV) pulse for the cell electropermeabilization and long low-voltage (LV) pulse for the DNA electrophoresis into cells. However, the voltages currently applied can still induce histological and functional damages to tissues. Pluronic L64 has been considered as a molecule possessing cell membrane-disturbing ability. For these reasons, we hope that L64 can be used as a substitute for the HV pulse in cell membrane permeabilization, and a safe LV pulse may still keep the ability to drive plasmid DNA across the permeabilized membrane. In this work, we optimized the electrotransfer parameters to establish a safe and efficient procedure using a clinically applied instrument, and found out that the critical condition for a successful combination of electrotransfer with L64 was that the injection of plasmid/L64 mixture should be applied 1 h before the electrotransfer. In addition, we revealed that the combined procedure could not efficiently transfer plasmid into solid tumor because the uncompressed plasmid may rapidly permeate the leaky tumor vessels and flow away. Altogether, the results demonstrate that the combined procedure has the potential for plasmid-based gene therapy through safe and efficient local gene delivery into skeletal muscles.
Collapse
Affiliation(s)
- S Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - L Ma
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - R Tan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Q Lu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Y Geng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - G Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Z Gu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Hetzler KL, Collins BC, Shanely RA, Sue H, Kostek MC. The homoeobox gene SIX1 alters myosin heavy chain isoform expression in mouse skeletal muscle. Acta Physiol (Oxf) 2014; 210:415-28. [PMID: 24102895 DOI: 10.1111/apha.12168] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 08/21/2013] [Accepted: 09/10/2013] [Indexed: 01/06/2023]
Abstract
AIM Six1 is necessary for the genesis of several tissues, but in adults, it is expressed primarily in skeletal muscle where its function is unclear. Overexpression of Six1 with a cofactor in skeletal muscle causes slow-to-fast fibre-type transition. We sought to characterize the effects of a physiologically relevant Six1 knockdown. METHODS The tibialis anterior (TA) muscles of C57BL/6 mice were electroporated with Six1 knockdown vector (siRNA) or empty vector. Muscles were collected at 2 or 14 days after transfection for Six1 and myosin heavy chain (MHC) expression analysis. C2C12 mouse myoblasts were grown in standard conditions. Cells were cotransfected with MHC promoter vectors and Six1 expression vectors. Cells were harvested after 4 days of differentiation. RESULTS In vivo, the Six1 siRNA caused a decreased expression of Six1,1.8-fold (±0.1, P < 0.05). With decreased Six1, MHC IIB expression decreased 2.7-fold (±0.7, P = 0.04). Proportion of muscle fibres expressing MHC IIB decreased (45.3 ± 4.8% vs. 65.1 ± 7.3% in control group, P = 0.04), and total area expressing MHC IIB decreased with decreased Six1 (59.6 ± 4.3% vs. 75.2 ± 5.4% in control group, P < 0.05). Decreased Six1 increased MHC IIA expression 1.9-fold (±0.3, P = 0.04). In vitro, Six1 overexpression increased promoter activation of MHC IIB 2.9-fold (±0.3, P < 0.01). Six1 knockdown repressed MHC IIB promoter 2.9-fold (±0.1, P < 0.05) and MHC IIX 3.7-fold (±0.08, P < 0.01). CONCLUSION Six1 knockdown caused a fast-to-slow shift in MHC isoform, and this was confirmed by promoter activity of MHC genes. Six1 may ultimately control the contractile and metabolic properties that define muscle fibre phenotype.
Collapse
Affiliation(s)
- K. L. Hetzler
- Department of Exercise Science; University of South Carolina; Columbia SC USA
| | - B. C. Collins
- Department of Exercise Science; University of South Carolina; Columbia SC USA
| | - R. A. Shanely
- Appalachian State University-North Carolina Research Campus Human Performance Laboratory; Appalachian State University; Kannapolis NC USA
| | - H. Sue
- Department of Exercise Science; University of South Carolina; Columbia SC USA
| | - M. C. Kostek
- Department of Exercise Science; University of South Carolina; Columbia SC USA
- Department of Physical Therapy; Duquesne University; Pittsburgh PA USA
| |
Collapse
|
34
|
Cornwell EW, Mirbod A, Wu CL, Kandarian SC, Jackman RW. C26 cancer-induced muscle wasting is IKKβ-dependent and NF-kappaB-independent. PLoS One 2014; 9:e87776. [PMID: 24489962 PMCID: PMC3906224 DOI: 10.1371/journal.pone.0087776] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 12/30/2013] [Indexed: 12/24/2022] Open
Abstract
Existing data suggest that NF-kappaB signaling is a key regulator of cancer-induced skeletal muscle wasting. However, identification of the components of this signaling pathway and of the NF-κB transcription factors that regulate wasting is far from complete. In muscles of C26 tumor bearing mice, overexpression of dominant negative (d.n.) IKKβ blocked muscle wasting by 69% and the IκBα-super repressor blocked wasting by 41%. In contrast, overexpression of d.n. IKKα or d.n. NIK did not block C26-induced wasting. Surprisingly, overexpression of d.n. p65 or d.n. c-Rel did not significantly affect muscle wasting. Genome-wide mRNA expression arrays showed upregulation of many genes previously implicated in muscle atrophy. To test if these upregulated genes were direct targets of NF-κB transcription factors, we compared genome-wide p65 binding to DNA in control and cachectic muscle using ChIP-sequencing. Bioinformatic analysis of ChIP-sequencing data from control and C26 muscles showed very little p65 binding to genes in cachexia and little to suggest that upregulated p65 binding influences the gene expression associated with muscle based cachexia. The p65 ChIP-seq data are consistent with our finding of no significant change in protein binding to an NF-κB oligonucleotide in a gel shift assay, no activation of a NF-κB-dependent reporter, and no effect of d.n.p65 overexpression in muscles of tumor bearing mice. Taken together, these data support the idea that although inhibition of IκBα, and particularly IKKβ, blocks cancer-induced wasting, the alternative NF-κB signaling pathway is not required. In addition, the downstream NF-κB transcription factors, p65 and c-Rel do not appear to regulate the transcriptional changes induced by the C26 tumor. These data are consistent with the growing body of literature showing that there are NF-κB-independent substrates of IKKβ and IκBα that regulate physiological processes.
Collapse
Affiliation(s)
- Evangeline W. Cornwell
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
| | - Azadeh Mirbod
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
| | - Chia-Ling Wu
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
| | - Susan C. Kandarian
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
| | - Robert W. Jackman
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
35
|
Hitachi K, Tsuchida K. Role of microRNAs in skeletal muscle hypertrophy. Front Physiol 2014; 4:408. [PMID: 24474938 PMCID: PMC3893574 DOI: 10.3389/fphys.2013.00408] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 12/25/2013] [Indexed: 01/05/2023] Open
Abstract
Skeletal muscle comprises approximately 40% of body weight, and is important for locomotion, as well as for metabolic homeostasis. Adult skeletal muscle mass is maintained by a fine balance between muscle protein synthesis and degradation. In response to cytokines, nutrients, and mechanical stimuli, skeletal muscle mass is increased (hypertrophy), whereas skeletal muscle mass is decreased (atrophy) in a variety of conditions, including cancer cachexia, starvation, immobilization, aging, and neuromuscular disorders. Recent studies have determined two important signaling pathways involved in skeletal muscle mass. The insulin-like growth factor-1 (IGF-1)/Akt pathway increases skeletal muscle mass via stimulation of protein synthesis and inhibition of protein degradation. By contrast, myostatin signaling negatively regulates skeletal muscle mass by reducing protein synthesis. In addition, the discovery of microRNAs as novel regulators of gene expression has provided new insights into a multitude of biological processes, especially in skeletal muscle physiology. We summarize here the current knowledge of microRNAs in the regulation of skeletal muscle hypertrophy, focusing on the IGF-1/Akt pathway and myostatin signaling.
Collapse
Affiliation(s)
- Keisuke Hitachi
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University Toyoake, Japan
| | - Kunihiro Tsuchida
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University Toyoake, Japan
| |
Collapse
|
36
|
van Drunen Littel-van den Hurk S, Hannaman D. Electroporation for DNA immunization: clinical application. Expert Rev Vaccines 2014; 9:503-17. [DOI: 10.1586/erv.10.42] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
37
|
Kósa M, Zádor E. Transfection efficiency along the regenerating soleus muscle of the rat. Mol Biotechnol 2013; 54:220-7. [PMID: 22580850 DOI: 10.1007/s12033-012-9555-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
We investigated the efficiency of a single plasmid transfection along the longitudinal axis of the regenerating soleus of young rats. This also reflected transfection efficiency along the fibers because the soleus is a nearly fusiform muscle in young animals. The complete regeneration was induced by notexin and the transfection was made by intramuscular injection of enhanced green fluorescent protein- or Discosoma red-coding plasmids after 4 days. One week after transfection the number of transfected fibers was higher at the place of injection (i.e., in the muscle belly) and lower or absent at the ends of the muscle. The inspection of longitudinal sections and neuromuscular endplates indicated that one of the reasons of uneven transfection might be the shortness of transfected myotubes and the other reason might be the limit of diffusion of transgenic proteins from the expressing nuclei. As a result, the efficiency of transfection in the whole regenerating muscle was much lower than it could be estimated from the most successfully transfected part.
Collapse
Affiliation(s)
- Magdolna Kósa
- Department of Biochemistry, University of Szeged, Szeged, Hungary.
| | | |
Collapse
|
38
|
Luo Y, Liu J, Wang Y, Su J, Wu Y, Hu G, Gao M, Quan F, Zhang Y. PhiC31 integrase-mediated genomic integration and stable gene expression in the mouse mammary gland after gene electrotransfer. J Gene Med 2013; 15:356-65. [PMID: 24288809 DOI: 10.1002/jgm.2723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND PhiC31 integrase is capable of conferring long-term transgene expression in various transfected tissues in vivo. In the present study, we investigated the activity of phiC31 integrase in mouse mammary glands. METHODS The normal mouse mammary epithelial cell line HC11 was transfected with FuGENE® HD Transfection Reagent (Roche Diagnostics, Shanghai, China). Transfection of the mouse mammary gland in vivo was performed by electrotransfer. Transgene expression was detected by western blotting and an enzyme-linked immunosorbent assay. Genomic integration and integration at mpsL1 was confirmed by a nested polymerase chain reaction. RESULTS An optimal electrotransfer protocol for the lactating mouse mammary gland was attained through investigation of different voltages and pulse durations. PhiC31 integrase mediated site-specific transgene integration in HC11 cells and the mouse mammary gland. In addition, the site-specific integration occurred efficiently at the ‘hot spot’ mpsL1. Co-delivery of PhiC31 integrase enhanced and prolonged transgene expression in the mouse mammary gland. CONCLUSIONS The results obtained in the present study show that the use of phiC31 integrase is a feasible and efficient method for high and stable transgene expression in the mouse mammary gland.
Collapse
|
39
|
Oddoux S, Zaal KJ, Tate V, Kenea A, Nandkeolyar SA, Reid E, Liu W, Ralston E. Microtubules that form the stationary lattice of muscle fibers are dynamic and nucleated at Golgi elements. ACTA ACUST UNITED AC 2013; 203:205-13. [PMID: 24145165 PMCID: PMC3812964 DOI: 10.1083/jcb.201304063] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Live imaging reveals that muscle microtubules are highly dynamic and build a durable network nucleated by static Golgi elements. Skeletal muscle microtubules (MTs) form a nonclassic grid-like network, which has so far been documented in static images only. We have now observed and analyzed dynamics of GFP constructs of MT and Golgi markers in single live fibers and in the whole mouse muscle in vivo. Using confocal, intravital, and superresolution microscopy, we find that muscle MTs are dynamic, growing at the typical speed of ∼9 µm/min, and forming small bundles that build a durable network. We also show that static Golgi elements, associated with the MT-organizing center proteins γ-tubulin and pericentrin, are major sites of muscle MT nucleation, in addition to the previously identified sites (i.e., nuclear membranes). These data give us a framework for understanding how muscle MTs organize and how they contribute to the pathology of muscle diseases such as Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Sarah Oddoux
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Konieczny P, Swiderski K, Chamberlain JS. Gene and cell-mediated therapies for muscular dystrophy. Muscle Nerve 2013; 47:649-63. [PMID: 23553671 DOI: 10.1002/mus.23738] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2012] [Indexed: 12/29/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating muscle disorder that affects 1 in 3,500 boys. Despite years of research and considerable progress in understanding the molecular mechanism of the disease and advancement of therapeutic approaches, there is no cure for DMD. The current treatment options are limited to physiotherapy and corticosteroids, and although they provide a substantial improvement in affected children, they only slow the course of the disorder. On a more optimistic note, more recent approaches either significantly alleviate or eliminate muscular dystrophy in murine and canine models of DMD and importantly, many of them are being tested in early phase human clinical trials. This review summarizes advancements that have been made in viral and nonviral gene therapy as well as stem cell therapy for DMD with a focus on the replacement and repair of the affected dystrophin gene.
Collapse
Affiliation(s)
- Patryk Konieczny
- Department of Neurology, The University of Washington School of Medicine, Seattle, Washington 98105, USA
| | | | | |
Collapse
|
41
|
Lally JSV, Herbst EAF, Matravadia S, Maher AC, Perry CGR, Ventura-Clapier R, Holloway GP. Over-expressing mitofusin-2 in healthy mature mammalian skeletal muscle does not alter mitochondrial bioenergetics. PLoS One 2013; 8:e55660. [PMID: 23383258 PMCID: PMC3561349 DOI: 10.1371/journal.pone.0055660] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 12/28/2012] [Indexed: 11/24/2022] Open
Abstract
The role of mitofusin-2 (MFN-2) in regulating mitochondrial dynamics has been well-characterized in lower order eukaryotic cell lines through the complete ablation of MFN-2 protein. However, to support the contractile function of mature skeletal muscle, the subcellular architecture and constituent proteins of this tissue differ substantially from simpler cellular organisms. Such differences may also impact the role of MFN-2 in mature mammalian muscle, and it is unclear if minor fluctuations in MFN-2, as observed in response to physiological perturbations, has a functional consequence. Therefore, we have transiently transfected MFN-2 cDNA into rat tibialis anterior muscle to determine the effect of physiolgically relevant increases in MFN-2 protein on mitochondrial bioenergetics. Permeabilized muscle fibres generated from muscle following MFN-2-transfection were used for functional assessments of mitochondrial bioenergetics. In addition, we have further established a novel method for selecting fibre bundles that are positively transfected, and using this approach transient transfection increased MFN-2 protein ∼2.3 fold in selected muscle fibres. However, this did not alter maximal rates of oxygen consumption or the sensitivity for ADP-stimulated respiration. In addition, MFN-2 over-expression did not alter rates of H2O2 emission. Altogether, and contrary to evidence from lower order cell lines, our results indicate that over-expressing MFN-2 in healthy muscle does not influence mitochondrial bioenergetics in mature mammalian skeletal muscle.
Collapse
Affiliation(s)
- James S V Lally
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
42
|
Andres-Mateos E, Brinkmeier H, Burks TN, Mejias R, Files DC, Steinberger M, Soleimani A, Marx R, Simmers JL, Lin B, Finanger Hedderick E, Marr TG, Lin BM, Hourdé C, Leinwand LA, Kuhl D, Föller M, Vogelsang S, Hernandez-Diaz I, Vaughan DK, Alvarez de la Rosa D, Lang F, Cohn RD. Activation of serum/glucocorticoid-induced kinase 1 (SGK1) is important to maintain skeletal muscle homeostasis and prevent atrophy. EMBO Mol Med 2012; 5:80-91. [PMID: 23161797 PMCID: PMC3569655 DOI: 10.1002/emmm.201201443] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 09/25/2012] [Accepted: 09/28/2012] [Indexed: 12/18/2022] Open
Abstract
Maintaining skeletal muscle mass is essential for general health and prevention of disease progression in various neuromuscular conditions. Currently, no treatments are available to prevent progressive loss of muscle mass in any of these conditions. Hibernating mammals are protected from muscle atrophy despite prolonged periods of immobilization and starvation. Here, we describe a mechanism underlying muscle preservation and translate it to non-hibernating mammals. Although Akt has an established role in skeletal muscle homeostasis, we find that serum- and glucocorticoid-inducible kinase 1 (SGK1) regulates muscle mass maintenance via downregulation of proteolysis and autophagy as well as increased protein synthesis during hibernation. We demonstrate that SGK1 is critical for the maintenance of skeletal muscle homeostasis and function in non-hibernating mammals in normal and atrophic conditions such as starvation and immobilization. Our results identify a novel therapeutic target to combat loss of skeletal muscle mass associated with muscle degeneration and atrophy.
Collapse
Affiliation(s)
- Eva Andres-Mateos
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Baligand C, Jouvion G, Schakman O, Gilson H, Wary C, Thissen JP, Carlier PG. Multiparametric functional nuclear magnetic resonance imaging shows alterations associated with plasmid electrotransfer in mouse skeletal muscle. J Gene Med 2012; 14:598-608. [DOI: 10.1002/jgm.2671] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
| | | | - Olivier Schakman
- Pôle d'Endocrinologie, de Diabétologie et Nutrition, Institut de Recherches expérimentales et cliniques (IREC); Université catholique de Louvain; Brussels; Belgium
| | - Helene Gilson
- Pôle d'Endocrinologie, de Diabétologie et Nutrition, Institut de Recherches expérimentales et cliniques (IREC); Université catholique de Louvain; Brussels; Belgium
| | | | - Jean-Paul Thissen
- Pôle d'Endocrinologie, de Diabétologie et Nutrition, Institut de Recherches expérimentales et cliniques (IREC); Université catholique de Louvain; Brussels; Belgium
| | | |
Collapse
|
44
|
Murphy KT, Ham DJ, Church JE, Naim T, Trieu J, Williams DA, Lynch GS. Parvalbumin gene transfer impairs skeletal muscle contractility in old mice. Hum Gene Ther 2012; 23:824-36. [PMID: 22455364 DOI: 10.1089/hum.2011.210] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Sarcopenia is the progressive age-related loss of skeletal muscle mass associated with functional impairments that reduce mobility and quality of life. Overt muscle wasting with sarcopenia is usually preceded by a slowing of the rate of relaxation and a reduction in maximum force production. Parvalbumin (PV) is a cytosolic Ca(2+) buffer thought to facilitate relaxation in muscle. We tested the hypothesis that restoration of PV levels in muscles of old mice would increase the magnitude and hasten relaxation of submaximal and maximal force responses. The tibialis anterior (TA) muscles of young (6 month), adult (13 month), and old (26 month) C57BL/6 mice received electroporation-assisted gene transfer of plasmid encoding PV or empty plasmid (pcDNA3.1). Contractile properties of TA muscles were assessed in situ 14 days after transfer. In old mice, muscles with increased PV expression had a 40% slower rate of tetanic force development (p<0.01), and maximum twitch and tetanic force were 22% and 16% lower than control values, respectively (p<0.05). Muscles with increased PV expression from old mice had an 18% lower maximum specific (normalized) force than controls, and absolute force was `26% lower at higher stimulation frequencies (150-300 Hz, p<0.05). In contrast, there was no effect of increased PV expression on TA muscle contractile properties in young and adult mice. The impairments in skeletal muscle function in old mice argue against PV overexpression as a therapeutic strategy for ameliorating aspects of contractile dysfunction with sarcopenia and help clarify directions for therapeutic interventions for age-related changes in skeletal muscle structure and function.
Collapse
Affiliation(s)
- Kate T Murphy
- Basic and Clinical Myology Laboratory, The University of Melbourne, Melbourne, VIC 3010, Australia
| | | | | | | | | | | | | |
Collapse
|
45
|
BMP-2 gene transfer under various conditions with in vivo electroporation and bone induction. JOURNAL OF ORAL AND MAXILLOFACIAL SURGERY MEDICINE AND PATHOLOGY 2012. [DOI: 10.1016/j.ajoms.2011.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
46
|
The E3 ubiquitin ligase TRAF6 intercedes in starvation-induced skeletal muscle atrophy through multiple mechanisms. Mol Cell Biol 2012; 32:1248-59. [PMID: 22290431 DOI: 10.1128/mcb.06351-11] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Starvation, like many other catabolic conditions, induces loss of skeletal muscle mass by promoting fiber atrophy. In addition to the canonical processes, the starvation-induced response employs many distinct pathways that make it a unique atrophic program. However, in the multiplex of the underlying mechanisms, several components of starvation-induced atrophy have yet to be fully understood and their roles and interplay remain to be elucidated. Here we unveiled the role of tumor necrosis factor receptor-associated factor 6 (TRAF6), a unique E3 ubiquitin ligase and adaptor protein, in starvation-induced muscle atrophy. Targeted ablation of TRAF6 suppresses the expression of key regulators of atrophy, including MAFBx, MuRF1, p62, LC3B, Beclin1, Atg12, and Fn14. Ablation of TRAF6 also improved the phosphorylation of Akt and FoxO3a and inhibited the activation of 5' AMP-activated protein kinase in skeletal muscle in response to starvation. In addition, our study provides the first evidence of the involvement of endoplasmic reticulum stress and unfolding protein response pathways in starvation-induced muscle atrophy and its regulation through TRAF6. Finally, our results also identify lysine 63-linked autoubiquitination of TRAF6 as a process essential for its regulatory role in starvation-induced muscle atrophy.
Collapse
|
47
|
Wu CL, Kandarian SC. Protein overexpression in skeletal muscle using plasmid-based gene transfer to elucidate mechanisms controlling fiber size. Methods Mol Biol 2012; 798:231-243. [PMID: 22130840 DOI: 10.1007/978-1-61779-343-1_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Plasmid DNA electrotransfer is a direct method of gene delivery to skeletal muscle commonly used to identify endogenous signaling pathways that mediate muscle remodeling or pathological states in adult rodents. When plasmids encoding a protein to be overexpressed are fused to a fluorescent protein or an epitope-tag, plasmid electrotransfer permits visualization of the expressed protein in muscle fibers. Here, we demonstrate the use of electrotransfer of plasmids encoding mutant or wild type proteins to identify the role of the endogenous protein in regulating muscle fiber atrophy. The plasmids used encode a dominant negative form of the inhibitor of kappaB kinase beta (IKKβ) fused to green fluorescent protein (GFP), a constitutively active form of IKKα fused to GFP, and a wild type IKKβ fused to an HA tag. We show the effects of overexpression of these proteins on rat or mouse fiber size either with disuse atrophy or in normal weight bearing muscle. The effects of overexpressed proteins on myofiber size are assessed by comparing cross-sectional area of the transfected, fluorescent myofibers to the nontransfected, nonfluorescent myofibers. Using optimized intramuscular plasmid DNA injection and electroporation, we illustrate high transfection efficiency with no overt muscle damage using medium sized fusion proteins (105 kDa).
Collapse
Affiliation(s)
- Chia-Ling Wu
- Department of Health Sciences, Boston University, Boston, MA, USA
| | | |
Collapse
|
48
|
Reed SA, Sandesara PB, Senf SM, Judge AR. Inhibition of FoxO transcriptional activity prevents muscle fiber atrophy during cachexia and induces hypertrophy. FASEB J 2011; 26:987-1000. [PMID: 22102632 DOI: 10.1096/fj.11-189977] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cachexia is characterized by inexorable muscle wasting that significantly affects patient prognosis and increases mortality. Therefore, understanding the molecular basis of this muscle wasting is of significant importance. Recent work showed that components of the forkhead box O (FoxO) pathway are increased in skeletal muscle during cachexia. In the current study, we tested the physiological significance of FoxO activation in the progression of muscle atrophy associated with cachexia. FoxO-DNA binding dependent transcription was blocked in the muscles of mice through injection of a dominant negative (DN) FoxO expression plasmid prior to inoculation with Lewis lung carcinoma cells or the induction of sepsis. Expression of DN FoxO inhibited the increased mRNA levels of atrogin-1, MuRF1, cathepsin L, and/or Bnip3 and inhibited muscle fiber atrophy during cancer cachexia and sepsis. Interestingly, during control conditions, expression of DN FoxO decreased myostatin expression, increased MyoD expression and satellite cell proliferation, and induced fiber hypertrophy, which required de novo protein synthesis. Collectively, these data show that FoxO-DNA binding-dependent transcription is necessary for normal muscle fiber atrophy during cancer cachexia and sepsis, and further suggest that basal levels of FoxO play an important role during normal conditions to depress satellite cell activation and limit muscle growth.
Collapse
Affiliation(s)
- Sarah A Reed
- Department of Physical Therapy, 101 S. Newell Dr., University of Florida, Gainesville, FL 32611, USA
| | | | | | | |
Collapse
|
49
|
Zhu H, Bhattacharyya BJ, Lin H, Gomez CM. Skeletal muscle IP3R1 receptors amplify physiological and pathological synaptic calcium signals. J Neurosci 2011; 31:15269-83. [PMID: 22031873 PMCID: PMC3237715 DOI: 10.1523/jneurosci.3766-11.2011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 08/24/2011] [Accepted: 09/01/2011] [Indexed: 01/11/2023] Open
Abstract
Ca(2+) release from internal stores is critical for mediating both normal and pathological intracellular Ca(2+) signaling. Recent studies suggest that the inositol 1,4,5-triphosphate (IP(3)) receptor mediates Ca(2+) release from internal stores upon cholinergic activation of the neuromuscular junction (NMJ) in both physiological and pathological conditions. Here, we report that the type I IP(3) receptor (IP(3)R(1))-mediated Ca(2+) release plays a crucial role in synaptic gene expression, development, and neuromuscular transmission, as well as mediating degeneration during excessive cholinergic activation. We found that IP(3)R(1)-mediated Ca(2+) release plays a key role in early development of the NMJ, homeostatic regulation of neuromuscular transmission, and synaptic gene expression. Reducing IP(3)R(1)-mediated Ca(2+) release via siRNA knockdown or IP(3)R blockers in C2C12 cells decreased calpain activity and prevented agonist-induced acetylcholine receptor (AChR) cluster dispersal. In fully developed NMJ in adult muscle, IP(3)R(1) knockdown or blockade effectively increased synaptic strength at presynaptic and postsynaptic sites by increasing both quantal release and expression of AChR subunits and other NMJ-specific genes in a pattern resembling muscle denervation. Moreover, in two mouse models of cholinergic overactivity and NMJ Ca(2+) overload, anti-cholinesterase toxicity and the slow-channel myasthenic syndrome (SCS), IP(3)R(1) knockdown eliminated NMJ Ca(2+) overload, pathological activation of calpain and caspase proteases, and markers of DNA damage at subsynaptic nuclei, and improved both neuromuscular transmission and clinical measures of motor function. Thus, blockade or genetic silencing of muscle IP(3)R(1) may be an effective and well tolerated therapeutic strategy in SCS and other conditions of excitotoxicity or Ca(2+) overload.
Collapse
MESH Headings
- Action Potentials/drug effects
- Action Potentials/genetics
- Animals
- Boron Compounds/pharmacology
- Calcium/metabolism
- Calcium Signaling/genetics
- Calcium Signaling/physiology
- Calpain/metabolism
- Carbachol/pharmacology
- Caspase 3/metabolism
- Caspase 9/metabolism
- Cell Line, Transformed
- Cholinergic Agonists/pharmacology
- Cholinesterase Inhibitors/toxicity
- Disease Models, Animal
- Electromyography
- Electroporation/methods
- Exercise Test
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/genetics
- Green Fluorescent Proteins/genetics
- Histone Deacetylases/metabolism
- Histones/genetics
- Histones/metabolism
- In Vitro Techniques
- Inositol 1,4,5-Trisphosphate Receptors/deficiency
- Inositol 1,4,5-Trisphosphate Receptors/physiology
- Male
- Membrane Potentials/drug effects
- Membrane Potentials/genetics
- Mice
- Mice, Transgenic
- Muscle, Skeletal/metabolism
- Myasthenic Syndromes, Congenital/genetics
- Myasthenic Syndromes, Congenital/pathology
- Myasthenic Syndromes, Congenital/therapy
- Neostigmine/toxicity
- Nerve Tissue Proteins/metabolism
- Neuromuscular Junction/metabolism
- Neuromuscular Junction/physiology
- Neurotoxicity Syndromes/etiology
- Neurotoxicity Syndromes/pathology
- Neurotoxicity Syndromes/therapy
- Patch-Clamp Techniques
- RNA, Small Interfering/pharmacology
- Receptors, Cholinergic/classification
- Receptors, Cholinergic/genetics
- Receptors, Cholinergic/metabolism
- Sciatic Nerve/physiopathology
- Time Factors
Collapse
Affiliation(s)
- Haipeng Zhu
- Department of Neurology, University of Chicago Medical Center, Chicago, Illinois 60637
| | - Bula J. Bhattacharyya
- Department of Molecular Pharmacology and Biological Chemistry, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, and
| | - Hong Lin
- Departments of Neurology and Pediatrics, the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104-4318
| | - Christopher M. Gomez
- Department of Neurology, University of Chicago Medical Center, Chicago, Illinois 60637
| |
Collapse
|
50
|
Abstract
For plasmid-mediated gene therapy applications, a major limitation to scale up from rodents to large animals is the low expression level of injected plasmid DNA. The electroporation technique, which results in the passage of foreign material through the cell membrane, is one method that has been shown to be effective at improving local plasmid uptake and consequently, expression levels. Previous studies have determined that optimized electroporation parameters (such as electric field intensity, number of pulses, lag time between plasmid injections and electroporations, and optimal plasmid formulation conditions) are dependent on the target muscle type and individual species. Here, we provide a detailed protocol to optimize conditions for the successful intramuscular electroporation of plasmid DNA to swine, a large animal model. Our results suggest that the technique is safe and effective for veterinary applications. Furthermore, these results provide evidence for the feasibility of upcoming human applications.
Collapse
|