1
|
Thakre PP, Rana S, Benevides ES, Fuller DD. Targeting drug or gene delivery to the phrenic motoneuron pool. J Neurophysiol 2023; 129:144-158. [PMID: 36416447 PMCID: PMC9829468 DOI: 10.1152/jn.00432.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/19/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
Phrenic motoneurons (PhrMNs) innervate diaphragm myofibers. Located in the ventral gray matter (lamina IX), PhrMNs form a column extending from approximately the third to sixth cervical spinal segment. Phrenic motor output and diaphragm activation are impaired in many neuromuscular diseases, and targeted delivery of drugs and/or genetic material to PhrMNs may have therapeutic application. Studies of phrenic motor control and/or neuroplasticity mechanisms also typically require targeting of PhrMNs with drugs, viral vectors, or tracers. The location of the phrenic motoneuron pool, however, poses a challenge. Selective PhrMN targeting is possible with molecules that move retrogradely upon uptake into phrenic axons subsequent to diaphragm or phrenic nerve delivery. However, nonspecific approaches that use intrathecal or intravenous delivery have considerably advanced the understanding of PhrMN control. New opportunities for targeted PhrMN gene expression may be possible with intersectional genetic methods. This article provides an overview of methods for targeting the phrenic motoneuron pool for studies of PhrMNs in health and disease.
Collapse
Affiliation(s)
- Prajwal P Thakre
- Department of Physical Therapy, University of Florida, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
- Breathing Research and Therapeutics Center, Gainesville, Florida
| | - Sabhya Rana
- Department of Physical Therapy, University of Florida, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
- Breathing Research and Therapeutics Center, Gainesville, Florida
| | - Ethan S Benevides
- Department of Physical Therapy, University of Florida, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
- Breathing Research and Therapeutics Center, Gainesville, Florida
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
- Breathing Research and Therapeutics Center, Gainesville, Florida
| |
Collapse
|
2
|
Benevides ES, Sunshine MD, Rana S, Fuller DD. Optogenetic activation of the diaphragm. Sci Rep 2022; 12:6503. [PMID: 35444167 PMCID: PMC9021282 DOI: 10.1038/s41598-022-10240-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 04/04/2022] [Indexed: 11/10/2022] Open
Abstract
Impaired diaphragm activation is common in many neuromuscular diseases. We hypothesized that expressing photoreceptors in diaphragm myofibers would enable light stimulation to evoke functional diaphragm activity, similar to endogenous bursts. In a mouse model, adeno-associated virus (AAV) encoding channelrhodopsin-2 (AAV9-CAG-ChR2-mVenus, 6.12 × 1011 vg dose) was delivered to the diaphragm using a minimally invasive method of microinjection to the intrapleural space. At 8-18 weeks following AAV injection, mice were anesthetized and studied during spontaneous breathing. We first showed that diaphragm electromyographic (EMG) potentials could be evoked with brief presentations of light, using a 473 nm high intensity LED. Evoked potential amplitude increased with intensity or duration of the light pulse. We next showed that in a paralyzed diaphragm, trains of light pulses evoked diaphragm EMG activity which resembled endogenous bursting, and this was sufficient to generate respiratory airflow. Light-evoked diaphragm EMG bursts showed no diminution after up to one hour of stimulation. Histological evaluation confirmed transgene expression in diaphragm myofibers. We conclude that intrapleural delivery of AAV9 can drive expression of ChR2 in the diaphragm and subsequent photostimulation can evoke graded compound diaphragm EMG activity similar to endogenous inspiratory bursting.
Collapse
Affiliation(s)
- Ethan S Benevides
- Rehabilitation Science PhD Program, University of Florida, Gainesville, Florida, USA.,Department of Physical Therapy, University of Florida, Gainesville, Florida, USA.,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA.,McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Michael D Sunshine
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA.,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA.,McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Sabhya Rana
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA.,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA.,McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA. .,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA. .,McKnight Brain Institute, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
3
|
Herrick SE, Mutsaers SE. The Potential of Mesothelial Cells in Tissue Engineering and Regenerative Medicine Applications. Int J Artif Organs 2018; 30:527-40. [PMID: 17628854 DOI: 10.1177/039139880703000611] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Injury to the serosa through injurious agents such as radiation, surgery, infection and disease results in the loss of the protective surface mesothelium and often leads to fibrous adhesion formation. Mechanisms that increase the rate of mesothialisation are therefore actively being investigated in order to reduce the formation of adhesions. These include intraperitoneal delivery of cultured mesothelial cells as well as administration of factors that are known to increase mesothelial proliferation and migration. An exciting alternative that has only recently received attention, is the possible role of mesothelial progenitor cells in the repair and regeneration of denuded serosal areas. Accumulating evidence suggests that such a population exists and under certain conditions is able to form a number of defined cell types indicating a degree of plasticity. Such properties may explain the extensive use of mesothelial cells in various tissue engineering applications including the development of vascular conduits and peripheral nerve replacements. It is likely that with the rapid explosion in the fields of tissue engineering and regenerative medicine, a greater understanding of the potential of mesothelial progenitor cells to repair, replace and possibly regenerate damaged or defective tissue will be uncovered.
Collapse
Affiliation(s)
- S E Herrick
- School of Medicine, Faculty of Medical and Human Sciences, University of Manchester, Oxford Road, Manchester, UK.
| | | |
Collapse
|
4
|
Abstract
Pleural malignancies remain a serious therapeutic challenge, and are frequently refractory to standard treatment; however, they have the advantage of occurring in an enclosed cavity readily accessible for examination, biopsy, and serial sampling. Novel therapeutics can be administered via intracavitary delivery to maximize efficacy by targeting the site of involvement and potentially mitigating the adverse effects of systemic therapies. The easy accessibility of the pleural space lends itself well to repeated sampling and analysis to determine efficacy and toxicity of a given treatment paradigm. These factors support the rationale for delivery of novel therapeutics directly into the pleural space.
Collapse
Affiliation(s)
- Vivek Murthy
- NYU PORT (Pulmonary Oncology Research Team), Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, NYU Langone Health, 550 First Avenue Suite 5D, New York, NY 10016, USA
| | - Keshav Mangalick
- NYU PORT (Pulmonary Oncology Research Team), Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, NYU Langone Health, 550 First Avenue Suite 5D, New York, NY 10016, USA
| | - Daniel H Sterman
- NYU PORT (Pulmonary Oncology Research Team), Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine, NYU Langone Health, 550 First Avenue Suite 5D, New York, NY 10016, USA.
| |
Collapse
|
5
|
Chiuchiolo MJ, Crystal RG. Gene Therapy for Alpha-1 Antitrypsin Deficiency Lung Disease. Ann Am Thorac Soc 2016; 13 Suppl 4:S352-69. [PMID: 27564673 PMCID: PMC5059492 DOI: 10.1513/annalsats.201506-344kv] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/28/2015] [Indexed: 12/16/2022] Open
Abstract
Alpha-1 antitrypsin (AAT) deficiency, characterized by low plasma levels of the serine protease inhibitor AAT, is associated with emphysema secondary to insufficient protection of the lung from neutrophil proteases. Although AAT augmentation therapy with purified AAT protein is efficacious, it requires weekly to monthly intravenous infusion of AAT purified from pooled human plasma, has the risk of viral contamination and allergic reactions, and is costly. As an alternative, gene therapy offers the advantage of single administration, eliminating the burden of protein infusion, and reduced risks and costs. The focus of this review is to describe the various strategies for AAT gene therapy for the pulmonary manifestations of AAT deficiency and the state of the art in bringing AAT gene therapy to the bedside.
Collapse
Affiliation(s)
- Maria J Chiuchiolo
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
6
|
Adenoviral vector encoding soluble Flt-1 engineered human endometrial mesenchymal stem cells effectively regress endometriotic lesions in NOD/SCID mice. Gene Ther 2016; 23:580-91. [DOI: 10.1038/gt.2016.30] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 02/26/2016] [Accepted: 03/01/2016] [Indexed: 12/24/2022]
|
7
|
Liu B, Lü X, Qi C, Zheng S, Zhou M, Wang J, Yin W. KGFR promotes Na+ channel expression in a rat acute lung injury model. Afr Health Sci 2014; 14:648-56. [PMID: 25352884 DOI: 10.4314/ahs.v14i3.21] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Binding of keratinocyte growth factor (KGF) to the KGF receptor (KGFR) plays an important role in the recovery of alveolar epithelial cells from acute lung injury (ALI). OBJECTIVES To evaluate the effect of gene therapy via adenovirus gene transfer of KGFR on the treatment of ALI. METHODS Sprague-Dawley rats were divided into four groups: normal controls, injury controls, normal adenovirus transduced group and injury adenovirus transduced group. The ALI model was induced by lipopolysaccharide (LPS) injection. Recombinant adenovirus (AdEasy-KGFR) was injected via the tail vein. Expression of the sodium (Na(+)) channel in rat alveolar type II (ATII) epithelial cells was determined by PCR, immunohistochemistry and immunoelectron microscopy of rat lung tissues. RESULTS Gene expression of the Na(+) channel and KGFR in ATII cells was higher in the normal adenovirus transduced group than the three other groups; expression of these two genes in the injury adenovirus transduced group was higher than the injury control group. Na(+) channel protein expression was lower in the injury adenovirus transduced group but higher than the injury control group. CONCLUSIONS KGFR over-expression induced Na channel expression could potentially be beneficial for ALI therapy.
Collapse
Affiliation(s)
- Binjian Liu
- Laboratory department, 161st Central Hospital of P.L.A., Wuhan 430010, Hubei Province, China
| | - Xin Lü
- Department of Microbiology, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Chaoling Qi
- Laboratory department, 161st Central Hospital of P.L.A., Wuhan 430010, Hubei Province, China
| | - Shuhui Zheng
- Laboratory department, 161st Central Hospital of P.L.A., Wuhan 430010, Hubei Province, China
| | - Muxiu Zhou
- Laboratory department, 161st Central Hospital of P.L.A., Wuhan 430010, Hubei Province, China
| | - Jianmin Wang
- Institute of Surgery Research, the Third Military Medical University, Chongqing 400042, China
| | - Wen Yin
- Department of Microbiology, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China ; Central Laboratory, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| |
Collapse
|
8
|
Phase I/II Study of Intrapleural Administration of a Serotype rh.10 Replication-Deficient Adeno-Associated Virus Gene Transfer Vector Expressing the Human α1-Antitrypsin cDNA to Individuals with α1-Antitrypsin Deficiency. HUM GENE THER CL DEV 2014; 25:112-33. [DOI: 10.1089/humc.2014.2513] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
9
|
Samaranayake H, Määttä AM, Pikkarainen J, Ylä-Herttuala S. Future prospects and challenges of antiangiogenic cancer gene therapy. Hum Gene Ther 2010; 21:381-96. [PMID: 20163246 DOI: 10.1089/hum.2010.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In 1971 Judah Folkman proposed the concept of antiangiogenesis as a therapeutic target for cancer. More than 30 years later, concept became reality with the approval of the antivascular endothelial growth factor (VEGF) monoclonal antibody bevacizumab as a first-line treatment for metastatic colorectal cancer. Monoclonal antibodies and small molecular drugs are the most widely applied methods for inhibition of angiogenesis. The efficacy of these antiangiogenic modalities has been proven, in both preclinical and clinical settings. Although angiogenesis plays a major role in wound healing, hypoxia, and in the female reproductive cycle, inhibition of angiogenesis seems to be a relatively safe therapeutic option against cancers, and has therefore become a logical arena for a wide range of experimentation. The twentieth century has shown the boom of gene therapy and thus it has been applied also in the antiangiogenic setting. This review summarizes methods to induce antiangiogenic responses with gene therapy and discusses the obstacles and future prospects of antiangiogenic cancer gene therapy.
Collapse
|
10
|
Carpenter M, Epperly MW, Agarwal A, Nie S, Hricisak L, Niu Y, Greenberger JS. Inhalation delivery of manganese superoxide dismutase-plasmid/liposomes protects the murine lung from irradiation damage. Gene Ther 2005; 12:685-93. [PMID: 15750616 DOI: 10.1038/sj.gt.3302468] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Intratracheal injection of manganese superoxide dismutase-plasmid/liposome (MnSOD-PL) complexes has been demonstrated to delay the onset and reduce the extent of ionizing irradiation-induced murine pulmonary organizing alveolitis/fibrosis. To facilitate translation of this modality to clinical fractionated radiotherapy, inhalation delivery of MnSOD-PL was developed using an ultrasonic nebulizer. Transgene product was quantitated by immunohistochemical quantitation and pulmonary tissue levels of MnSOD biochemical activity. C57BL/6NHsd female mice demonstrated a plasmid dose-dependent increased expression of MnSOD transgene product over the range of 250 microg-2.5 mg of MnSOD-PL administered over a constant 5 min interval. Delivery of a constant concentration of 500 microg of MnSOD-PL with varying times of administration ranging from 0.5 to 10 min demonstrated optimal MnSOD expression at 5 min. Mice pretreated by inhalation delivery of MnSOD-PL demonstrated significantly improved survival after 20 Gy single fraction irradiation to both lungs compared to LacZ-PL inhalation-treated or irradiated control mice. Mice receiving 10 fractions of 3.5 cGy demonstrated increased pulmonary MnSOD transgene product activity by a protocol of every Monday-Wednesday or daily inhalation of MnSOD-PL. Thus, inhalation radioprotective gene therapy using MnSOD-PL provides a practical and effective method for delivery of lung-specific radioprotection during fractionated radiotherapy protocols in a mouse model.
Collapse
Affiliation(s)
- M Carpenter
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
De B, Heguy A, Leopold PL, Wasif N, Korst RJ, Hackett NR, Crystal RG. Intrapleural administration of a serotype 5 adeno-associated virus coding for α1-antitrypsin mediates persistent, high lung and serum levels of α1-antitrypsin. Mol Ther 2004; 10:1003-10. [PMID: 15564132 DOI: 10.1016/j.ymthe.2004.08.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2004] [Accepted: 08/30/2004] [Indexed: 10/26/2022] Open
Abstract
alpha1-Antitrypsin (alpha1AT) is a serine proteinase inhibitor that protects the lung from degradation by neutrophil proteases. In alpha1AT deficiency, an autosomal recessive disorder resulting from mutations in the alpha1AT (approved symbol SERPINA1) gene, serum alpha1AT levels of < 570 microg/ml are associated with development of emphysema. Adeno-associated virus (AAV) serotype 2 (AAV2) vectors expressing alpha1AT administered intramuscularly or intravenously mediate sustained serum levels of alpha1AT in experimental animals. Since the lung is only 2% of the body weight, AAV vector delivery to the muscle or liver is inefficient, as most of the alpha1AT does not reach the lung. The present study evaluates AAV2- and AAV5-mediated delivery of human alpha1AT (halpha1AT) to C57BL/6 mice using the intrapleural space as a platform for local production of alpha1AT. Intrapleural administration of either an AAV5-halpha1AT or an AAV2-halpha1AT vector achieves higher lung and serum levels of alpha1AT than intramuscular delivery. AAV5-mediated serum and lung alpha1AT levels were 10-fold higher than those achieved by AAV2 delivery via either route. The diaphragm, lung, and heart are the major sites of transgene expression following intrapleural administration of an AAV5 reporter vector. At 40 weeks postadministration, intrapleural administration of the AAV5-halpha1AT vector mediated serum alpha1AT levels of 900 +/- 50 microg/ml, 1.6-fold higher than the accepted therapeutic level of 570 microg/ml. In the context that the pleura is a safe site for administration, intrapleural administration using AAV5 vectors may represent an attractive gene therapy strategy for alpha1AT deficiency in humans.
Collapse
Affiliation(s)
- Bishnu De
- Belfer Gene Therapy Core Facility, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Mahtabifard A, Merritt RE, Yamada RE, Crystal RG, Korst RJ. In vivo gene transfer of pigment epithelium-derived factor inhibits tumor growth in syngeneic murine models of thoracic malignancies. J Thorac Cardiovasc Surg 2003; 126:28-38. [PMID: 12878936 DOI: 10.1016/s0022-5223(02)73616-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Pigment epithelium-derived factor is known to be an inhibitor of angiogenesis. We hypothesized that in vivo gene transfer of pigment epithelium-derived factor may inhibit tumor angiogenesis and growth in syngeneic models of thoracic malignancies. METHODS An adenovirus vector encoding the human pigment epithelium-derived factor cDNA (AdPEDF) was used to transduce human lung cancer cells in vitro. Transgene expression was assessed using Western analysis. Three different murine flank tumors (2 lung, 1 colon) were then established in syngeneic mice and treated intratumorally with phosphate-buffered saline, AdPEDF, or an empty vector (AdNull). Endpoints measured included transgene expression, tumor size, and animal survival, as well as microvessel density within the tumor. Additionally, a murine pulmonary metastasis model was established by intravenous injection of a syngeneic colon adenocarcinoma cell line expressing a marker gene (beta-galactosidase). One day later, treatment (phosphate-buffered saline, AdNull, or AdPEDF) was administered intrapleurally. Tumor burden (gross and histologic inspection, lung weight, and beta-galactosidase expression) was then evaluated 13 days after vector dosing, and survival was recorded. RESULTS AdPEDF-derived expression of pigment epithelium-derived factor was demonstrated in vitro and in vivo. In syngeneic murine lung cancer flank tumors, intratumoral administration of AdPEDF significantly inhibited tumor growth (P <.01), prolonged mouse survival (P <.01), and decreased microvessel density (P <.01) compared with control groups. In the pulmonary metastasis model, AdPEDF-treated mice exhibited significantly reduced lung lesions, lung weight (P <.0005), beta-galactosidase expression (P <.05), and animal survival was prolonged (P <.05). CONCLUSION Gene transfer of pigment epithelium-derived factor suppresses tumor vascularization and growth, while prolonging survival in syngeneic murine models of thoracic malignancies.
Collapse
MESH Headings
- Adenoviruses, Human/genetics
- Animals
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/mortality
- Disease Models, Animal
- Eye Proteins
- Female
- Gene Expression/genetics
- Gene Transfer Techniques
- Genetic Vectors/administration & dosage
- Genetic Vectors/antagonists & inhibitors
- Genetic Vectors/genetics
- Injections, Intravenous
- Lung Neoplasms/genetics
- Lung Neoplasms/mortality
- Lung Neoplasms/secondary
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Models, Cardiovascular
- Nerve Growth Factors
- Protein Biosynthesis
- Proteins/antagonists & inhibitors
- Proteins/genetics
- Serpins/biosynthesis
- Serpins/genetics
- Survival Analysis
- Thoracic Neoplasms/genetics
- Thoracic Neoplasms/metabolism
- Thoracic Neoplasms/mortality
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Ali Mahtabifard
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | | | | | |
Collapse
|