1
|
Vitry S, Mendia C, Maudoux A, El-Amraoui A. Advancing precision ear medicine: leveraging animal models for disease insights and therapeutic innovations. Mamm Genome 2025:10.1007/s00335-025-10126-y. [PMID: 40263131 DOI: 10.1007/s00335-025-10126-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/28/2025] [Indexed: 04/24/2025]
Abstract
Gene therapy offers significant promise for treating inner ear disorders, but its clinical translation requires robust preclinical validation, often reliant on animal models. This review examines the role of these models in advancing gene therapeutics for inherited inner ear disorders, focusing on successes, challenges, and treatment solutions. By providing a precise understanding of disease mechanisms, these models offer a versatile preclinical platform that is essential for assessing and validating therapies. Successful gene supplementation and editing have shown potential in restoring hearing and balance functions and preventing their decline. However, challenges such as limitations in gene delivery methods, surgical access, immune responses, and discrepancies in disease manifestation between animal models and humans hinder clinical translation. Current efforts are dedicated to developing innovative strategies aimed at enhancing the efficiency of gene delivery, overcoming physical barriers such as the blood-labyrinth barrier, improving target specificity, and maximizing therapeutic efficacy while minimizing adverse immune responses. Diverse gene supplementation and editing strategies, along with evolving technologies, hold promise for maximizing therapeutic outcomes using disease relevant models. The future of inner ear gene therapeutics will hinge on personalized therapies and team science fueling interdisciplinary collaborations among researchers, clinicians, companies, and regulatory agencies to expedite the translation from bench to bedside and unlock the immense potential of precision medicine in the inner ear.
Collapse
Affiliation(s)
- Sandrine Vitry
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Progressive Sensory Disorders, Pathophysiology and Therapy, F-75012, Paris, France.
- Unit Progressive Sensory Disorders, Pathophysiology and Therapy, Institut Pasteur, Institut de l'Audition, Paris, France.
| | - Clara Mendia
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Progressive Sensory Disorders, Pathophysiology and Therapy, F-75012, Paris, France
- Collège Doctoral, Sorbonne Université, 75005, Paris, France
| | - Audrey Maudoux
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Progressive Sensory Disorders, Pathophysiology and Therapy, F-75012, Paris, France
- Otolaryngology Department, Assistance Publique des Hôpitaux de Paris, Robert Debré University Hospital-APHP, Paris, France
| | - Aziz El-Amraoui
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Progressive Sensory Disorders, Pathophysiology and Therapy, F-75012, Paris, France.
- Unit Progressive Sensory Disorders, Pathophysiology and Therapy, Institut Pasteur, Institut de l'Audition, Paris, France.
| |
Collapse
|
2
|
Uribe PM, Hudson AM, Lockard G, Jiang M, Harding J, Steyger PS, Coffin AB. Hepatocyte growth factor mimetic confers protection from aminoglycoside-induced hair cell death in vitro. Hear Res 2023; 434:108786. [PMID: 37192594 DOI: 10.1016/j.heares.2023.108786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 04/18/2023] [Accepted: 05/05/2023] [Indexed: 05/18/2023]
Abstract
Loss of sensory hair cells from exposure to certain licit drugs, such as aminoglycoside antibiotics, can result in permanent hearing damage. Exogenous application of the neurotrophic molecule hepatocyte growth factor (HGF) promotes neuronal cell survival in a variety of contexts, including protecting hair cells from aminoglycoside ototoxicity. HGF itself is not an ideal therapeutic due to a short half-life and limited blood-brain barrier permeability. MM-201 is a chemically stable, blood-brain barrier permeable, synthetic HGF mimetic that serves as a functional ligand to activate the HGF receptor and its downstream signaling cascade. We previously demonstrated that MM-201 robustly protects zebrafish lateral line hair cells from aminoglycoside ototoxicity. Here, we examined the ability of MM-201 to protect mammalian sensory hair cells from aminoglycoside damage to further evaluate MM-201's clinical potential. We found that MM-201 exhibited dose-dependent protection from neomycin and gentamicin ototoxicity in mature mouse utricular explants. MM-201's protection was reduced following inhibition of mTOR, a downstream target of HGF receptor activation, implicating the activation of endogenous intracellular substrates by MM-201 as critical for the observed protection. We then asked if MM-201 altered the bactericidal properties of aminoglycosides. Using either plate or liquid growth assays we found that MM-201 did not alter the bactericidal efficacy of aminoglycoside antibiotics at therapeutically relevant concentrations. We therefore assessed the protective capacity of MM-201 in an in vivo mouse model of kanamycin ototoxicity. In contrast to our in vitro data, MM-201 did not attenuate kanamycin ototoxicity in vivo. Further, we found that MM-201 was ototoxic to mice across the dose range tested here. These data suggest species- and tissue-specific differences in otoprotective capacity. Next generation HGF mimetics are in clinical trials for neurodegenerative diseases and show excellent safety profiles, but neither preclinical studies nor clinical trials have examined hearing loss as a potential consequence of pharmaceutical HGF activation. Further research is needed to determine the consequences of systemic MM-201 application on the auditory system.
Collapse
Affiliation(s)
- Phillip M Uribe
- Department of Integrative Physiology and Neuroscience, Washington State University, 14204 NE Salmon Creek Ave, Vancouver, WA 98686 USA
| | - Alexandria M Hudson
- Department of Integrative Physiology and Neuroscience, Washington State University, 14204 NE Salmon Creek Ave, Vancouver, WA 98686 USA
| | - Gavin Lockard
- Department of Integrative Physiology and Neuroscience, Washington State University, 14204 NE Salmon Creek Ave, Vancouver, WA 98686 USA
| | - Meiyan Jiang
- Oregon Hearing Research Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Joseph Harding
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA
| | - Peter S Steyger
- Translational Hearing Center, Creighton University, Omaha, NE, 68178, USA
| | - Allison B Coffin
- Department of Integrative Physiology and Neuroscience, Washington State University, 14204 NE Salmon Creek Ave, Vancouver, WA 98686 USA.
| |
Collapse
|
3
|
Employing siRNA tool and its delivery platforms in suppressing cisplatin resistance: Approaching to a new era of cancer chemotherapy. Life Sci 2021; 277:119430. [PMID: 33789144 DOI: 10.1016/j.lfs.2021.119430] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/10/2021] [Accepted: 03/23/2021] [Indexed: 12/18/2022]
Abstract
Although chemotherapy is a first option in treatment of cancer patients, drug resistance has led to its failure, requiring strategies to overcome it. Cancer cells are capable of switching among molecular pathways to ensure their proliferation and metastasis, leading to their resistance to chemotherapy. The molecular pathways and mechanisms that are responsible for cancer progression and growth, can be negatively affected for providing chemosensitivity. Small interfering RNA (siRNA) is a powerful tool extensively applied in cancer therapy in both pre-clinical (in vitro and in vivo) and clinical studies because of its potential in suppressing tumor-promoting factors. As such oncogene pathways account for cisplatin (CP) resistance, their targeting by siRNA plays an important role in reversing chemoresistance. In the present review, application of siRNA for suppressing CP resistance is discussed. The first priority of using siRNA is sensitizing cancer cells to CP-mediated apoptosis via down-regulating survivin, ATG7, Bcl-2, Bcl-xl, and XIAP. The cancer stem cell properties and related molecular pathways including ID1, Oct-4 and nanog are inhibited by siRNA in CP sensitivity. Cell cycle arrest and enhanced accumulation of CP in cancer cells can be obtained using siRNA. In overcoming siRNA challenges such as off-targeting feature and degradation, carriers including nanoparticles and biological carriers have been applied. These carriers are important in enhancing cellular accumulation of siRNA, elevating gene silencing efficacy and reversing CP resistance.
Collapse
|
4
|
Takehana S, Murata Y, Jo JI, Tabata Y. Complexation design of cationized gelatin and molecular beacon to visualize intracellular mRNA. PLoS One 2021; 16:e0245899. [PMID: 33493232 PMCID: PMC7833158 DOI: 10.1371/journal.pone.0245899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
The objective of this study is to prepare cationized gelatin-molecular beacon (MB) complexes for the visualization of intracellular messenger RNA (mRNA). The complexes were prepared from cationized gelatins with different extents of cationization and different mixing ratios of MB to cationized gelatin. The apparent size of complexes was almost similar, while the zeta potential was different among the complexes. Irrespective of the preparation conditions, the complexes had a sequence specificity against the target oligonucleotides in hybridization. The cytotoxicity and the amount of complexes internalized into cells increased with an increase in the cationization extent and the concentration of cationized gelatin. After the incubation with complexes prepared from cationized gelatin with the highest extent of cationization and at mixing ratios of 10 and 20 pmole MB/μg cationized gelatin, a high fluorescent intensity was detected. On the other hand, the complex prepared with the mixing ratio at 20 pmole/μg did not show any cytotoxicity. The complex was the most effective to visualize the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA endogenously present. In addition, even for enhanced green fluorescent protein (EGFP) mRNA exogenously transfected, the complex permitted to effectively detect it as well. It is concluded that both the endogenous and exogenous mRNA can be visualized in living cells by use of cationized gelatin-MB complexes designed.
Collapse
Affiliation(s)
- Sho Takehana
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yuki Murata
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Jun-ichiro Jo
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
5
|
Bousfiha A, Riahi Z, Elkhattabi L, Bakhchane A, Charoute H, Snoussi K, Bonnet C, Petit C, Barakat A. Further Evidence for the Implication of the MET Gene in Non-Syndromic Autosomal Recessive Deafness. Hum Hered 2019; 84:109-116. [PMID: 31801140 DOI: 10.1159/000503450] [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: 07/02/2019] [Accepted: 09/18/2019] [Indexed: 11/19/2022] Open
Abstract
Mutations in the mesenchymal epithelial transition factor (MET) gene are frequently associated with multiple human cancers but can also lead to human non-syndromic autosomal recessive deafness (DFNB97). In the present study, we identified a novel homozygous missense mutation in the METgene causing a non-syndromic hearing impairment DFNB97 form. Whole-exome sequencing was performed to determine the genetic causes of hearing loss in a Moroccan consanguineous family with an affected daughter. The structural analysis of native and mutant in the SEMA domain of the MET receptor was investigated using a molecular dynamics simulation (MDS) approach. We identified a novel pathogenic homozygous c.948A>G (p.Ile316Met) mutation in the MET gene in one deaf Moroccan young girl carrying a total bilateral non-syndromic hearing impairment. The results of the MDS approach show that an Ile316Met mutation in the SEMA domain leads to protein flexibility loss. This may produce a major impact on the structural conformation of the MET receptor, which also affects the function and binding site of the receptor. This is the first time that a mutation in the MET gene is described in a Moroccan family. Moreover, this study reports the second family in the world associating deafness and mutation in the MET gene.
Collapse
Affiliation(s)
- Amale Bousfiha
- Laboratoire de Génomique et Génétique Humaine, Institut Pasteur du Maroc, Casablanca, Morocco.,Laboratoire de Physiopathologie et Génétique Moléculaire, Faculté des Sciences Ben M'Sik, Université Hassan II, Casablanca, Morocco
| | - Zied Riahi
- INSERM UMRS1120, Institut de la Vision, Paris, France.,UPMC-Sorbonnes Universités Paris VI, Paris, France
| | - Lamiae Elkhattabi
- Laboratoire de Génomique et Génétique Humaine, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Amina Bakhchane
- Laboratoire de Génomique et Génétique Humaine, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Hicham Charoute
- Laboratoire de Génomique et Génétique Humaine, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Khalid Snoussi
- Laboratoire de Génomique et Génétique Humaine, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Crystel Bonnet
- INSERM UMRS1120, Institut de la Vision, Paris, France.,UPMC-Sorbonnes Universités Paris VI, Paris, France
| | - Christine Petit
- INSERM UMRS1120, Institut de la Vision, Paris, France.,UPMC-Sorbonnes Universités Paris VI, Paris, France.,Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, Paris, France.,Collège de France, Paris, France
| | - Abdelhamid Barakat
- Laboratoire de Génomique et Génétique Humaine, Institut Pasteur du Maroc, Casablanca, Morocco,
| |
Collapse
|
6
|
Naz S, Friedman TB. Growth factor and receptor malfunctions associated with human genetic deafness. Clin Genet 2019; 97:138-155. [PMID: 31506927 DOI: 10.1111/cge.13641] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/22/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022]
Abstract
A variety of different signaling pathways are necessary for development and maintenance of the human auditory system. Normal hearing allows for the detection of soft sounds within the frequency range of 20 to 20 000 Hz, but more importantly to perceive the human voice frequency band of 250 to 6000 Hz. Loss of hearing is common, and is a clinically heterogeneous disorder that can be caused by environmental factors such as exposure to loud noise, infections and ototoxic drugs. In addition, variants of hundreds of genes have been reported to disrupt processes required for hearing. Noncoding regulatory variants and variants of additional genes necessary for hearing remain to be discovered as many individuals with inherited deafness are without a genetic diagnosis, despite the advent of whole exome sequencing. Here, we discuss in detail some of these deafness-causing variants of genes encoding a ligand or its receptor. Spotlighted in this review are three growth factor-receptor-pairs EDN3/EDNRB, HGF/MET and JAG/NOTCH, which individually are necessary for normal hearing. We also offer our perspective on unanswered questions, future challenges and potential opportunities for treatments emerging from molecular genetic and mechanistic studies of deafness due to these causes.
Collapse
Affiliation(s)
- Sadaf Naz
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| | - Thomas B Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
7
|
Tai JA, Chang CY, Nishikawa T, Kaneda Y. Cancer immunotherapy using the Fusion gene of Sendai virus. Cancer Gene Ther 2019; 27:498-508. [PMID: 31383952 DOI: 10.1038/s41417-019-0126-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 07/17/2019] [Accepted: 07/20/2019] [Indexed: 12/17/2022]
Abstract
Inactivated Sendai virus particle (or hemagglutinating virus of Japan envelope; HVJ-E) has been previously reported to possess antitumour properties that activate antitumour immunity. Two glycoproteins, fusion (F) and hemagglutinin-neuraminidase (HN), are present on the surface of HVJ-E. HN is necessary for binding to receptors such as acidic gangliosides, and F induces membrane fusion by associating with membrane lipids. We previously reported that liposomes reconstituted with F but not HN showed antitumour activity by inducing IL-6 secretion in dendritic cells (DCs), suggesting that F protein is capable of eliciting antitumour activity. Here, we attempted to deliver F gene into tumour tissue in mice by electroporation and demonstrated that F gene therapy retarded tumour growth, increased CD4+ and CD8+ T-cell infiltration into tumours and induced tumour-specific IFN-γ T-cell response. However, neutralisation of IL-6R signalling did not impact F plasmid-mediated antitumour effect. Instead, we found that F plasmid treatment resulted in a significant increase in the secretion of the chemokine RANTES (regulated upon activation, normal T cell expressed and secreted) by tumour-infiltrating T cells. Neutralising antibody against RANTES abolished the antitumour effect of F plasmid treatment in a dose-dependent manner. Thus, F gene therapy may show promise as a novel therapeutic for single or combined cancer immunotherapy.
Collapse
Affiliation(s)
- Jiayu A Tai
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Chin Yang Chang
- Department of Device Application for Molecular Therapeutics, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Tomoyuki Nishikawa
- Department of Device Application for Molecular Therapeutics, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yasufumi Kaneda
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
8
|
Lin HC, Ren Y, Lysaght AC, Kao SY, Stankovic KM. Proteome of normal human perilymph and perilymph from people with disabling vertigo. PLoS One 2019; 14:e0218292. [PMID: 31185063 PMCID: PMC6559673 DOI: 10.1371/journal.pone.0218292] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/29/2019] [Indexed: 12/20/2022] Open
Abstract
The vast majority of hearing loss, the most common sensory impairment, and vertigo, which commonly causes falls, both reflect underlying dysfunction of inner ear cells. Perilymph sampling can thus provide molecular cues to hearing and balance disorders. While such "liquid biopsy" of the inner ear is not yet in routine clinical practice, previous studies have uncovered alterations in perilymph in patients with certain types of hearing loss. However, the proteome of perilymph from patients with intact hearing has been unknown. Furthermore, no complete characterization of perilymph from patients with vestibular dysfunction has been reported. Here, using liquid-chromatography with tandem mass spectrometry, we analyzed samples of normal perilymph collected from three patients with skull base meningiomas and intact hearing. We identified 228 proteins that were common across the samples, establishing a greatly expanded proteome of the previously inferred normal human perilymph. Further comparison to perilymph obtained from three patients with vestibular dysfunction with drop attacks due to Meniere's disease showed 38 proteins with significantly differential abundance. The abundance of four protein candidates with previously unknown roles in inner ear biology was validated in murine cochleae by immunohistochemistry and in situ hybridization: AACT, HGFAC, EFEMP1, and TGFBI. Together, these results motivate future work in characterizing the normal human perilymph and identifying biomarkers of inner ear disease.
Collapse
Affiliation(s)
- Hsiao-Chun Lin
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton Peabody Laboratories and Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
| | - Yin Ren
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton Peabody Laboratories and Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
| | - Andrew C. Lysaght
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton Peabody Laboratories and Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, United States of America
| | - Shyan-Yuan Kao
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton Peabody Laboratories and Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
| | - Konstantina M. Stankovic
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton Peabody Laboratories and Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, United States of America
- Harvard Program in Therapeutic Science, Harvard University, Boston, United States of America
| |
Collapse
|
9
|
Targeting the cMET pathway augments radiation response without adverse effect on hearing in NF2 schwannoma models. Proc Natl Acad Sci U S A 2018; 115:E2077-E2084. [PMID: 29440379 DOI: 10.1073/pnas.1719966115] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neurofibromatosis type II (NF2) is a disease that needs new solutions. Vestibular schwannoma (VS) growth causes progressive hearing loss, and the standard treatment, including surgery and radiotherapy, can further damage the nerve. There is an urgent need to identify an adjunct therapy that, by enhancing the efficacy of radiation, can help lower the radiation dose and preserve hearing. The mechanisms underlying deafness in NF2 are still unclear. One of the major limitations in studying tumor-induced hearing loss is the lack of mouse models that allow hearing testing. Here, we developed a cerebellopontine angle (CPA) schwannoma model that faithfully recapitulates the tumor-induced hearing loss. Using this model, we discovered that cMET blockade by crizotinib (CRZ) enhanced schwannoma radiosensitivity by enhancing DNA damage, and CRZ treatment combined with low-dose radiation was as effective as high-dose radiation. CRZ treatment had no adverse effect on hearing; however, it did not affect tumor-induced hearing loss, presumably because cMET blockade did not change tumor hepatocyte growth factor (HGF) levels. This cMET gene knockdown study independently confirmed the role of the cMET pathway in mediating the effect of CRZ. Furthermore, we evaluated the translational potential of cMET blockade in human schwannomas. We found that human NF2-associated and sporadic VSs showed significantly elevated HGF expression and cMET activation compared with normal nerves, which correlated with tumor growth and cyst formation. Using organoid brain slice culture, cMET blockade inhibited the growth of patient-derived schwannomas. Our findings provide the rationale and necessary data for the clinical translation of combined cMET blockade with radiation therapy in patients with NF2.
Collapse
|
10
|
Abstract
Many drugs have been developed and optimized for the treatment of cancer; however, it is difficult to completely cure cancer with anticancer drugs alone. Therefore, the development of new therapeutic technologies, in addition to new anticancer drugs, is necessary for more effective oncotherapy. Oncolytic viruses are one potential new anticancer strategy. Various oncolytic viruses have been developed for safe and effective oncotherapy. Recently, Sendai virus-based oncotherapy has been reported by several groups, and attention has been drawn to its unique anticancer mechanisms, which are different from those of the conventional oncolytic viruses that kill cancer cells by cancer cell-selective replication. Here, we introduce Sendai virus-based virotherapy and its anticancer mechanisms.
Collapse
Affiliation(s)
- Kotaro Saga
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yasufumi Kaneda
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
11
|
Mujtaba G, Schultz JM, Imtiaz A, Morell RJ, Friedman TB, Naz S. A mutation of MET, encoding hepatocyte growth factor receptor, is associated with human DFNB97 hearing loss. J Med Genet 2015; 52:548-52. [PMID: 25941349 DOI: 10.1136/jmedgenet-2015-103023] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 04/15/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND Hearing loss is a heterogeneous neurosensory disorder. Mutations of 56 genes are reported to cause recessively inherited non-syndromic deafness. OBJECTIVE We sought to identify the genetic lesion causing hearing loss segregating in a large consanguineous Pakistani family. METHODS AND RESULTS Mutations of GJB2 and all other genes reported to underlie recessive deafness were ruled out as the cause of the phenotype in the affected members of the participating family. Homozygosity mapping with a dense array of one million SNP markers allowed us to map the gene for recessively inherited severe hearing loss to chromosome 7q31.2, defining a new deafness locus designated DFNB97 (maximum logarithm of the odds score of 4.8). Whole-exome sequencing revealed a novel missense mutation c.2521T>G (p.F841V) in MET (mesenchymal epithelial transition factor), which encodes the receptor for hepatocyte growth factor. The mutation cosegregated with the hearing loss phenotype in the family and was absent from 800 chromosomes of ethnically matched control individuals as well as from 136 602 chromosomes in public databases of nucleotide variants. Analyses by multiple prediction programmes indicated that p.F841V likely damages MET function. CONCLUSIONS We identified a missense mutation of MET, encoding the hepatocyte growth factor receptor, as a likely cause of hearing loss in humans.
Collapse
Affiliation(s)
- Ghulam Mujtaba
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| | - Julie M Schultz
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA Present address, GeneDx, Gaithersburg, Maryland, USA
| | - Ayesha Imtiaz
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| | - Robert J Morell
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas B Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Sadaf Naz
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
12
|
Uribe PM, Kawas LH, Harding JW, Coffin AB. Hepatocyte growth factor mimetic protects lateral line hair cells from aminoglycoside exposure. Front Cell Neurosci 2015; 9:3. [PMID: 25674052 PMCID: PMC4309183 DOI: 10.3389/fncel.2015.00003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/05/2015] [Indexed: 11/13/2022] Open
Abstract
Loss of sensory hair cells from exposure to certain licit drugs (e.g., aminoglycoside antibiotics, platinum-based chemotherapy agents) can result in permanent hearing loss. Here we ask if allosteric activation of the hepatocyte growth factor (HGF) cascade via Dihexa, a small molecule drug candidate, can protect hair cells from aminoglycoside toxicity. Unlike native HGF, Dihexa is chemically stable and blood-brain barrier permeable. As a synthetic HGF mimetic, it forms a functional ligand by dimerizing with endogenous HGF to activate the HGF receptor and downstream signaling cascades. To evaluate Dihexa as a potential hair cell protectant, we used the larval zebrafish lateral line, which possesses hair cells that are homologous to mammalian inner ear hair cells and show similar responses to toxins. A dose-response relationship for Dihexa protection was established using two ototoxins, neomycin and gentamicin. We found that a Dihexa concentration of 1 μM confers optimal protection from acute treatment with either ototoxin. Pretreatment with Dihexa does not affect the amount of fluorescently tagged gentamicin that enters hair cells, indicating that Dihexa’s protection is likely mediated by intracellular events and not by inhibiting aminoglycoside entry. Dihexa-mediated protection is attenuated by co-treatment with the HGF antagonist 6-AH, further evidence that HGF activation is a component of the observed protection. Additionally, Dihexa’s robust protection is partially attenuated by co-treatment with inhibitors of the downstream HGF targets Akt, TOR and MEK. Addition of an amino group to the N-terminal of Dihexa also attenuates the protective response, suggesting that even small substitutions greatly alter the specificity of Dihexa for its target. Our data suggest that Dihexa confers protection of hair cells through an HGF-mediated mechanism and that Dihexa holds clinical potential for mitigating chemical ototoxicity.
Collapse
Affiliation(s)
- Phillip M Uribe
- Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA
| | - Leen H Kawas
- Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA ; M3 Biotechnology, Inc. Seattle, WA, USA
| | - Joseph W Harding
- Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA ; M3 Biotechnology, Inc. Seattle, WA, USA
| | - Allison B Coffin
- Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA ; College of Arts and Sciences, Washington State University Vancouver, WA, USA
| |
Collapse
|
13
|
Virosome presents multimodel cancer therapy without viral replication. BIOMED RESEARCH INTERNATIONAL 2013; 2013:764706. [PMID: 24369016 PMCID: PMC3866828 DOI: 10.1155/2013/764706] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 10/31/2013] [Indexed: 12/11/2022]
Abstract
A virosome is an artificial envelope that includes viral surface proteins and lacks the ability to produce progeny virus. Virosomes are able to introduce an encapsulated macromolecule into the cytoplasm of cells using their viral envelope fusion ability. Moreover, virus-derived factors have an adjuvant effect for immune stimulation. Therefore, many virosomes have been utilized as drug delivery vectors and adjuvants for cancer therapy. This paper introduces the application of virosomes for cancer treatment. In Particular, we focus on virosomes derived from the influenza and Sendai viruses which have been widely used for cancer therapy. Influenza virosomes have been mainly applied as drug delivery vectors and adjuvants. By contrast, the Sendai virosomes have been mainly applied as anticancer immune activators and apoptosis inducers.
Collapse
|
14
|
XIA LI, YIN SHANKAI. Local gene transfection in the cochlea (Review). Mol Med Rep 2013; 8:3-10. [DOI: 10.3892/mmr.2013.1496] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 12/13/2012] [Indexed: 11/06/2022] Open
|
15
|
Gene therapy for cisplatin-induced ototoxicity: a systematic review of in vitro and experimental animal studies. Otol Neurotol 2012; 33:302-10. [PMID: 22388732 DOI: 10.1097/mao.0b013e318248ee66] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Ototoxicity is a frequent adverse event of cisplatin treatment. No therapy is currently available for cisplatin-induced ototoxicity. A systematic review of experimental animal studies and in vitro experiments was conducted to evaluate gene therapy as a potential future therapeutic option. DATA SOURCES Eligible studies were identified through searches of electronic databases Ovid MEDLINE, Ovid MEDLINE In-Process, Embase, PubMed, Biosis Previews, Scopus, ISI Web of Science, and The Cochrane Library. STUDY SELECTION Articles obtained from the search were independently reviewed by 2 authors using specific criteria to identify experimental animal studies and in vitro experiments conducted to evaluate gene therapy for cisplatin-induced ototoxicity. No restriction was applied to publication dates or languages. DATA EXTRACTION Data extracted included experiment type, cell type, species, targeted gene, gene expression, method, administration, inner ear site evaluated, outcome measures for cytotoxicity, and significant results. RESULTS Fourteen articles were included in this review. In vitro and in vivo experiments have been performed to evaluate the potential of gene expression manipulation for cisplatin-induced ototoxicity. Twelve different genes were targeted including NTF3, GDNF, HO-1, XIAP, Trpv1, BCL2, Otos, Nfe2l2, Nox1, Nox3, Nox4, and Ctr1. All of the included articles demonstrated a benefit of gene therapy on cytotoxicity caused by cisplatin. CONCLUSION Experimental animal studies and in vitro experiments have demonstrated the efficacy of gene therapy for cisplatin-induced ototoxicity. However, further investigation regarding safety, immunogenicity, and consequences of genetic manipulation in the inner ear tissues must be completed to develop future therapeutic options.
Collapse
|
16
|
Abstract
This protocol describes techniques for construction of fusion-mediated vectors based on inactivated HVJ (hemagglutinating virus of Japan; Sendai virus). HVJ liposomes are constructed by fusing liposomes containing DNA with inactivated HVJ. The HVJ envelope vector, a more simplified vector, incorporates DNA into inactivated HVJ particles without liposomes. Both vectors have many advantages. They can be used to introduce proteins, peptides, oligonucleotides (including antisense oligonucleotides, decoy oligonucleotides, and ribozymes), and short interfering RNA (siRNA), as well as plasmid DNA, into cultured cells in vitro and into organs in vivo. Fusion-mediated delivery avoids the degradation of therapeutic molecules before reaching the cytoplasm. Finally, repeated injection of the vector in vivo is not inhibited and even enhances the effects of the delivered molecules. These vectors have been used in many gene therapy experiments in animal models to address problems such as liver cirrhosis, hearing impairment, ischemic brain damage, peripheral arterial diseases, and cancers. This protocol describes methods for the preparation of HVJ liposomes and of HVJ envelope vectors and their use in delivery of plasmid DNA into various cells and tissues.
Collapse
|
17
|
Devarajan K, Staecker H, Detamore MS. A review of gene delivery and stem cell based therapies for regenerating inner ear hair cells. J Funct Biomater 2011; 2:249-70. [PMID: 24956306 PMCID: PMC4030941 DOI: 10.3390/jfb2030249] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 08/31/2011] [Accepted: 09/05/2011] [Indexed: 12/13/2022] Open
Abstract
Sensory neural hearing loss and vestibular dysfunction have become the most common forms of sensory defects, affecting millions of people worldwide. Developing effective therapies to restore hearing loss is challenging, owing to the limited regenerative capacity of the inner ear hair cells. With recent advances in understanding the developmental biology of mammalian and non-mammalian hair cells a variety of strategies have emerged to restore lost hair cells are being developed. Two predominant strategies have developed to restore hair cells: transfer of genes responsible for hair cell genesis and replacement of missing cells via transfer of stem cells. In this review article, we evaluate the use of several genes involved in hair cell regeneration, the advantages and disadvantages of the different viral vectors employed in inner ear gene delivery and the insights gained from the use of embryonic, adult and induced pluripotent stem cells in generating inner ear hair cells. Understanding the role of genes, vectors and stem cells in therapeutic strategies led us to explore potential solutions to overcome the limitations associated with their use in hair cell regeneration.
Collapse
Affiliation(s)
| | - Hinrich Staecker
- Department of Otolaryngology Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, KS 66160, USA.
| | | |
Collapse
|
18
|
Kopecky B, Fritzsch B. Regeneration of Hair Cells: Making Sense of All the Noise. Pharmaceuticals (Basel) 2011; 4:848-879. [PMID: 21966254 PMCID: PMC3180915 DOI: 10.3390/ph4060848] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 06/04/2011] [Accepted: 06/08/2011] [Indexed: 12/17/2022] Open
Abstract
Hearing loss affects hundreds of millions of people worldwide by dampening or cutting off their auditory connection to the world. Current treatments for sensorineural hearing loss (SNHL) with cochlear implants are not perfect, leaving regenerative medicine as the logical avenue to a perfect cure. Multiple routes to regeneration of damaged hair cells have been proposed and are actively pursued. Each route not only requires a keen understanding of the molecular basis of ear development but also faces the practical limitations of stem cell regulation in the delicate inner ear where topology of cell distribution is essential. Improvements in our molecular understanding of the minimal essential genes necessary for hair cell formation and recent advances in stem cell manipulation, such as seen with inducible pluripotent stem cells (iPSCs) and epidermal neural crest stem cells (EPI-NCSCs), have opened new possibilities to advance research in translational stem cell therapies for individuals with hearing loss. Despite this, more detailed network maps of gene expression are needed, including an appreciation for the roles of microRNAs (miRs), key regulators of transcriptional gene networks. To harness the true potential of stem cells for hair cell regeneration, basic science and clinical medicine must work together to expedite the transition from bench to bedside by elucidating the full mechanisms of inner ear hair cell development, including a focus on the role of miRs, and adapting this knowledge safely and efficiently to stem cell technologies.
Collapse
Affiliation(s)
- Benjamin Kopecky
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
19
|
Sun H, Huang A, Cao S. Current status and prospects of gene therapy for the inner ear. Hum Gene Ther 2011; 22:1311-22. [PMID: 21338273 DOI: 10.1089/hum.2010.246] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Inner ear diseases are common and often result in hearing disability. Sensorineural hearing loss is the main cause of hearing disability. So far, no effective treatment is available although some patients may benefit from a hearing aid equipped with a hearing amplifier or from cochlear implantation. Inner ear gene therapy has become an emerging field of study for the treatment of hearing disability. Numerous new discoveries and tremendous advances have been made in inner ear gene therapy including gene vectors, routes of administration, and therapeutic genes and targets. Gene therapy may become a treatment option for inner ear diseases in the near future. In this review, we summarize the current state of inner ear gene therapy including gene vectors, delivery routes, and therapeutic genes and targets by examining and analyzing publications on inner ear gene therapy from the literature and patent documents, and identify promising patents, novel techniques, and vital research projects. We also discuss the progress and prospects of inner ear gene therapy, the advances and shortcomings, with possible solutions in this field of research.
Collapse
Affiliation(s)
- Hong Sun
- Department of Otolaryngology, Head and Neck Surgery, Xiang Ya Hospital, Central South University, Changsha, Hunan 410008, China
| | | | | |
Collapse
|
20
|
Nakamura T, Mizuno S. The discovery of hepatocyte growth factor (HGF) and its significance for cell biology, life sciences and clinical medicine. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2010; 86:588-610. [PMID: 20551596 PMCID: PMC3081175 DOI: 10.2183/pjab.86.588] [Citation(s) in RCA: 374] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
It has been more than 25 years since HGF was discovered as a mitogen of hepatocytes. HGF is produced by stromal cells, and stimulates epithelial cell proliferation, motility, morphogenesis and angiogenesis in various organs via tyrosine phosphorylation of its receptor, c-Met. In fetal stages, HGF-neutralization, or c-Met gene destruction, leads to hypoplasia of many organs, indicating that HGF signals are essential for organ development. Endogenous HGF is required for self-repair of injured livers, kidneys, lungs and so on. In addition, HGF exerts protective effects on epithelial and non-epithelial organs (including the heart and brain) via anti-apoptotic and anti-inflammatory signals. During organ diseases, plasma HGF levels significantly increased, while anti-HGF antibody infusion accelerated tissue destruction in rodents. Thus, endogenous HGF is required for minimization of diseases, while insufficient production of HGF leads to organ failure. This is the reason why HGF supplementation produces therapeutic outcomes under pathological conditions. Moreover, emerging studies delineated key roles of HGF during tumor metastasis, while HGF-antagonism leads to anti-tumor outcomes. Taken together, HGF-based molecules, including HGF-variants, HGF-fragments and c-Met-binders are available as regenerative or anti-tumor drugs. Molecular analysis of the HGF-c-Met system could provide bridges between basic biology and clinical medicine.
Collapse
Affiliation(s)
- Toshikazu Nakamura
- Kringle Pharma Joint Research Division for Regenerative Drug Discovery, Center for Advanced Science and Innovation, Osaka University, Osaka, Japan.
| | | |
Collapse
|
21
|
Kikkawa YS, Nakagawa T, Tsubouchi H, Ido A, Inaoka T, Ono K, Ito J. Hepatocyte growth factor protects auditory hair cells from aminoglycosides. Laryngoscope 2009; 119:2027-31. [DOI: 10.1002/lary.20602] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
22
|
Schultz JM, Khan SN, Ahmed ZM, Riazuddin S, Waryah AM, Chhatre D, Starost MF, Ploplis B, Buckley S, Velásquez D, Kabra M, Lee K, Hassan MJ, Ali G, Ansar M, Ghosh M, Wilcox ER, Ahmad W, Merlino G, Leal SM, Riazuddin S, Friedman TB, Morell RJ. Noncoding mutations of HGF are associated with nonsyndromic hearing loss, DFNB39. Am J Hum Genet 2009; 85:25-39. [PMID: 19576567 DOI: 10.1016/j.ajhg.2009.06.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 05/22/2009] [Accepted: 06/01/2009] [Indexed: 11/16/2022] Open
Abstract
A gene causing autosomal-recessive, nonsyndromic hearing loss, DFNB39, was previously mapped to an 18 Mb interval on chromosome 7q11.22-q21.12. We mapped an additional 40 consanguineous families segregating nonsyndromic hearing loss to the DFNB39 locus and refined the obligate interval to 1.2 Mb. The coding regions of all genes in this interval were sequenced, and no missense, nonsense, or frameshift mutations were found. We sequenced the noncoding sequences of genes, as well as noncoding genes, and found three mutations clustered in intron 4 and exon 5 in the hepatocyte growth factor gene (HGF). Two intron 4 deletions occur in a highly conserved sequence that is part of the 3' untranslated region of a previously undescribed short isoform of HGF. The third mutation is a silent substitution, and we demonstrate that it affects splicing in vitro. HGF is involved in a wide variety of signaling pathways in many different tissues, yet these putative regulatory mutations cause a surprisingly specific phenotype, which is nonsydromic hearing loss. Two mouse models of Hgf dysregulation, one in which an Hgf transgene is ubiquitously overexpressed and the other a conditional knockout that deletes Hgf from a limited number of tissues, including the cochlea, result in deafness. Overexpression of HGF is associated with progressive degeneration of outer hair cells in the cochlea, whereas cochlear deletion of Hgf is associated with more general dysplasia.
Collapse
|
23
|
Hori R, Nakagawa T, Sugimoto Y, Sakamoto T, Yamamoto N, Hamaguchi K, Ito J. Prostaglandin E receptor subtype EP4 agonist protects cochleae against noise-induced trauma. Neuroscience 2009; 160:813-9. [DOI: 10.1016/j.neuroscience.2009.03.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 02/02/2009] [Accepted: 03/08/2009] [Indexed: 12/19/2022]
|
24
|
Inaoka T, Nakagawa T, Kikkawa YS, Tabata Y, Ono K, Yoshida M, Tsubouchi H, Ido A, Ito J. Local application of hepatocyte growth factor using gelatin hydrogels attenuates noise-induced hearing loss in guinea pigs. Acta Otolaryngol 2009; 129:453-7. [PMID: 19214837 DOI: 10.1080/00016480902725197] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
CONCLUSION Local application of hepatocyte growth factor using biodegradable gelatin hydrogels attenuates noise-induced hearing loss in guinea pigs. OBJECTIVES To develop an inner ear drug delivery system using gelatin hydrogels that is capable of a sustained delivery of growth factors to the cochlea. We examined the efficacy of the local application of gelatin hydrogels containing hepatocyte growth factor (HGF) in protecting cochlear hair cells from noise-induced damage. MATERIALS AND METHODS A piece of gelatin hydrogel previously immersed in either HGF or saline was placed on the round window membrane of a guinea pig 1 h after noise exposure (4 kHz octave band noise at 120 dB sound pressure level for 3 h). Auditory function was monitored using auditory brainstem responses (ABRs), and the loss of hair cells was evaluated quantitatively. RESULTS Local HGF treatment significantly reduced the noise exposure-caused ABR threshold shifts and the loss of outer hair cells in the basal portion of the cochleae.
Collapse
|
25
|
Matsubara H, Tsuchiya H, Watanabe K, Takeuchi A, Tomita K. Percutaneous nonviral delivery of hepatocyte growth factor in an osteotomy gap promotes bone repair in rabbits: a preliminary study. Clin Orthop Relat Res 2008; 466:2962-72. [PMID: 18813894 PMCID: PMC2628245 DOI: 10.1007/s11999-008-0493-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2008] [Accepted: 08/19/2008] [Indexed: 01/31/2023]
Abstract
Hepatocyte growth factor (HGF) was initially identified in cultured hepatocytes and subsequently reported to induce angiogenic, morphogenic, and antiapoptotic activity in various tissues. These properties suggest a potential influence of HGF on bone healing. We asked if gene transfer of human HGF (hHGF) into an osteotomy gap with a hemagglutinating virus of Japan-envelope (HVJ-E) vector promotes bone healing in rabbits. HVJ-E that contained either hHGF or control plasmid was percutaneously injected into the osteotomy gap of rabbit tibias on Day 14. The osteotomy gap was evaluated by radiography, pQCT, mechanical tests, and histology at Week 8. The expression of hHGF was evaluated by reverse transcriptase-polymerase chain reaction and immunohistochemistry at Week 3. Radiography, pQCT, and histology suggested the hHGF group had faster fracture healing. Mechanical tests demonstrated the hHGF group had greater mechanical strength. The injected tissues at 3 weeks expressed hHGF mRNA by reverse transcriptase-polymerase chain reaction. hHGF-positive immunohistochemical staining was observed in various cells at the osteotomy gap at Week 3. The data suggest delivery of hHGF plasmid into the osteotomy gap promotes fracture repair, and HGF could become a novel agent for fracture treatment.
Collapse
Affiliation(s)
- Hidenori Matsubara
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Koji Watanabe
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Akihiko Takeuchi
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Katsuro Tomita
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| |
Collapse
|
26
|
Trapp T, Kögler G, El-Khattouti A, Sorg RV, Besselmann M, Föcking M, Bührle CP, Trompeter I, Fischer JC, Wernet P. Hepatocyte Growth Factor/c-MET Axis-mediated Tropism of Cord Blood-derived Unrestricted Somatic Stem Cells for Neuronal Injury. J Biol Chem 2008; 283:32244-53. [DOI: 10.1074/jbc.m800862200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
27
|
Hughes TS, Langer SJ, Johnson KW, Chavez RA, Watkins LR, Milligan ED, Leinwand LA. Intrathecal injection of naked plasmid DNA provides long-term expression of secreted proteins. Mol Ther 2008; 17:88-94. [PMID: 18941439 DOI: 10.1038/mt.2008.230] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Therapeutic benefit has been reported to result from intrathecal (i.t.) injection of transgene vectors, including naked DNA. However, most studies using naked DNA have measured only the transgene expression of intracellular proteins. Here we demonstrate that i.t. injection of naked DNA can result in long-term expression of secreted proteins. Plasmids expressing either secreted alkaline phosphatase (SEAP) or human interleukin-10 (hIL-10) were injected into the i.t. space in rats, and transgene products were repeatedly measured in the cerebrospinal fluid (CSF). Both SEAP and hIL-10 were maximal at 1 and 2 days after the injection and still detectable at 4 months. The utilization of a plasmid having two features that are hypothesized to increase gene expression (matrix attachment regions (MARs) and lack of CpG dinucleotides) resulted in a significant increase in gene expression. Reinjection of SEAP or hIL-10 plasmids after 4 months significantly increased protein levels at 1 and 14 days after the reinjection. SEAP was uniformly distributed between the DNA delivery site (approximately vertebral level T13) and the lumbar puncture site (L5/L6 inter-vertebral space), was reduced at the cisterna magna, and was detectable, though at much lower levels, in serum. These data suggest that naked DNA has the potential to be used as a therapeutic tool for applications that require long-term release of transgenes into the CSF.
Collapse
Affiliation(s)
- Travis S Hughes
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado 80309-0347, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Alleviation of Abeta-induced cognitive impairment by ultrasound-mediated gene transfer of HGF in a mouse model. Gene Ther 2008; 15:561-71. [PMID: 18288214 DOI: 10.1038/sj.gt.3303094] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A new therapeutic approach to treat Alzheimer's disease (AD) is needed, and the use of growth factors is considered to be a candidate. Hepatocyte growth factor (HGF) is a unique multifunctional growth factor, which has the potential effect to exert neurotrophic action and induce angiogenesis. In this study, we examined the effects of overexpression of human HGF plasmid DNA using ultrasound-mediated gene transfer into the brain in an Abeta-infused cognitive dysfunction mouse model. We demonstrated that HGF gene transfer significantly alleviated Abeta-induced cognitive impairment in mice in behavioral tests. These beneficial effects of HGF might be due to (1) significant recovery of the vessel density in the dentate gyrus of the hippocampus, (2) upregulation of BDNF, (3) a significant decrease in oxidative stress and (4) synaptic enhancement. A pharmacological approach including gene therapy to increase the HGF level in combination with anti-Abeta therapy might be a new therapeutic option for the treatment of AD.
Collapse
|
29
|
Kaneda Y. Applications of Hemagglutinating Virus of Japan in therapeutic delivery systems. Expert Opin Drug Deliv 2008; 5:221-33. [DOI: 10.1517/17425247.5.2.221] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
30
|
Advances in Molecular and Cellular Therapies for Hearing Loss. Mol Ther 2008; 16:224-236. [DOI: 10.1038/sj.mt.6300351] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Accepted: 10/10/2007] [Indexed: 02/07/2023] Open
|
31
|
Fujihara A, Kurooka M, Miki T, Kaneda Y. Intratumoral injection of inactivated Sendai virus particles elicits strong antitumor activity by enhancing local CXCL10 expression and systemic NK cell activation. Cancer Immunol Immunother 2008; 57:73-84. [PMID: 17602226 PMCID: PMC11030187 DOI: 10.1007/s00262-007-0351-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Accepted: 05/30/2007] [Indexed: 11/25/2022]
Abstract
We have already demonstrated that inactivated, replication-defective Sendai virus particles (HVJ-E) have a powerful antitumor effect by both the generation of tumor-specific cytotoxic T cells and inhibition of regulatory T cell activity. Here, we report that HVJ-E also has an antitumor effect through non-T cell immunity. Microarray analysis revealed that direct injection of HVJ-E induced the expression of CXCL10 in established Renca tumors. CXCL10 was secreted by dendritic cells in the tumors after HVJ-E injection. Quantitative real-time RT-PCR and immunohistochemistry revealed that CXCR3+ cells (predominantly NK cells) infiltrated the HVJ-E-injected tumors. Moreover, HVJ-E injection caused systemic activation of NK cells and enhanced their cytotoxity against tumor cells. In an in vivo experiment, approximately 50% of tumors were eradicated by HVJ-E injection, and this activity of HVJ-E against Renca tumors was largely abolished by NK cell depletion using anti-asialo GM1 antibody. Since HVJ-E injection induced systemic antitumor immunity by enhancing or correcting the chemokine-chemokine receptor axis, it might be a potential new therapy for cancer.
Collapse
Affiliation(s)
- Atsuko Fujihara
- Division of Gene Therapy Science, Osaka University Medical School, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
- Department of Urology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masayuki Kurooka
- Division of Gene Therapy Science, Osaka University Medical School, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Tsuneharu Miki
- Department of Urology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasufumi Kaneda
- Division of Gene Therapy Science, Osaka University Medical School, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| |
Collapse
|
32
|
Sharif S, Nakagawa T, Ohno T, Matsumoto M, Kita T, Riazuddin S, Ito J. The potential use of bone marrow stromal cells for cochlear cell therapy. Neuroreport 2007; 18:351-4. [PMID: 17435601 DOI: 10.1097/wnr.0b013e3280287a9a] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study investigated the potential of bone-marrow stromal cell transplantation for cell replacement therapy in the cochlea. Bone-marrow stromal cells labeled with enhanced green fluorescent protein were injected into the perilymphatic space of normal cochleae in mice. Histological analysis 2 weeks after transplantation demonstrated that transplanted cells settled within the cochlear tissues, especially in the spiral ligament and the spiral limbus, although most transplants were located in the perilymphatic space. Some of the transplanted cells expressed the cochlear gap-junction protein connexin 26. These findings indicate the potential of bone-marrow stromal cells for delivering therapeutic molecules and for the restoration of cochlear cells, particularly in the spiral ligament and the spiral limbus.
Collapse
Affiliation(s)
- Sadia Sharif
- Department of Otolaryngology-Head and Neck Surgery, Kyoto University Graduate School of Medicine, Shogoin, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
33
|
Suvanasuthi S, Tamai K, Kaneda Y. Rapid transport of plasmid DNA into the nucleolus via actin depolymerization using the HVJ envelope vector. J Gene Med 2007; 9:55-62. [PMID: 17149789 DOI: 10.1002/jgm.989] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Although nuclear transport of therapeutic genes is an essential requirement of human gene therapy, factors required for nuclear entry of DNA remain to be elucidated. Non-viral vector systems have led to numerous improvements in the efficiency of delivery of exogenous DNA into cells. However, nuclear transport of plasmid is difficult to achieve. METHODS We examined nuclear translocation efficiency of Cy3-labeled plasmid DNA (Cy3-pDNA) delivered by the hemagglutinating virus of Japan envelope (HVJ-E) vector, Lipofectamine or microinjection. We also examined the effect of actin depolymerization on nuclear transport of Cy3-pDNA. RESULTS Cy3-pDNA reached the nucleus, particularly in the nucleolus, in 30 min after fusion-mediated delivery using the HVJ-E vector, while the DNA was retained in the cytoplasm during the observed period after the delivery by cationic liposomes. HVJ-E treatment transiently depolymerized actin filaments, and acceleration of nucleolar entry of microinjected DNA was achieved when treated with either empty HVJ-E or cytochalasin D, an inhibitor of actin depolymerization, prior to microinjection. CONCLUSIONS These results suggest that plasmid DNA can be transported rapidly from the cytoplasm to the nucleolus when actin filaments are depolymerized. Thus, the HVJ-E vector can accelerate the transport of DNA to the nucleolus by actin depolymerization.
Collapse
Affiliation(s)
- Saroj Suvanasuthi
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | |
Collapse
|
34
|
Koyama S, Kimura T, Ogita K, Nakamura H, Tabata C, Md Abu Hadi Noor Ali K, Temma-Asano K, Shimoya K, Tsutsui T, Koyama M, Kaneda Y, Murata Y. Simple and highly efficient method for transient in vivo gene transfer to mid-late pregnant mouse uterus. J Reprod Immunol 2006; 70:59-69. [PMID: 16488483 DOI: 10.1016/j.jri.2005.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2005] [Revised: 10/14/2005] [Accepted: 12/20/2005] [Indexed: 11/24/2022]
Abstract
Up- and down-regulation of various genes in the placenta, decidua and amnion has been reported during the mid-late period of pregnancy and in pregnancy-related complications, such as preeclampsia and preterm labor. However, whether this gene regulation at the feto-maternal interface directly influences the physiology/pathophysiology of disease remains unknown. In order to study this problem, transient gene transfer into the pregnant uterus at mid-late term would be a useful tool. We injected exogenous plasmid entrapped using a commercially available Hemagglutinating Virus of Japan Envelope (HVJ-E) vector system (GenomONE Neo, Ishihara Sangyo) into the extra-amniotic space of the upper part of the pregnant mouse uterus on day 14.5 post-coitus (p.c.). Luciferase activity driven by the cytomegalovirus promoter was detectable for 3 days after transfection in the upper, middle and lower part of the uterus. beta-Galactsidase activity was localized in the basal lamina of the placenta, the decidual membrane and the fetal membrane. Exogenous plasmid was not transmitted to the fetus. The course of pregnancy was not disturbed by this procedure; rupture of membranes, intrauterine fetal growth restriction and preterm birth were not observed. Thus, we demonstrated that this transient gene transfer method is highly efficient and minimally invasive, and expect that this procedure will be a useful tool to analyze the pathophysiology of pregnancy-related disorders.
Collapse
Affiliation(s)
- Shinsuke Koyama
- Division of Obstetrics and Gynecology, Department of Specific Organ Regulation, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Shimamura M, Sato N, Waguri S, Uchiyama Y, Hayashi T, Iida H, Nakamura T, Ogihara T, Kaneda Y, Morishita R. Gene Transfer of Hepatocyte Growth Factor Gene Improves Learning and Memory in the Chronic Stage of Cerebral Infarction. Hypertension 2006; 47:742-51. [PMID: 16505200 DOI: 10.1161/01.hyp.0000208598.57687.3e] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
There is no specific treatment to improve the functional recovery in the chronic stage of ischemic stroke. To provide the new therapeutic options, we examined the effect of overexpression of hepatocyte growth factor (HGF) in the chronic stage of cerebral infarction by transferring the HGF gene into the brain using hemagglutinating virus of Japan envelope vector. Sixty rats were exposed to permanent middle cerebral artery occlusion (day 1). Based on the sensorimotor deficits at day 7, the rats were divided equally into control vector or HGF-treated rats. At day 56, rats transfected with the HGF gene showed a significant recovery of learning and memory in Morris water maze tests (control vector 50±4 s; HGF 33±5 s;
P
<0.05) and passive avoidance task (control vector 132.4±37.5 s; HGF 214.8±26.5 s;
P
<0.05). Although the total volume of cerebral infarction was not related to the outcome, immunohistochemical analysis for Cdc42 and synaptophysin in the peri-infarct region revealed that HGF enhanced the neurite extension and increased synapses. Immunohistochemistry for glial fibriary acidic protein revealed that the formation of glial scar was also prevented by HGF gene treatment. Additionally, the number of the arteries was increased in the HGF group at day 56. These data demonstrated that HGF has a pivotal role for the functional recovery after cerebral infarction through neuritogenesis, improved microcirculation, and the prevention of gliosis. Our results also provide evidence for the feasibility of gene therapy in the chronic stage of cerebral infarction.
Collapse
Affiliation(s)
- Munehisa Shimamura
- Division of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Maiorana CR, Staecker H. Advances in inner ear gene therapy: exploring cochlear protection and regeneration. Curr Opin Otolaryngol Head Neck Surg 2006; 13:308-12. [PMID: 16160526 DOI: 10.1097/01.moo.0000179248.51476.11] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW To review the application of gene therapy in the inner ear. Gene delivery to the inner ear was first reported in 1996. Since then the field has progressed on multiple fronts. RECENT DEVELOPMENTS More diverse and sophisticated vectors are improving the efficiency of delivery to the inner ear. Research is transitioning from the delivery of marker genes to the delivery of therapeutic genes in animal models of inner ear disease. Three distinct areas of research are developing: (1) delivery of genes for protection of spiral ganglion neurons with potential application in cochlear implantation, (2) delivery of genes for protection of hair cells and hearing preservation in degenerative diseases and cochlear insults and (3) the use of gene therapy to transform cells from one phenotype to another and replace lost cells, potentially restoring lost function. SUMMARY Currently, no specific drugs are targeted at inner ear disease. The use of gene therapy in the inner ear is being applied in animal models of ototoxicity and ischemia reperfusion injury. Gene therapy can protect the inner ear from damage and even restore function through the regeneration of hair cells.
Collapse
Affiliation(s)
- Carrie R Maiorana
- Department of Otolaryngology, University of Kansas, Kansas City, KS 66160, USA
| | | |
Collapse
|
37
|
Johanson CE, Duncan JA, Stopa EG, Baird A. Enhanced Prospects for Drug Delivery and Brain Targeting by the Choroid Plexus–CSF Route. Pharm Res 2005; 22:1011-37. [PMID: 16028003 DOI: 10.1007/s11095-005-6039-0] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2005] [Accepted: 04/12/2005] [Indexed: 02/07/2023]
Abstract
The choroid plexus (CP), i.e., the blood-cerebrospinal fluid barrier (BCSFB) interface, is an epithelial boundary exploitable for drug delivery to brain. Agents transported from blood to lateral ventricles are convected by CSF volume transmission (bulk flow) to many periventricular targets. These include the caudate, hippocampus, specialized circumventricular organs, hypothalamus, and the downstream pia-glia and arachnoid membranes. The CSF circulatory system normally provides micronutrients, neurotrophins, hormones, neuropeptides, and growth factors extensively to neuronal networks. Therefore, drugs directed to CSF can modulate a variety of endocrine, immunologic, and behavioral phenomema; and can help to restore brain interstitial and cellular homeostasis disrupted by disease and trauma. This review integrates information from animal models that demonstrates marked physiologic effects of substances introduced into the ventricular system. It also recapitulates how pharmacologic agents administered into the CSF system prevent disease or enhance the brain's ability to recover from chemical and physical insults. In regard to drug distribution in the CNS, the BCSFB interaction with the blood-brain barrier is discussed. With a view toward translational CSF pharmacotherapy, there are several promising innovations in progress: bone marrow cell infusions, CP encapsulation and transplants, neural stem cell augmentation, phage display of peptide ligands for CP epithelium, CSF gene transfer, regulation of leukocyte and cytokine trafficking at the BCSFB, and the purification of neurotoxic CSF in degenerative states. The progressively increasing pharmacological significance of the CP-CSF nexus is analyzed in light of treating AIDS, multiple sclerosis, stroke, hydrocephalus, and Alzheimer's disease.
Collapse
Affiliation(s)
- Conrad E Johanson
- Department of Clinical Neurosciences, Rhode Island Hospital, Brown Medical School, Providence, Rhode Island 02912, USA.
| | | | | | | |
Collapse
|
38
|
Ito M, Yamamoto S, Nimura K, Hiraoka K, Tamai K, Kaneda Y. Rad51 siRNA delivered by HVJ envelope vector enhances the anti-cancer effect of cisplatin. J Gene Med 2005; 7:1044-52. [PMID: 15756713 DOI: 10.1002/jgm.753] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Every cancer therapy appears to be transiently effective for cancer regression, but cancers gradually transform to be resistant to the therapy. Cancers also develop machineries to resist chemotherapy. Short interfering RNA (siRNA) has been evaluated as an attractive and effective tool for suppressing a target protein by specifically digesting its mRNA. Suppression of the machineries using siRNA may enhance the sensitivity to chemotherapy in cancers when combined with an effective delivery system. METHODS To enhance the anti-cancer effect of chemotherapy, we transferred siRNA against Rad51 into various human cancer cells using the HVJ (hemagglutinating virus of Japan, Sendai virus) envelope vector in the presence or absence of cis-diamminedichloroplatinum(II) (CDDP, cisplatin). The inhibition of cell growth was assessed by a modified MTT assay, counting cell number, or fluorescence-activated cell sorting (FACS) analysis after Annexin V labeling. The synthetic Rad51 siRNA was also introduced into subcutaneous tumor masses of HeLa cells in SCID mice with or without intraperitoneal injection of CDDP, and tumor growth was monitored. RESULTS When synthetic Rad51 siRNA was delivered into HeLa cells using the HVJ envelope vector, no Rad51 transcripts were detected on day 2, and Rad51 protein completely disappeared for 4 days after siRNA transfer. When HeLa cells were incubated with 0.02 microg/ml CDDP for 3 h after siRNA transfer, the number of colonies decreased to approximately 10% of that with scrambled siRNA. The sensitivity to CDDP was enhanced in various human cancer cells, but not in normal human fibroblasts. When Rad51 siRNA was delivered into tumors using the HVJ envelope vector, the Rad51 transcript level was reduced to approximately 25%. Rad51 siRNA combined with CDDP significantly inhibited tumor growth when compared to siRNA or CDDP alone. CONCLUSIONS Rad51 siRNA could enhance the sensitivity to CDDP in cancer cells both in vitro and in vivo. Our results suggest that the combination of CDDP and Rad51 siRNA will be an effective anti-cancer protocol.
Collapse
Affiliation(s)
- Makoto Ito
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565 -0871, Japan
| | | | | | | | | | | |
Collapse
|
39
|
Kaneda Y, Yamamoto S, Nakajima T. Development of HVJ Envelope Vector and Its Application to Gene Therapy. NON-VIRAL VECTORS FOR GENE THERAPY, SECOND EDITION: PART 1 2005; 53PA:307-332. [PMID: 16243069 DOI: 10.1016/s0065-2660(05)53012-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
To create a highly efficient vector system that is minimally invasive, we initially developed liposomes that contained fusion proteins from the hemagglutinating virus of Japan (HVJ; Sendai virus). These HVJ-liposomes delivered genes and drugs to cultured cells and tissues. To simplify the vector system and develop more efficient vectors, the next approach was to convert viruses to non-viral vectors. Based on this concept, we recently developed the HVJ envelope vector. HVJ with robust fusion activity was inactivated, and exogenous DNA was incorporated into the viral envelope by detergent treatment and centrifugation. The resulting HVJ envelope vector introduced plasmid DNA efficiently and rapidly into both cultured cells in vitro and organs in vivo. Furthermore, proteins, synthetic oligonucleotides, and drugs have also been effectively introduced into cells using the HVJ envelope vector. The HVJ envelope vector is a promising tool for both ex vivo and in vivo gene therapy experiments. Hearing impairment in rats was prevented and treated by hepatocyte growth factor gene transfer to cerebrospinal fluid using HVJ envelope vector. For cancer treatment, tumor-associated antigen genes were delivered efficiently to mouse dendritic cells to evoke an anti-cancer immune response. HVJ envelope vector fused dendritic cells and tumor cells and simultaneously delivered cytokine genes, such as IL-12, to the hybrid cells. This strategy successfully prevented and treated cancers in mice by stimulating the presentation of tumor antigens and the maturation of T cells. For human gene therapy, a pilot plant to commercially produce clinical grade HVJ envelope vector has been established.
Collapse
Affiliation(s)
- Yasufumi Kaneda
- Division of Gene Therapy Science, Graduate School of Medicine Osaka University, Suita, Osaka 565–0871, Japan
| | | | | |
Collapse
|
40
|
Duan M, Venail F, Spencer N, Mezzina M. Treatment of peripheral sensorineural hearing loss: gene therapy. Gene Ther 2004; 11 Suppl 1:S51-6. [PMID: 15454957 DOI: 10.1038/sj.gt.3302369] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Noise, chemicals and genetic defects are all common causes of irreversible hearing loss, which at present have no cure. Gene therapy may soon be utilized in both the protection and the treatment of these exogenous and endogenous sources of hearing loss. Gene therapy technology is rapidly developing and the inner ear is a particularly feasible model for gene therapy. This review outlines our current understanding of the mechanisms behind deafness and prospects for treatment, discusses the inner ear model in detail and reviews the efforts that have been made in inner ear gene therapy. Finally, the proposed next steps will be discussed. The viral mediated delivery of neurotrophins and antioxidants offers imminent promise in preventing and treating exogenous hearing loss and improving cochlear implant therapy.
Collapse
Affiliation(s)
- M Duan
- Department of Clinical Neuroscience and Center for Hearing and Communication Research, Building MI-ENT, Karolinska Hospital, Stockholm, Sweden
| | | | | | | |
Collapse
|