Your browser doesn't support javascript.
loading
Systemic administration of AAV-Slc25a46 mitigates mitochondrial neuropathy in Slc25a46-/- mice.
Yang, Li; Slone, Jesse; Li, Zhuo; Lou, Xiaoting; Hu, Yueh-Chiang; Queme, Luis F; Jankowski, Michael P; Huang, Taosheng.
Affiliation
  • Yang L; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
  • Slone J; Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
  • Li Z; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
  • Lou X; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
  • Hu YC; State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410078, China.
  • Queme LF; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
  • Jankowski MP; School of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
  • Huang T; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
Hum Mol Genet ; 29(4): 649-661, 2020 03 13.
Article in En | MEDLINE | ID: mdl-31943007
ABSTRACT
Mitochondrial disorders are the result of nuclear and mitochondrial DNA mutations that affect multiple organs, with the central and peripheral nervous system often affected. Currently, there is no cure for mitochondrial disorders. Currently, gene therapy offers a novel approach for treating monogenetic disorders, including nuclear genes associated with mitochondrial disorders. We utilized a mouse model carrying a knockout of the mitochondrial fusion-fission-related gene solute carrier family 25 member 46 (Slc25a46) and treated them with neurotrophic AAV-PHP.B vector carrying the mouse Slc25a46 coding sequence. Thereafter, we used immunofluorescence staining and western blot to test the transduction efficiency of this vector. Toluidine blue staining and electronic microscopy were utilized to assess the morphology of optic and sciatic nerves following treatment, and the morphology and respiratory chain activity of mitochondria within these tissues were determined as well. The adeno-associated virus (AAV) vector effectively transduced in the cerebrum, cerebellum, heart, liver and sciatic nerves. AAV-Slc25a46 treatment was able to rescue the premature death in the mutant mice (Slc25a46-/-). The treatment-improved electronic conductivity of the peripheral nerves increased mobility and restored mitochondrial complex activities. Most notably, mitochondrial morphology inside the tissues of both the central and peripheral nervous systems was normalized, and the neurodegeneration, chronic neuroinflammation and loss of Purkinje cell dendrites observed within the mutant mice were alleviated. Overall, our study shows that AAV-PHP.B's neurotrophic properties are plausible for treating conditions where the central nervous system is affected, such as many mitochondrial diseases, and that AAV-Slc25a46 could be a novel approach for treating SLC25A46-related mitochondrial disorders.
Subject(s)

Full text: 1 Database: MEDLINE Main subject: Ataxia / Genetic Therapy / Central Nervous System Diseases / Dependovirus / Mitochondrial Diseases / Phosphate Transport Proteins / Genetic Vectors Limits: Animals Language: En Year: 2020 Type: Article

Full text: 1 Database: MEDLINE Main subject: Ataxia / Genetic Therapy / Central Nervous System Diseases / Dependovirus / Mitochondrial Diseases / Phosphate Transport Proteins / Genetic Vectors Limits: Animals Language: En Year: 2020 Type: Article