Your browser doesn't support javascript.
loading
Gliosis-dependent expression of complement factor H truncated variants attenuates retinal neurodegeneration following ischemic injury.
Biber, Josef; Jabri, Yassin; Glänzer, Sarah; Dort, Aaron; Hoffelner, Patricia; Schmidt, Christoph Q; Bludau, Oliver; Pauly, Diana; Grosche, Antje.
Afiliación
  • Biber J; Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
  • Jabri Y; Department of Ophthalmology, University Hospital Regensburg, Regensburg, Germany.
  • Glänzer S; Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
  • Dort A; Experimental Ophthalmology, University of Marburg, Marburg, Germany.
  • Hoffelner P; Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
  • Schmidt CQ; Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Center, Ulm, Germany.
  • Bludau O; Institute of Pharmacy, Biochemical Pharmacy Group, Martin Luther University Halle-Wittenberg, Halle, Germany.
  • Pauly D; Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
  • Grosche A; Experimental Ophthalmology, University of Marburg, Marburg, Germany. diana.pauly@uni-marburg.de.
J Neuroinflammation ; 21(1): 56, 2024 Feb 22.
Article en En | MEDLINE | ID: mdl-38388518
ABSTRACT
Inherited, age-related, and acute retinal diseases are often exacerbated by an aberrant or excessive activity of the complement system. Consequently, cells not directly affected by an acute event or genetic variants may degenerate, resulting in enhanced visual impairment. The therapeutic potential of supplementation of complement factor H (FH), a key regulator of the complement cascade, is therefore particularly promising in the context of retinal diseases caused by complement activation. In this study, we engineered adeno-associated viruses (AAVs) containing sequences of two truncated human FH variants. The expression of these variants was regulated by the glial fibrillary acidic protein (GFAP) promoter, which is selectively active in gliotic Müller cells. Both FH variants consisted of FH domains 19-20, which were connected to domains 1-4 and 1-7, respectively, by a polyglycine linker. These AAVs were intravitreally injected following ischemic injury of C57BL/6J mouse retinas. We observed transgene expression in gliotic Müller cells and to some extent in astrocytes. The expression correlated directly with damage severity. Interventions resulted in decreased complement activation, accelerated normalization of microglia activity and morphological improvements. Reduced levels of C3 transcripts and C3d protein in conjunction with higher transcript levels of inhibitory regulators like Cfi and Cfh, hinted at attenuated complement activity. This study demonstrates the great potential of complement regulatory gene addition therapy. With further in vivo testing it could be applied to treat a wide range of retinal diseases where no causative therapies are available.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Enfermedades de la Retina / Gliosis Límite: Animals / Humans Idioma: En Revista: J Neuroinflammation Asunto de la revista: NEUROLOGIA Año: 2024 Tipo del documento: Article País de afiliación: Alemania

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Enfermedades de la Retina / Gliosis Límite: Animals / Humans Idioma: En Revista: J Neuroinflammation Asunto de la revista: NEUROLOGIA Año: 2024 Tipo del documento: Article País de afiliación: Alemania