Synthetic human ABCB4 mRNA therapy rescues severe liver disease phenotype in a BALB/c.Abcb4-/- mouse model of PFIC3

Progressive familial intrahepatic cholestasis type 3 (PFIC3) is a rare lethal autosomal recessive liver disorder caused by loss-of-function variations of the ABCB4 gene, encoding a phosphatidylcholine transporter (ABCB4/MDR3). Currently, no effective treatment exists for PFIC3 outside of liver trans...

Full description

Saved in:
Bibliographic Details
Published inJournal of hepatology Vol. 74; no. 6; pp. 1416 - 1428
Main Authors Wei, Guangyan, Cao, Jingsong, Huang, Pinzhu, An, Ping, Badlani, Disha, Vaid, Kahini A., Zhao, Shuangshuang, Wang, David Q-H., Zhuo, Jenny, Yin, Ling, Frassetto, Andrea, Markel, Arianna, Presnyak, Vladimir, Gandham, Srujan, Hua, Serenus, Lukacs, Christine, Finn, Patrick F., Giangrande, Paloma H., Martini, Paolo G.V., Popov, Yury V.
Format Journal Article
LanguageEnglish
Published Netherlands Elsevier B.V 01.06.2021
Elsevier Science Ltd
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Progressive familial intrahepatic cholestasis type 3 (PFIC3) is a rare lethal autosomal recessive liver disorder caused by loss-of-function variations of the ABCB4 gene, encoding a phosphatidylcholine transporter (ABCB4/MDR3). Currently, no effective treatment exists for PFIC3 outside of liver transplantation. We have produced and screened chemically and genetically modified mRNA variants encoding human ABCB4 (hABCB4 mRNA) encapsulated in lipid nanoparticles (LNPs). We examined their pharmacological effects in a cell-based model and in a new in vivo mouse model resembling human PFIC3 as a result of homozygous disruption of the Abcb4 gene in fibrosis-susceptible BALB/c.Abcb4-/- mice. We show that treatment with liver-targeted hABCB4 mRNA resulted in de novo expression of functional hABCB4 protein and restored phospholipid transport in cultured cells and in PFIC3 mouse livers. Importantly, repeated injections of the hABCB4 mRNA effectively rescued the severe disease phenotype in young Abcb4-/- mice, with rapid and dramatic normalisation of all clinically relevant parameters such as inflammation, ductular reaction, and liver fibrosis. Synthetic mRNA therapy also promoted favourable hepatocyte-driven liver regeneration to restore normal homeostasis, including liver weight, body weight, liver enzymes, and portal vein blood pressure. Our data provide strong preclinical proof-of-concept for hABCB4 mRNA therapy as a potential treatment option for patients with PFIC3. This report describes the development of an innovative mRNA therapy as a potential treatment for PFIC3, a devastating rare paediatric liver disease with no treatment options except liver transplantation. We show that administration of our mRNA construct completely rescues severe liver disease in a genetic model of PFIC3 in mice. [Display omitted] •Synthetic liver-targeted hABCB4 mRNA therapy was designed for PFIC3, a devastating rare paediatric liver disease.•Single injection restores hepatocyte ABCB4 expression and biliary phosphatidylcholine secretion in a genetic model of PFIC3.•Repeated administration rapidly and completely rescues PFIC3 disease in young Abcb4-/- mice.•Treatment ameliorated liver injury, inflammation, ductular reaction, fibrosis, portal hypertension and ‘failure to thrive’.
Bibliography:These authors share senior authorship.
Authors’ contributions
Study concept and design: PGVM, YVP. Designed experiments: GW, JC, PZH. Designed the mRNA constructs: VP, CL. Important intellectual input on study design of bile duct cannulation experiments: DQHW. Conducted experiments: PZH, DB, PA, KV, SSZ, JZ, LY, AF, AM, SG, SH. Conducted most experiments: GW, JC. Data acquisition: DB, PA, KV, SSZ, JZ, LY, AF, AM. Data analysis: GW, JC, PZH, SG, SH, YVP. Data interpretation: DQHW. Interpretation of results: YVP. Study supervision: PF, PHG, PGVM, YVP. Wrote the manuscript: GW, YVP. Edited the manuscript: VP, CL, SG, SH, DQHW, PGVM. Revised the manuscript: GW, JC, PZH. Critically revised the manuscript: PF, PHG. Read and approved the manuscript: all authors.
These authors contributed equally.
ISSN:0168-8278
1600-0641
DOI:10.1016/j.jhep.2020.12.010