Elsevier

Biomaterials

Volume 241, May 2020, 119906
Biomaterials

Membrane fusion FerA domains enhance adeno-associated virus vector transduction

https://doi.org/10.1016/j.biomaterials.2020.119906Get rights and content

Highlights

  • Diverse FerA domains augment AAV transduction in AAV serotypes-independent manner.

  • FerA domains directly interact with AAV virion.

  • FerA domains fuel AAV binding, trafficking, and traversing ability.

  • FerA domains slow down the blood clearance of AAV.

  • ScAAV8/hFIX-FerA injection improves hemostasis in FIX-/- hemophilia B mice.

Abstract

The recombinant adeno-associated virus (rAAV) vector has been successfully employed in clinical trials for patients with blindness and bleeding diseases as well as neuromuscular disorders. To date, it remains a major challenge to achieve higher transduction efficiency with a lower dose of rAAV vector. Our previous studies have demonstrated that serum proteins are able to directly interact with AAV virions for transduction enhancement. Herein, we explored the effect of the FerA domains, which are derived from ferlin proteins and possess membrane-fusion activity, on AAV transduction. Our results show that FerA domains from dysferlin, myoferlin, and otoferlin proteins are able to directly interact with AAV vectors and enhance AAV transduction in vitro and in mice through either intravenous or intramuscular injections. The enhanced AAV transduction induced by human/mouse FerA domains is achieved in various cell lines and in mice regardless of AAV serotypes. Mechanism studies demonstrated that the FerA domains could effectively enhance the ability of AAV vectors to bind to target cells and cross the vascular barrier. Additionally, FerA domains slow down the blood clearance of AAV. Systemic administration of AAV8/hFIX-FerA complex induced approximate 4-fold more human coagulation factor IX expression and improved hemostasis in hemophilia B mice than that of AAV8/hFIX. Collectively, we show, for the first time, that multiple FerA domains could be tethered on the AAV capsid and enhance widespread tissue distribution in an AAV serotypes-independent manner. This approach therefore holds a promise for future clinical application.

Introduction

Recombinant adeno-associated virus (rAAV) vector has been a leading vehicle for clinical application of gene therapies in view of its safety, broad tissue tropism, and sustained effectiveness [1,2]. Currently, 13 serotypes and numerous AAV variants and mutants have been isolated and studied as gene delivery vehicles [3]. Several AAV serotypes, such as AAV2, AAV8, and AAV9, have been extensively employed in clinical trials and achieved therapeutic effects [[4], [5], [6]]. Nevertheless, emerging concerns about the rAAV limited transduction efficacy and the high vector dose requirement remain crucial barriers for ongoing AAV-based gene therapy in preclinical and clinical settings [7,8]. Data from AAV-human factor IX (hFIX)-related clinical trials in hemophilia B patients have shown that high vector dose directly correlates with the cellular immune responses to vector capsid [9,10]. Meanwhile, several studies have demonstrated that AAV capsid or transgene-specific cytotoxic T lymphocytes (CTLs) response and late innate immune activation after long-term AAV transduction hinder the AAV vector transduction efficiency [[10], [11], [12]], which could be strengthened under the condition of high dose of vehicles. In this context, tremendous efforts have been directed to improve the AAV vector transduction efficacy while simultaneously decreasing the vector dose. Several approaches, including but not limited to direct evolution, capsid/genome engineering and polyploid AAV capsids modification, have been undertaken in an effort to produce and impel AAV variants into desired tissues or cell types and achieve therapeutic efficacy [[13], [14], [15], [16], [17]]. Meanwhile, numerous studies with cell surface targeting of AAV vectors by genetic modification of the capsid proteins VP1, VP2, or VP3 have been extensively explored to improve safety and efficacy in preclinical/clinical application [[18], [19], [20]]. Additionally, covalent coupling of targeting ligands to intact AAV particles and magnetically guided AAV delivery system have been also exploited to achieve selective gene transfer in distinct cell types, even in non-permissive cell types [21,22]. However, the achieved results being implemented in specific mouse strains and other small-animal models cannot always be extrapolated to that of other species such as non-human primates (NHPs) [[23], [24], [25], [26]]. One study has shown that the therapeutic efficacy for liver transduction in large-animal models compared with mouse models is approximately 50-100-fold less efficient [27]. Notably, engineered modification of the AAV capsid may lead to unpredicted structural and tropism changes [28]. Although those risks could be mitigated or eliminated by direct evolution approaches [17], the most mutants developed from direct evolution were isolated from cell lines in vitro or in animal models and have unknown human tropism. Therefore, further exploration of safer and more effective strategies to improve AAV transduction at a lower vector dose, especially in a manner that induces consistent transgene expression without limitation of species and alteration of tissue tropism, is still of critical clinical importance.

It has been demonstrated by multiple studies that serum proteins effectively bind to AAV and affect transduction efficacy via different mechanisms in a serotypes- and species-specific manner [[29], [30], [31], [32], [33], [34], [35]]. For instance, the mouse-derived but not human-derived C-reactive protein can bind to the AAV6 virions [29]. However, both of them did not appreciably interact with either AAV8 or AAV9 [35]. Our recent studies have demonstrated that several human serum proteins are able to directly interact with the specific AAV capsid and increase its transduction [[32], [33], [34]]. Pei et al. found that the low-density lipoprotein and transferrin could be appropriated by AAV8 virions to enhance the liver transduction by increasing AAV's ability to bind to target cells, although the enhancement is only limited to hepatocytes after systemic administration [33]. Chai et al. has demonstrated that serum proteins could directly interact with AAV9 and further enhance AAV9 vascular permeability for global transduction [34]. Nevertheless, it is worthy to note that the effect of serum proteins may have limited application only to specific AAV serotypes [29,30,[32], [33], [34], [35]]. In light of the described events, it will undoubtedly require the exploration of novel proteins with the characterization of generally working with multiple AAV serotypes to achieve broader AAV preclinical/clinical application.

Ferlins are a family of type II transmembrane proteins and are characterized by the multiple tandem C2 domains (Ca2+-regulated, phospholipid-binding domains), a centrally positioned FerA domain, and anchored by a C-terminal transmembrane domain [36,37]. Ferlins are implicated in membrane repair and membrane fusion processes. To date, there are six Fer-1-like genes that form the ferlin family: dysferlin (Fer1L1), otoferlin (Fer1L2), and myoferlin (Fer1L3), as well as three additional yet not well-characterized members Fer1L4, Fer1L5, and Fer1L6 in humans and most mammals [38]. A recent study found that the α-helical ~15-kDa FerA domain possesses Ca2+-dependent membrane-associating activity, and may contribute to the overall membrane fusion activity of ferlin proteins [39]. If the isolated FerA domain can help mediate vesicle fusion, can it then be used to improve the AAV transduction process?

In the present study, we demonstrated for the first time that the isolated FerA domains from the dysferlin, myoferlin, and otoferlin proteins could generally enhance the AAV transduction in vitro and in vivo in a species- and AAV serotypes-independent manner, thus overcoming some of the limitations of the previous AAV adjuvants. Underlying mechanism studies suggest that FerA domains directly bind to AAV, promote the AAV binding activity, delay the blood clearance, and enhance the transcytosis ability of AAV. We applied this approach for treating FIX-deficient hemophilic mice. After systemic administration of self-complementary (sc) AAV8/hFIX vector preincubated with FerA domain, enhanced transgene hFIX expression and hemostasis improvement were achieved.

Section snippets

Cell lines

Huh7, HeLa, and C2C12 cells were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% (v/v) heat inactivated fetal bovine serum (FBS), and antibiotics (penicillin 100-U/mL and streptomycin 100-μg/mL). Human brain endothelial capillary hCMEC/D3 cells (Millipore Sigma, USA), as a model of human blood-brain barrier (BBB), were cultured as previously described [40]. All cells were maintained at 37 °C in an atmosphere of 5% CO2.

Expression and purification of FerA domains

FerA samples were prepared as previously

Mouse otoferlin FerA enhances the transduction of multiple AAV serotypes in vitro without cytotoxicity

Our previous studies have shown that several serum proteins could directly and specifically bind to several AAV serotypes for transduction enhancement [[32], [33], [34]]. To study whether the FerA domains from ferlin proteins have an effect on AAV transduction, we first investigated the effect of the FerA from mouse otoferlin (mOto-FerA) on AAV transduction in vitro. The purity of AAV vectors were confirmed by SDS-PAGE and Western blot, which showed no protein contamination in all the AAV

Discussion

In the current study, we demonstrated that isolated FerA domains derived from different ferlin proteins were able to serve as “native molecules” and augment divergent AAV serotypes transduction by systemic or intramuscular administration. Mechanism researches showed that the FerA domain could directly interact with AAV, increase AAV's binding activity on target cells, and its transcytosis ability. Meanwhile, it could delay the clearance of AAV particles from circulation to enhance AAV global

Conclusion

In summary, we provide a proof of concept experiment that the FerA domains from different ferlin proteins could be leveraged as an “assistant cargo” to augment gene delivery of multiple AAV serotypes without altering inherent tissue tropism. Further deciphering the structural features of how the FerA domain interacts with AAV virions may assist in the design of more efficient AAV vectors for future clinical application.

Author contributions

X. Z and C.L designed the study. X.Z implemented experiments, collected and analyzed the data, and wrote the initial manuscript; B.A contributed to the expression and purification of multiple FerA domains. Z.C and X.Z contributed to the tissue processing, hFIX expression, aPTT activity assay, and discussion; R.B.S discussed the results, provided advice, and revised the manuscript; C.L supervised the project and revised the manuscript. All authors reviewed, edited, and approved the manuscript.

Declaration of competing interest

Chengwen Li and Roger Bryan Sutton are cofounders of Bedrock Therapeutics, Inc. Chengwen Li has licensed patents by UNC and received royalties from Bedrock Therapeutics and Asklepios BioPharmaceutical, Inc.

Acknowledgments

We would like to thank Ellie Frost and Taylor Ralph for their great technical assistance. We acknowledge the UNC Biomedical Research Imaging Center (BRIC) Small Animal Imaging (SAI) facility for assistance with mouse imaging and UNC Histology Research Core Facility for histological services. This work was supported by National Institutes of Health Grants R01AI117408, R01HL144661, R01HL125749 (to C.L.), R01AR063634 (to R.B.S), P30-CA016086-35-37, and U54-CA151652-01-04 (to the BRIC SAI facility).

References (53)

  • G.D. Hurlbut et al.

    Preexisting immunity and low expression in primates highlight translational challenges for liver-directed AAV8-mediated gene therapy

    Mol. Ther. : the journal of the American Society of Gene Therapy

    (2010)
  • J. Denard et al.

    AAV-8 and AAV-9 vectors cooperate with serum proteins differently than AAV-1 and AAV-6

    Molecular therapy Methods & clinical development

    (2018)
  • K. Rapti et al.

    Neutralizing antibodies against AAV serotypes 1, 2, 6, and 9 in sera of commonly used animal models

    Mol. Ther.

    (2012)
  • X. Pei et al.

    AAV8 virions hijack serum proteins to increase hepatocyte binding for transduction enhancement

    Virology

    (2018)
  • Z. Chai et al.

    Cryoprecipitate augments the global transduction of the adeno-associated virus serotype 9 after a systemic administration

    J. Contr. Release : official journal of the Controlled Release Society

    (2018)
  • A.D. Posey et al.

    Ferlin proteins in myoblast fusion and muscle growth

    Curr. Top. Dev. Biol.

    (2011)
  • C. Therrien et al.

    Mutation impact on dysferlin inferred from database analysis and computer-based structural predictions

    J. Neurol. Sci.

    (2006)
  • X. Zhang et al.

    Blood-brain barrier shuttle peptides enhance AAV transduction in the brain after systemic administration

    Biomaterials

    (2018)
  • Z. Wu et al.

    Optimization of self-complementary AAV vectors for liver-directed expression results in sustained correction of hemophilia B at low vector dose

    Mol. Ther. : the journal of the American Society of Gene Therapy

    (2008)
  • D.Y. Jin et al.

    Creation of a mouse expressing defective human factor IX

    Blood

    (2004)
  • Z. Wu et al.

    Optimization of self-complementary AAV vectors for liver-directed expression results in sustained correction of hemophilia B at low vector dose

    Mol. Ther.

    (2008)
  • S. Yasunaga et al.

    OTOF encodes multiple long and short isoforms: genetic evidence that the long ones underlie recessive deafness DFNB9

    Am. J. Hum. Genet.

    (2000)
  • N.M. Kotchey et al.

    A potential role of distinctively delayed blood clearance of recombinant adeno-associated virus serotype 9 in robust cardiac transduction

    Mol. Ther. : the journal of the American Society of Gene Therapy

    (2011)
  • K.R. Doherty et al.

    Repairing the tears: dysferlin in muscle membrane repair

    Trends Mol. Med.

    (2003)
  • S.C. Harrison

    Viral membrane fusion

    Virology

    (2015)
  • L.-Y. Huang et al.

    Parvovirus glycan interactions

    Current Opinion in Virology

    (2014)
  • Cited by (6)

    View full text