Sie sind auf Seite 1von 7

Send Orders for Reprints to reprints@benthamscience.

ae 43
Current Gene Therapy, 2017, 17, 43-49

REVIEW ARTICLE
ISSN: 1566-5232
eISSN: 1875-5631

Perspectives of Gene Therapies in Autosomal Dominant Polycystic Kidney Impact


Factor:
2.78

Disease
BENTHAM
SCIENCE

Yuchen Xu1, Ao Li1, Guanqing Wu1,2,* and Chaozhao Liang1,*

1
Institute/Department of Urology, The Anhui Province PKD Center, The First Affiliated Hospital of Anhui Medical Uni-
versity, Hefei, Anhui Province, 230022, China; 2State Key Laboratory of Molecular Oncology, Cancer Hospital and
Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China

Abstract: Background: Autosomal dominant polycystic kidney disease (ADPKD) is the most com-
mon inherited kidney disease in the clinic. The predominant clinical manifestation is bilateral and pro-
gressive cysts formation in the kidneys, impairs normal renal parenchyma, and ultimately leads to end-
stage renal disease (ESRD). ADPKD is a heterogenic disease which is resulted from the mutations of
ARTICLE HISTORY PKD1 or PKD2 genes which encode polycystin-1 (PC1) and -2 (PC2), thereby multiple cell signaling
Received: April 03, 2017 pathways are involved.
Revised: May 07, 2017
Accepted: May 09, 2017 Method: Although causative genes and aberrant signaling pathways have been investigated for dec-
ades, lack of effective and less side-effect treatment for the disease still perplex vast clinicians. There-
DOI:
10.2174/1566523217666170510152808 fore, development of new therapeutic approaches for ADPKD is currently very much desired.
Conclusion: This review will center on pathogenesis of ADPKD, and thereafter gene transfer will be
discussed as potential treatment for the disease. New therapeutic interventions will bring further hope
Current Gene Therapy

to improve prognosis of this incurable disease.

Keywords: Aberrant signaling pathways, ADPKD, causative genes, gene therapy, kidney disease, translational medicine.

1. INTRODUCTION 2. GENETIC CHARACTERISTICS OF ADPKD


Autosomal dominant polycystic kidney disease ADPKD is a heterogenic disease resulted from the muta-
(ADPKD) is the most common genetic disease of the kidney. tions in the PKD1 or PKD2 genes. The PKD1 gene (~50kb)
In the past decades, diverse morbidities (from 1/4000 to located on chromosome 16p13.3, contains 46 exons and en-
1/400) have been reported in different regions of the world codes transcript with an open reading frame of 12909 bp
and a European incidence of the disease is recently consid- [18]. In addition, there are 6 pseudogenes on the upstream of
ered to be 1/2500 [1-3]. The main life-threatening manifesta- the same chromosomal location which are highly homolo-
tion of ADPKD is renal cyst growth which would eventually gous with exons 1 to 33 of PKD1, complicating mutation
destroy parenchyma and ruin function of the kidney. The detection and molecular diagnosis [19]. Another ADPKD
extrarenal lesions include cystic liver and pancreas [4-6], causal gene PKD2 (68 kb) locates on chromosome 4q22,
intracranial aneurysm [7-10], cardiovascular abnormalities contains 15 exons, encodes transcript with an ORF of 2904
[11-15], colon diverticulum and inguinal hernia [16, 17]. bp [20]. It is generally thought that 85% mutations occur in
ADPKD patients account for approximate 10% population of PKD1 and 15% in PKD2 [21-23]. However, an investigation
patients that need to receive renal replacement therapy in North America showed a relative high prevalence of
(RRT) and became the fourth most common disease for RRT PKD2, accounting for 26% of the patients [24], and some
[1, 2]. Despite first reported 500 years ago [2], ADPKD has reports indicated that much lower prevalence (<10%) of
not been clearly understood and still challenges clinicians as PKD2 mutations was seen in Chinese populations [25, 26].
a refractory disease due to lack of effective and safe treat- Furthermore, in approximate 10% of ADPKD patients, no
ment. With intensive reveal of pathogenesis and translational mutation was detected in PKD1 or PKD2 genes [21, 27]. The
study towards clinic, new hopes emerging on finding potential latest study reported that mutation in the gene encoding
therapeutic targets and novel treatment interventions, which glucosidase IIα subunit (GANAB) could disrupt normal ex-
may delay or halt progression of this disease. pression and function of PC1, thereby inducing ADPKD
phenotypes [27]. This finding indicates that other gene muta-
tions may also be responsible for the onset of ADPKD.
*Address correspondence to these authors at the Anhui Province PKD Cen-
ter, Institute/Department of Urology, The First Affiliated Hospital, Anhui The PKD1 gene encodes protein with 4303 amino acids
Medical University, Hefei, 230022, China; Tel: (86) 0551+62923001; Fax: and named as polycistin-1(PC1). PC1 consists of a giant
(86)0551+62923932;
E-mails: guanqing.wu@ahmu.edu.cn; liang_chaozhao@ahmu.edu.cn
extracellular domain, 11 transmembrane domains and a short

1875-5631/17 $58.00+.00 © 2017 Bentham Science Publishers


44 Current Gene Therapy, 2017, Vol. 17, No. 1 Xu et al.

cytoplasmic tail with a coiled-coil domain [28, 29]. At the homozygous Pkd2nf3 alleles could retain one third functional
early stage of embryonic development or cell sub-fusion PC2 expression and developed hepatorenal cysts without
status, PC1, paxillin, α-actinin and tensin were co-localized embryonic lethality [58]. Similar result was also found in a
at the focal adhesions. While in mature epithelial cells with Pkd1 mutant mouse model (Pkd1nl) [59]. In addition, incom-
polarity, PC1 and E-cadherin were located at adhesion junc- plete penetrance of mutated PKD1 or PKD2 genes has also
tion of cells [30]. PC1 also exists in the primary cilia and the been discovered in human ADPKD patients [60]. If an
centrosome of epithelial cells. It was therefore considered to ADPKD patient inherited with one complete inactivated al-
play important roles in the mechano/chemosensory functions lele and another allele with incomplete penetrance, early
of primary cilia in epithelium [31-36]. onset diseases phenotypes would occur [61]. These finding
indicated that cysts formation could be induced when PCs
PKD2 encodes polycystin-2 (PC2), a protein with 968
amino acids which contains 6 transmembrane domains and expression was under a specific threshold, and the disease
severity are dose-dependently associated with residual func-
cytoplasmic N- and C-termini [20, 37, 38]. There is a Ca2+
tional PCs [58, 60, 62, 63].
binding motif, EF hand domain, in the C-terminus of PC2,
and is therefore named as TRPP2 which belongs to transient Non-genetic factors such as ischemic renal injury [64-
receptor potential channel superfamily [27, 39, 40]. Four 67], toxic renal injury [68] and compensatory renal hypertro-
PC2 constitute a membrane cation channel which is more phy [69] could also induce or accelerate cystogenesis in the
selective to Na+ and K+ than Ca2+ [38, 41]. PC2 is expressed ADPKD models and patients. Prevention of these factors
along all segments of the nephrons except glomeruli. At sub- may improve prognosis of ADPKD patients.
cellular level, PC2 is localized in the endoreticulum and pri-
mary cilia [42, 43]. There is also a coiled-coil domain in C- 4. TRANSLATIONAL MEDICINE IN ADPKD
terminus of PC2 which can physically bind to the coiled-coil
Toward curing ADPKD, considerable progresses and un-
domain of PC1. This interaction contributes to the formation
of PC1/PC2 complex, by which multiple downstream signal- derstanding have been recently achieved in the molecular
mechanism and pathogenesis of ADPKD [1, 2, 70]. Most
ing pathways are regulated [31, 37, 44-47].
studies indicated the cyst formation of ADPKD can be re-
As the most common causal gene, PKD1 mutations often sulted from both excessive fluid secretion into cyst lumens
lead to more severe clinical manifestations in ADPKD pa- and/or abnormal proliferation of the cyst-lining epithelium
tients, with a relative early onset of ESRD 20 years ahead [32]. The molecules rectifying the both epithelial defects by
compared to the patients with PKD2 mutations (58.1 vs. 79.7 regulating diverse associated cell signalings have been inten-
years old, respectively) [48]. Based on the data of ADPKD sively investigated [34, 35, 38, 46, 55, 71]. Based on the
Mutation Database (PKDB), 2323 mutations of PKD1 were findings, multiple clinical trials with the cell signaling acti-
identified in 2080 pedigrees, and 278 PKD2 mutations were vators or inhibitors have been carried out and showed low-
identified in 463 pedigrees [49]. Ever-increasing list of new ered cyst formation in ADPKD patients, but only tolvaptan, a
mutations is continuously discovered in both causal genes vasopressin V2 receptor antagonist, has improved the renal
[26]. In ADPKD patients, over 70% of causal mutations was function in the patients [72-80]. Since conspicuous side ef-
identified to be truncating mutations, and remaining nont- fects of tolvaptan, its wide-application in clinic has been
runcating mutations include missense mutations, in-frame challenged [81, 82]. Development of more effective thera-
deletions/insertions or splicing mutations [21]. Some of these peutic interventions for human ADPKD is clinically desired.
nontruncating mutations might be hypomorphic since they
could preserve functional residues and/or exert partial func- 5. GENE THERAPY IN ADPKD
tions. Compared to patients with truncating mutations, pa-
tients with nontruncating mutations could have mild disease In consideration of the inherited nature of ADPKD, gene
phenotypes, presenting relative late onset of ESRD (67 vs. 55 replacement therapy holds great promise although no clinical
years old, respectively) [48, 50]. trial has yet been reported. Exogenously transferring wild-
type PKD1 or PKD2 genes into tubular epithelia of the kid-
3. PATHOGENESIS OF ADPKD ney may have an inhibiting potential on cystogenesis in
ADPKD patients. Recently, we have established a mouse
The most widely accepted theory of ADPKD pathogene- model with human PKD2 transgene (PKD2tg). Although
sis is “two-hit hypothesis”. Since homozygous mutations of highly expressed human PC2 was detected in diverse tissues
PKD1 or PKD2 are embryonic lethal, most ADPKD patients and organs at varying degrees (e.g. sevenfold in the heart,
have typical heterozygous genotypes, and the mutant allele fourfold in the kidney, and twofold in the liver and pan-
obtained by inheritance is the primary hit [51-55]. The sec- creas), mice with hemizygous and homozygous PKD2 trans-
ond hit refers to mutations of the remaining normal allele gene(s) did not exhibited any deleterious effects during em-
which occur in the growth and differentiation of somatic bryogenesis and organogenesis [83]. We applied this trans-
cells, leading to the homozygous inactivation of PKD1 or genic model crossing with Pkd2 mutant mice which we have
PKD2 at the cellular level. If the second hit occurred in a generated previously [53-55, 84]. Resulting Pkd2 mutant
renal tubular epithelium, the cell will be abnormally trans- mice with human PKD2 transgene(s) (e.g. Pkd2-/-; PKD2tg,
formed to cyst epithelium and promoted cyst formation in Pkd2-/-; PKD2tg/tg, Vil-Cre; Pkd2f3/f3; PKD2tg or Vil-Cre;
the kidney [53, 56, 57]. Pkd2f3/f3; PKD2tg/tg) rescued all disease phenotypes in PC2
The two-hit theory is not the only pathogenesis of dose-dependent manner [83] (and unpublished observations).
ADPKD. Our group have previously generated a mouse These findings suggested that functional restoration of hu-
model with hypomorphic Pkd2 allele (Pkd2nf3). Mice with man PC2 sufficiently delayed and arrested ADPKD progres-
Perspectives of Gene Therapies in Autosomal Dominant Polycystic Kidney Disease Current Gene Therapy, 2017, Vol. 17, No. 1 45

sion in animals. Moreover, the findings also laid on theoreti- which entire viral genome is deleted, has been developed.
cal basis for gene therapy in treatment of ADPKD [84]. This modified adenovirus exhibit very low immunogenicity
and large packaging capacity [98], but low production effi-
To achieve therapeutic effects for ADPKD, an efficient
ciency challenges its wide application in clinic [99].
gene delivery system is required for transferring the target
gene into the kidney. Currently often used delivery systems Lentivirus is another sort of well-characterized viral vec-
include naked plasmids, nonviral vectors and viral vectors tors with low immunogenicity (Table 2). Lentiviral vectors
(Table 1). As the first and simplest delivery system been injected through renal artery and vein only obtained gene
studied, naked plasmids are easily produced and safe. How- expression in inner medullary collecting ducts (IMCDs). If
ever, it is not only insufficient to pass through lipophilic cell being directly injected into renal parenchyma, transgene can
membrane, but also prone to have transient expression in be detected in the cortex of the kidney, but only along the
targeted cells [85]. Other concerns are that naked plasmids injection track [100]. Lentivirus-mediated transgene can be
are easily degraded by serum nucleases and liver metabolism integrated into the host genome, and persistently expressed
[86]. in the targeted cells and tissues [101]. As the flipside, this
genome integration may lead to leukemia-like mutations
Nonviral vectors (lipid based and polymeric vectors) which may activate or serve as some proto-oncogenes [102].
have been focused for gene delivery because of their safety
and low immunogenicity. The synthetic cationic lipid and Adeno-associated virus (AAV) can be found in normal
polymeric vectors, by which plasmid DNA was protected human populations and has endemic characteristics. It is
from circulating nucleases and was easily absorbed by cell therefore considered to be one of ideal vectors owing to its
membrane, were developed to improve plasmid-delivery non-pathogenicity [103], broad tropism for renal cells [104],
efficiency [87-89]. However, particles with cationic charge persistent transgene expression and rarely integrating into
host genome [105, 106]. Through renal artery injection of
may aggregated in physiological fluid, as a result of reduced
AAV, successful transgene expression was observed in tubu-
electrostatic repulsion or interaction with blood component
lar epithelial cells of the proximal tubule and intercalated
[90]. Aggregation during systemic administration may in-
cells of collecting duct [107]. A recent study also reported
duce rapid clearance by circulating macrophages and may
that renal vein injection of rAAV-9 provided high transduc-
lead to blood cell aggregation even embolism [91]. In addi- tion efficiency in both the medulla and cortex, even in
tion, vector transfer and expression efficiency did also not glomeruli, suggesting that AAV might be a choice in treat-
reach therapeutic expectations for renal epithelia associated ment of renal diseases [108].
diseases.
In comparison with gene deliveries through artery, vein
By decades of investigation, viral vectors were shown to or parenchyma injection, retrograde transureteral gene deliv-
have the highest efficiency for transferring specific genes to ery facilitates transgene to reach renal epithelia and mini-
mammalian cells compared to non-viral approaches. Cur- mizes immune responses and macrophage-mediated clear-
rently, often used viral vectors include adenovirus, lentivirus ance, thus may has more benefits for treating ADPKD.
and adeno-associated virus (AAVs) (Table 2). Adenoviral Through retrograde transureteral injection, successful trans-
vectors are non-integrating viruses that can infect both divid- gene expression was detected in medullary tubules of the
ing and non-dividing cells in wide spectrum of tissues, in- kidney by AAV-mediated gene transfer in mouse model
cluding the kidney [92, 93]. However, adenovirus mediated [109]. By injection of recombinant AAV2/8 and AAV2/9
gene transfer to tubular epithelia, glomeruli and interstitial with the same method, high gene transfer efficiency was
cells of the kidney has relatively poor efficiency and only obtained in tubular epithelia of the kidney [110]. Given that
keeps transgene expression for few weeks [94-96]. In rare viral vectors have shown high efficiency of gene transduc-
case, the adenovirus can cause severe side effects, even le- tion and elongated duration of transgene expression, viral
thal in some of recipients due to immunogenic response [97]. mediated gene transfer combined with retrograde transuret-
To bypass this difficulty, the next generation of adenoviral eral injection may provide promising therapeutic potential
vectors, named as helper-dependent or gutless adenovirus in for treatment of ADPKD.

Table 1. Advantage and disadvantage of different gene-delivery systems.

Delivery System Advantage Disadvantage

Naked plasmids Safe and Low immunogenicity Inefficient gene delivery


Easy to produce with strict quality control (Rapid clearance and difficult to enter cell)
Easy to store and transport

Nonviral vectors No size limitation for DNA Relative low transfer efficiency compared to viral vectors
Low immunogenicity Potential toxicities (aggregation in blood)
Protect DNA from degradation and facilitate cell absorption

Viral vectors High transfection efficiency in a spectrum of mammalian cells Limited size of recombinant DNA
Diverse serotype targeting different tissues Immunogeneity may lead to adverse effects or rapid clearance
Long period even persistent expression Potential oncogenicity
46 Current Gene Therapy, 2017, Vol. 17, No. 1 Xu et al.

Table 2. Compression of diverse viral-associated vectors for gene transfer.

Viral Vectors Advantage Disadvantage

Adenovirus Large transgene size (~8kb, up to ~36kb*) Immunogenicity and induction of inflammatory re-
No insertional mutagenesis sponses
Broad tropism to tissues and cells Transient expression

Lentivirus Large transgene size (~10kb) Insertional mutagenesis or potential proto-oncogene


Low immunogenicity activation
Persistent expression Relative low delivery efficiency to kidney

Adeno-associated virus High transfection efficiency in a spectrum of kidney cells Limited packaging capacity (~4.7kb)
(AAVs) Low immunogenicity and non-pathogenicity
Persistent expression
No insertional mutagenesis
*:gutless adenovirus.

PERSPECTIVES AND CONCLUSION routine screening and management. Nephrol Dial Transplant 2014;
29(2): 247-54.
Currently, there is a lack of suitable treatment in ADPKD [6] Kim Y, Bae SK, Cheng T, et al. Automated segmentation of liver
patients. Conventional surgical options for ADPKD patients and liver cysts from bounded abdominal MR images in patients
seem only to relieve the patient symptoms, while there is no with autosomal dominant polycystic kidney disease. Phys Med Biol
2016; 61(22): 7864-80.
evidence that it can delay disease progression and prevent
[7] Ring T, Spiegelhalter D. Risk of intracranial aneurysm bleeding in
onset of ESRD [111]. As the only curative treatment, kidney autosomal-dominant polycystic kidney disease. Kidney Int 2007;
transplantation has been proven to benefit ADPKD patients 72(11): 1400-2.
[112-116]. Lack of donors is a key issue to limit the applica- [8] Ong AC. Screening for intracranial aneurysms in ADPKD. BMJ
tion of kidney transplantation in the patients [117]. Drug 2009; 339(7723): b3763.
therapy of ADPKD has been highly expected, but several [9] Xu HW, Yu SQ, Mei CL, Li MH. Screening for intracranial aneu-
rysm in 355 patients with autosomal-dominant polycystic kidney
potential drugs did not achieve satisfying clinical effect. In
disease. Stroke 2011; 42(1): 204-6.
light of the inherited nature of ADPKD, gene therapy has [10] Rozenfeld MN, Ansari SA, Mohan P, Shaibani A, Russell EJ,
shown a great potential to treat this refractory disease. Our Hurley MC. Autosomal dominant polycystic kidney disease and in-
recent results of human PKD2 transgene [83] provide an tracranial aneurysms: is there an increased risk of treatment? Am J
encouraging evidence that the disease could be treated by Neuroradiol 2016; 37(2): 290-93.
appropriate gene delivery system. Further, study on novel [11] Liu D, Wang CJ, Judge DP, et al. A Pkd1-Fbn1 Genetic interaction
implicates tgf-beta signaling in the pathogenesis of vascular com-
therapeutic intervention will bring new hope and prospect in
plications in autosomal dominant polycystic kidney disease. J Am
the treatment of ADPKD. Soc Nephrol 2014; 25(1): 81-91.
[12] Perrone RD, Malek AM, Watnick T. Vascular complications in
CONFLICT OF INTEREST autosomal dominant polycystic kidney disease. Nat Rev Nephrol
2015; 11(10): 589-98.
The authors declare no conflict of interest, financial or [13] Neves JB, Rodrigues FB, Lopes JA. Autosomal dominant polycys-
otherwise. tic kidney disease and coronary artery dissection or aneurysm: a
systematic review. Ren Fail 2016; 38(4): 493-502.
ACKNOWLEDGEMENTS [14] Feng J, Ge S, Zhang L, Che H, Liang C. Aortic dissection is asso-
Declared none. ciated with reduced polycystin-1 expression, an abnormality that
leads to increased ERK phosphorylation in vascular smooth muscle
cells. Eur J Histochem 2016; 60(4): 2711.
REFERENCES [15] Dimarakis I, Kadir I. Acute aortic syndrome in autosomal dominant
[1] Harris PC, Torres VE. Genetic mechanisms and signaling pathways polycystic kidney disease. Kidney Int 2017; 91(2): 512.
in autosomal dominant polycystic kidney disease. J Clin Invest [16] Lucon M, Ianhez LE, Lucon AM, Chambo JL, Sabbaga E, Srougi
2014; 124(6): 2315-24. M. Bilateral nephrectomy of huge polycystic kidneys associated
[2] Ong AC, Devuyst O, Knebelmann B, Walz G. Autosomal domi- with a rectus abdominis diastasis and umbilical hernia. Clinics
nant polycystic kidney disease: the changing face of clinical 2006; 61(6): 529-34.
management. Lancet 2015; 385(9981): 1993-2002. [17] Mikolajczyk AE, Te HS, Chapman AB. Gastrointestinal manifesta-
[3] Willey CJ, Blais JD, Hall AK, Krasa HB, Makin AJ, Czerwiec FS. tions of autosomal-dominant polycystic kidney disease. Clin Gas-
Prevalence of autosomal dominant polycystic kidney disease in the troenterol Hepatol 2017; 15(1): 17-24.
European Union. Nephrol Dial Transplant 2016; pii: gfw240. [18] The European Polycystic Kidney Disease Consortium. The poly-
[Epub ahead of print] cystic kidney disease 1 gene encodes a 14 kb transcript and lies
[4] Yazdanpanah K, Manouchehri N, Hosseinzadeh E, Emami MH, within a duplicated region on chromosome 16. Cell 1994; 78(4):
Karami M, Sarrami AH. Recurrent acute pancreatitis and cholangi- 725.
tis in a patient with autosomal dominant polycystic kidney disease. [19] Loftus BJ, Kim UJ, Sneddon VP, et al. Genome duplications and
Int J Prev Med 2013; 4(2): 233-6. other features in 12 Mb of DNA sequence from human chromo-
[5] Luciano RL, Dahl NK. Extra-renal manifestations of autosomal some 16p and 16q. Genomics 1999; 60(3): 295-308.
dominant polycystic kidney disease (ADPKD): considerations for
Perspectives of Gene Therapies in Autosomal Dominant Polycystic Kidney Disease Current Gene Therapy, 2017, Vol. 17, No. 1 47

[20] Mochizuki T, Wu G, Hayashi T, et al. PKD2, a gene for polycystic [44] Qian F, Germino FJ, Cai Y, Zhang X, Somlo S, Germino GG.
kidney disease that encodes an integral membrane protein. Science PKD1 interacts with PKD2 through a probable coiled-coil domain.
1996; 272(5266): 1339-42. Nat Genet 1997; 16(2): 179-83.
[21] Rossetti S, Consugar MB, Chapman AB, et al. Comprehensive [45] Hanaoka K, Qian F, Boletta A, et al. Co-assembly of polycystin-1
molecular diagnostics in autosomal dominant polycystic kidney and -2 produces unique cation-permeable currents. Nature 2000;
disease. J Am Soc Nephrol 2007; 18(7): 2143-60. 408(6815): 990-4.
[22] Igarashi P, Somlo S. Polycystic kidney disease. J Am Soc Nephrol [46] Yang Y, Ehrlich BE. Structural studies of the C-terminal tail of
2007; 18(5): 1371-3. polycystin-2 (PC2) reveal insights into the mechanisms used for
[23] Fedeles SV, Gallagher AR, Somlo S. Polycystin-1: a master regula- the functional regulation of the PC2. J Physiol 2016; 594(15):
tor of intersecting cystic pathways. Trends Mol Med 2014; 20(5): 4141-49.
251-60. [47] Kim S, Nie H, Nesin V, et al. The polycystin complex mediates
[24] Barua M, Cil O, Paterson AD, et al. Family history of renal disease Wnt/Ca(2+) signalling. Nat Cell Biol 2016; 18(7): 752-64.
severity predicts the mutated gene in ADPKD. J Am Soc Nephrol [48] Cornec-Le Gall E, Audrezet MP, Chen JM, et al. Type of PKD1
2009; 20(8): 1833-8. mutation influences renal outcome in ADPKD. J Am Soc Nephrol
[25] Liu B, Chen SC, Yang YM, et al. Identification of novel PKD1 and 2013; 24(6): 1006-13.
PKD2 mutations in a Chinese population with autosomal dominant [49] ADPKD. Autosomal Dominant Polycystic Kidney Disease: Muta-
polycystic kidney disease. Sci Rep 2015; 5: 17468-79. tion Database. PKD Foundation Web site. http://pkdb.mayo.edu
[26] Jin M, Xie Y, Chen Z, et al. System analysis of gene mutations and 2016.
clinical phenotype in Chinese patients with autosomal-dominant [50] Harris PC, Hopp K. The mutation, a key determinant of phenotype
polycystic kidney disease. Sci Rep 2016; 6: 35945. in ADPKD. J Am Soc Nephrol 2013; 24(6): 868-70.
[27] Porath B, Gainullin VG, Cornec-Le Gall E, et al. Mutations in [51] Lu W, Peissel B, Babakhanlou H, et al. Perinatal lethality with
GANAB, encoding the glucosidase IIalpha subunit, cause autoso- kidney and pancreas defects in mice with a targetted Pkd1 muta-
mal-dominant polycystic kidney and liver disease. Am J Hum tion. Nat Genet 1997; 17(2): 179-81.
Genet 2016; 98(6): 1193-207. [52] Brill SR, Ross KE, Davidow CJ, Ye M, Grantham JJ, Caplan MJ.
[28] Hughes J, Ward CJ, Peral B, et al. The polycystic kidney disease 1 Immunolocalization of ion transport proteins in human autosomal
(PKD1) gene encodes a novel protein with multiple cell recognition dominant polycystic kidney epithelial cells. Proc Natl Acad Sci
domains. Nat Genet 1995; 10(2): 151-60. USA 1996; 93(19): 10206-11.
[29] Nims N, Vassmer D, Maser RL. Transmembrane domain analysis [53] Wu G, D'Agati V, Cai Y, et al. Somatic inactivation of Pkd2 results
of polycystin-1, the product of the polycystic kidney disease-1 in polycystic kidney disease. Cell 1998; 93(2): 177-88.
(PKD1) gene: evidence for 11 membrane-spanning domains. Bio- [54] Wu G, Markowitz GS, Li L, et al. Cardiac defects and renal failure
chemistry 2003; 42(44): 13035-48. in mice with targeted mutations in Pkd2. Nat Genet 2000; 24(1):
[30] Geng L, Burrow CR, Li HP, Wilson PD. Modification of the com- 75-8.
position of polycystin-1 multiprotein complexes by calcium and ty- [55] Li A, Fan S, Xu Y, et al. Rapamycin treatment dose-dependently
rosine phosphorylation. Biochim Biophys Acta 2000; 1535(1): 21- improves the cystic kidney in a new ADPKD mouse model via the
35. mTORC1 and cell-cycle-associated CDK1/cyclin axis. J Cell Mol
[31] Nauli SM, Alenghat FJ, Luo Y, et al. Polycystins 1 and 2 mediate Med 2017; doi: 10.1111/jcmm.13091. [Epub ahead of print]
mechanosensation in the primary cilium of kidney cells. Nat Genet [56] Qian F, Watnick TJ, Onuchic LF, Germino GG. The molecular
2003; 33(2): 129-37. basis of focal cyst formation in human autosomal dominant poly-
[32] Chapin HC, Caplan MJ. The cell biology of polycystic kidney cystic kidney disease type I. Cell 1996; 87(6): 979-87.
disease. J Cell Biol 2010; 191(4): 701-10. [57] Watnick TJ, Torres VE, Gandolph MA, et al. Somatic mutation in
[33] Hildebrandt F, Benzing T, Katsanis N. Ciliopathies. N Engl J Med individual liver cysts supports a two-hit model of cystogenesis in
2011; 364(16): 1533-43. autosomal dominant polycystic kidney disease. Mol Cell 1998;
[34] Cai Y, Fedeles SV, Dong K, et al. Altered trafficking and stability 2(2): 247-51.
of polycystins underlie polycystic kidney disease. J Clin Invest [58] Kim I, Li C, Liang D, et al. Polycystin-2 expression is regulated by
2014; 124(12): 5129-44. a PC2-binding domain in the intracellular portion of fibrocystin. J
[35] Gainullin VG, Hopp K, Ward CJ, Hommerding CJ, Harris PC. Biol Chem 2008; 283(46): 31559-66.
Polycystin-1 maturation requires polycystin-2 in a dose-dependent [59] Lantinga-van Leeuwen IS, Dauwerse JG, Baelde HJ. Lowering of
manner. J Clin Invest 2015; 125(2): 607-20. Pkd1 expression is sufficient to cause polycystic kidney disease.
[36] Delling M, Indzhykulian AA, Liu X, et al. Primary cilia are not Hum Mol Genet 2004; 13(24): 3069-77.
calcium-responsive mechanosensors. Nature 2016; 531(7596): 656- [60] Rossetti S, Hopp K, Sikkink RA, et al. Identification of gene muta-
60. tions in autosomal dominant polycystic kidney disease through tar-
[37] Celic A, Petri ET, Demeler B, Ehrlich BE, Boggon TJ. Domain geted resequencing. J Am Soc Nephrol 2012; 23(5): 915-33.
mapping of the polycystin-2 C-terminal tail using de novo molecu- [61] Rossetti S, Kubly VJ, Consugar MB, et al. Incompletely penetrant
lar modeling and biophysical analysis. J Biol Chem 2008; 283(42): PKD1 alleles suggest a role for gene dosage in cyst initiation in
28305-12. polycystic kidney disease. Kidney Int 2009; 75(8): 848-55.
[38] Shen PS, Yang X, DeCaen PG, et al. The structure of the polycys- [62] Vujic M, Heyer CM, Ars E, et al. Incompletely penetrant PKD1
tic kidney disease channel PKD2 in lipid nanodiscs. Cell 2016; alleles mimic the renal manifestations of ARPKD. J Am Soc Neph-
167(3): 763-73 e11. rol 2010; 21(7): 1097-102.
[39] Montell C, Birnbaumer L, Flockerzi V. The TRP channels, a re- [63] Hopp K, Ward CJ, Hommerding CJ, et al. Functional polycystin-1
markably functional family. Cell 2002; 108(5): 595-98. dosage governs autosomal dominant polycystic kidney disease se-
[40] Qamar S, Vadivelu M, Sandford R. TRP channels and kidney dis- verity. J Clin Invest 2012; 122(11): 4257-73.
ease: lessons from polycystic kidney disease. Biochem Soc Trans [64] Bastos AP, Piontek K, Silva AM, et al. Pkd1 haploinsufficiency
2007; 35(1): 124-28. increases renal damage and induces microcyst formation following
[41] Grieben M, Pike AC, Shintre CA, et al. Structure of the polycystic ischemia/reperfusion. J Am Soc Nephrol 2009; 20(11): 2389-402.
kidney disease TRP channel Polycystin-2 (PC2). Nat Struct Mol [65] Patel V, Li L, Cobo-Stark P, et al. Acute kidney injury and aberrant
Biol 2017; 24(2): 114-22. planar cell polarity induce cyst formation in mice lacking renal
[42] Koulen P, Cai Y, Geng L, et al. Polycystin-2 is an intracellular cilia. Hum Mol Genet 2008; 17(11): 1578-90.
calcium release channel. Nat Cell Biol 2002; 4(3): 191-97. [66] Takakura A, Contrino L, Zhou X, et al. Renal injury is a third hit
[43] Geng L, Okuhara D, Yu Z, et al. Polycystin-2 traffics to cilia inde- promoting rapid development of adult polycystic kidney disease.
pendently of polycystin-1 by using an N-terminal RVxP motif. J Hum Mol Genet 2009; 18(14): 2523-31.
Cell Sci 2006; 119(Pt 7): 1383-95.
48 Current Gene Therapy, 2017, Vol. 17, No. 1 Xu et al.

[67] Kurbegovic A, Trudel M. Acute kidney injury induces hallmarks of [91] Zhang Y, Satterlee A, Huang L. In vivo gene delivery by nonviral
polycystic kidney disease. Am J Physiol Renal Physiol 2016; vectors: overcoming hurdles? Mol Ther 2012; 20(7): 1298-304.
311(4): F740-F51. [92] Sandovici M, Deelman LE, Benigni A, Henning RH. Towards
[68] Happé H, Leonhard WN, van der Wal A et al. Toxic tubular injury graft-specific immune suppression: Gene therapy of the trans-
in kidneys from Pkd1-deletion mice accelerates cystogenesis ac- planted kidney. Adv Drug Deliv Rev 2010; 62(14): 1358-68.
companied by dysregulated planar cell polarity and canonical Wnt [93] Isaka Y. Gene therapy targeting kidney diseases: routes and vehi-
signaling pathways. Hum Mol Genet 2009; 18: 2532-42. cles. Clin Exp Nephrol 2006; 10(4): 229-35.
[69] Bell PD, Fitzgibbon W, Sas K, et al. Loss of primary cilia upregu- [94] Moullier P, Friedlander G, Calise D, Ronco P, Perricaudet M, Ferry
lates renal hypertrophic signaling and promotes cystogenesis. J Am N. Adenoviral-mediated gene transfer to renal tubular cells in vivo.
Soc Nephrol 2011; 22(5): 839-48. Kidney Int 1994; 45(4): 1220-5.
[70] Grantham JJ, Torres VE. The importance of total kidney volume in [95] Zhu G, Nicolson AG, Cowley BD, Rosen S, Sukhatme VP. In vivo
evaluating progression of polycystic kidney disease. Nat Rev adenovirus-mediated gene transfer into normal and cystic rat kid-
Nephrol 2016; 12(11): 667-77. neys. Gene Ther 1996; 3(4): 298-304.
[71] Torres VE. Vasopressin antagonists in polycystic kidney disease. [96] Heikkila P, Parpala T, Lukkarinen O, Weber M, Tryggvason K.
Kidney Int 2005; 68(5): 2405-18. Adenovirus-mediated gene transfer into kidney glomeruli using an
[72] Barash I, Ponda MP, Goldfarb DS, Skolnik EY. A pilot clinical study ex vivo and in vivo kidney perfusion system - first steps towards
to evaluate changes in urine osmolality and urine cAMP in response gene therapy of Alport syndrome. Gene Ther 1996; 3(1): 21-7.
to acute and chronic water loading in autosomal dominant polycystic [97] Raper SE, Chirmule N, Lee FS, et al. Fatal systemic inflammatory
kidney disease. Clin J Am Soc Nephrol 2010; 5(4): 693-7. response syndrome in a ornithine transcarbamylase deficient pa-
[73] Yamaguchi T, Reif GA, Calvet JP, Wallace DP. Sorafenib inhibits tient following adenoviral gene transfer. Mol Genet Metab 2003;
cAMP-dependent ERK activation, cell proliferation, and in vitro 80(1-2): 148-58.
cyst growth of human ADPKD cyst epithelial cells. Am J Physiol [98] Cots D, Bosch A, Chillon M. Helper dependent adenovirus vectors:
Renal Physiol 2010; 299(5): F944-51. progress and future prospects. Curr Gene Ther 2013; 13(5): 370-81.
[74] Xia S, Li X, Johnson T, Seidel C, Wallace DP, Li R. Polycystin- [99] Alba R, Bosch A, Chillon M. Gutless adenovirus: last-generation
dependent fluid flow sensing targets histone deacetylase 5 to pre- adenovirus for gene therapy. Gene Ther 2005; 12 (Suppl 1): S18-
vent the development of renal cysts. Development 2010; 137(7): 27.
1075-84. [100] Gusella GL, Fedorova E, Hanss B, Marras D, Klotman ME, Klot-
[75] Serra AL, Poster D, Kistler AD, et al. Sirolimus and kidney growth man PE. Lentiviral gene transduction of kidney. Hum Gene Ther
in autosomal dominant polycystic kidney disease. N Engl J Med 2002; 13(3): 407-14.
2010; 363(9): 820-9. [101] Park F, Gow KW. Gene therapy: future or flop. Pediatr Clin North
[76] Walz G, Budde K, Mannaa M, et al. Everolimus in patients with Am 2006; 53(4): 621-38.
autosomal dominant polycystic kidney disease. N Engl J Med [102] Hacein-Bey-Abina S, Von Kalle C, Schmidt M, et al. LMO2-
2010; 363(9): 830-40. associated clonal T cell proliferation in two patients after gene
[77] Calvet JP. MEK inhibition holds promise for polycystic kidney therapy for SCID-X1. Science 2003; 302(5644): 415-9.
disease. J Am Soc Nephrol 2006; 17(6): 1498-500. [103] Amado D, Mingozzi F, Hui D, et al. Safety and efficacy of subreti-
[78] Perico N, Antiga L, Caroli A, et al. Sirolimus therapy to halt the nal readministration of a viral vector in large animals to treat con-
progression of ADPKD. J Am Soc Nephrol 2010; 21(6): 1031-40. genital blindness. Sci Transl Med 2010; 2(21): 21ra16.
[79] Torres VE, Chapman AB, Devuyst O, et al. Tolvaptan in patients [104] Lipkowitz MS, Hanss B, Tulchin N, et al. Transduction of renal
with autosomal dominant polycystic kidney disease. N Engl J Med cells in vitro and in vivo by adeno-associated virus gene therapy
2012; 367(25): 2407-18. vectors. J Am Soc Nephrol 1999; 10(9): 1908-15.
[80] Caroli A, Perico N, Perna A, et al. Effect of longacting soma- [105] Blessing D, Deglon N. Adeno-associated virus and lentivirus vec-
tostatin analogue on kidney and cyst growth in autosomal dominant tors: a refined toolkit for the central nervous system. Curr Opin Vi-
polycystic kidney disease (ALADIN): a randomised, placebo- rol 2016; 21: 61-6.
controlled, multicentre trial. Lancet 2013; 382(9903): 1485-95. [106] Janssen MJ, Arcolino FO, Schoor P, Kok RJ, Mastrobattista E.
[81] Blair HA, Keating GM. Tolvaptan: A review in autosomal domi- Gene based therapies for kidney regeneration. Eur J Pharmacol
nant polycystic kidney disease. Drugs 2015; 75(15): 1797-806. 2016; 790: 99-108.
[82] Horie S. Will introduction of tolvaptan change clinical practice in [107] Chen S, Agarwal A, Glushakova OY, et al. Gene delivery in renal
autosomal dominant polycystic kidney disease? Kidney Int 2015; tubular epithelial cells using recombinant adeno-associated viral
88(1): 14-6. vectors. J Am Soc Nephrol 2003; 14(4): 947-58.
[83] Li A, Tian X, Zhang X, et al. Human polycystin-2 transgene dose- [108] Rocca CJ, Ur SN, Harrison F, Cherqui S. rAAV9 combined with
dependently rescues adpkd phenotypes in pkd2 mutant mice. Am J renal vein injection is optimal for kidney-targeted gene delivery:
Pathol 2015; 185(10): 2843-60. conclusion of a comparative study. Gene Ther 2014; 21(6): 618-
[84] Kim I, Ding T, Fu Y, et al. Conditional mutation of Pkd2 causes 28.
cystogenesis and upregulates beta-catenin. J Am Soc Nephrol [109] Ito K, Chen J, Khodadadian JJ, et al. Adeno-associated viral vector
2009; 20(12): 2556-69. transduction of green fluorescent protein in kidney: effect of unilat-
[85] Schmidt-Wolf GD, Schmidt-Wolf IG. Non-viral and hybrid vectors eral ureteric obstruction. BJU Int 2008; 101(3): 376-81.
in human gene therapy: an update. Trends Mol Med 2003; 9(2): 67- [110] Chung DC, Fogelgren B, Park KM, et al. Adeno-associated virus-
72. mediated gene transfer to renal tubule cells via a retrograde ureteral
[86] Liu F, Shollenberger LM, Conwell CC, Yuan X, Huang L. Mecha- approach. Nephron Extra 2011; 1(1): 217-23.
nism of naked DNA clearance after intravenous injection. J Gene [111] Millar M, Tanagho YS, Haseebuddin M, Clayman RV, Bhayani
Med 2007; 9(7): 613-9. SB, Figenshau RS. Surgical cyst decortication in autosomal domi-
[87] He D, Wagner E. Defined polymeric materials for gene delivery. nant polycystic kidney disease. J Endourol 2013; 27(5): 528-34.
Macromol Biosci 2015; 15(5): 600-12. [112] Ahmad SB, Inouye B, Phelan MS, et al. Live donor renal transplant
[88] Allen TM, Cullis PR. Liposomal drug delivery systems: from con- with simultaneous bilateral nephrectomy for autosomal dominant
cept to clinical applications. Adv Drug Deliv Rev 2013; 65(1): 36- polycystic kidney disease is feasible and satisfactory at long-term
48. follow-up. Transplantation 2016; 100(2): 407-15.
[89] Yin H, Kanasty RL, Eltoukhy AA, Vegas AJ, Dorkin JR, Anderson [113] Mehrabi A, Golriz M, Maier J, et al. Long-Term follow-up of
DG. Non-viral vectors for gene-based therapy. Nat Rev Genet kidney transplant recipients with polycystic kidney disease. Exp
2014; 15(8): 541-55. Clin Transplant 2015; 13(5): 413-20.
[90] Wiethoff CM, Middaugh CR. Barriers to nonviral gene delivery. J [114] Mosconi G, Persici E, Cuna V, et al. Renal transplant in patients
Pharm Sci 2003; 92(2): 203-17. with polycystic disease: the Italian experience. Transplant Proc
2013; 45(7): 2635-40.
Perspectives of Gene Therapies in Autosomal Dominant Polycystic Kidney Disease Current Gene Therapy, 2017, Vol. 17, No. 1 49

[115] Martinez V, Comas J, Arcos E, et al. Renal replacement therapy in cystic kidney disease in Denmark. Clin J Am Soc Nephrol 2010;
ADPKD patients: a 25-year survey based on the Catalan registry. 5(11): 2034-9.
BMC Nephrol 2013; 14: 186. [117] Chapman AB, Devuyst O, Eckardt KU, et al. Autosomal-dominant
[116] Orskov B, Romming Sorensen V, Feldt-Rasmussen B, Strandgaard polycystic kidney disease (ADPKD): executive summary from a
S. Improved prognosis in patients with autosomal dominant poly- Kidney Disease: Improving Global Outcomes (KDIGO) Controver-
sies Conference. Kidney Int 2015; 88(1): 17-27.

Das könnte Ihnen auch gefallen