1,461
Views
6
CrossRef citations to date
0
Altmetric
Review Article

Nano-sized carriers in gene therapy for renal fibrosis in vivo

, , , &
Article: 1331099 | Received 28 Jan 2017, Accepted 12 May 2017, Published online: 13 Jun 2017

ABSTRACT

Renal fibrosis is the final common pathway leading to end-stage renal failure regardless of underlying initial nephropathies. No specific therapy has been established for renal fibrosis. Gene therapy is a promising strategy for the treatment of renal fibrosis. Nano-sized carriers including viral vectors and non-viral vectors have been shown to enhance the delivery and treatment effects of gene therapy for renal fibrosis in vivo. This review focuses on the mechanisms of renal fibrosis and the in vivo technologies and methodologies of nano-sized carriers in gene therapy for renal fibrosis.

RESPONSIBLE EDITOR Alexander Seifalian Director of Nanotechnology & Regenerative Medicine Ltd., The London BioScience Innovation Centre, London, UNITED KINGDOM

1. Introduction

Chronic kidney disease (CKD), defined as a decreasing glomerular filtration rate and/or the presence of histological or biochemical markers of kidney damage over 3 months, is a worldwide public health problem [Citation1]. A recent meta-analysis reported that the global prevalence of CKD is estimated as 13.4% in general populations [Citation2]. In the pathology of CKD, tubulointerstitial fibrosis, called renal fibrosis, is the final common pathway leading to end-stage renal disease regardless of initial nephropathies [Citation3,Citation4]. As no specific therapy has been established for renal fibrosis, the development of treatment options is crucial to improve the prognosis for CKD patients. Understanding the mechanisms of renal fibrosis enables targeting of the processes that take place to prevent them from occurring. Many different kinds of cells, such as immune, vascular endothelial, tubular epithelial, and fibroblast cells, have been considered to contribute to renal fibrosis in vivo [Citation5Citation10]. Studying therapeutic approaches for the treatment of renal fibrosis in vivo is important. Gene therapy is a potentially promising strategy for the treatment of renal fibrosis in vivo because it can target molecules that were previously difficult to set as therapeutic targets using small molecules or antibodies. The development of nano-sized carriers including viral vectors and non-viral vectors has been shown to enhance the delivery and treatment effects of gene therapy for various diseases, including renal fibrosis in vivo [Citation11Citation14].

This review focuses on the mechanisms of renal fibrosis and the in vivo technologies and methodologies of nano-sized carriers in gene therapy for renal fibrosis.

2. Mechanisms of renal fibrosis

Renal fibrosis is pathologically characterized by the proliferation of myofibroblasts and the excessive accumulation of extracellular matrix components such as fibrotic collagen in the tubulointerstitial space [Citation15]. Although the precise mechanisms of renal fibrosis have not been completely determined, activation of pro-fibrotic signaling pathways and chronic inflammation are thought to play central roles in renal fibrosis () [Citation16Citation25].

Figure 1. Mechanisms of renal fibrosis.

TGF: transforming growth factor; Smads: small mothers against decapentaplegic; P: phosphorylation.

Figure 1. Mechanisms of renal fibrosis.TGF: transforming growth factor; Smads: small mothers against decapentaplegic; P: phosphorylation.

2.1. Pro-fibrotic signaling

Numerous studies have demonstrated that transforming growth factor (TGF)-β1 and downstream intracellular proteins, known as small mothers against decapentaplegic (Smad), play a central role as a pro-fibrotic pathway in renal fibrosis () [Citation16Citation22]. TGF-β1 receptor-activated Smad3 combines with a common Smad, called Smad4, and this complex is translocated into the nucleus where it binds to DNA elements to promote the transcription of various pro-fibrotic genes () [Citation16Citation19]. Therapeutic agents that inhibit TGF-β1–Smad signaling have been shown to reduce extracellular matrix accumulation in the tubulointerstitial space, resulting in the inhibition of renal fibrosis [Citation16,Citation26,Citation27].

2.2. Chronic inflammation

Unresolved inflammation characterized by leukocyte infiltration is considered to be a main factor contributing to renal fibrosis () [Citation23Citation25,Citation28,Citation29]. Infiltrated leukocytes excrete various pro-fibrotic growth factors and cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β, and IL-6, which promote the proliferation of myofibroblasts () [Citation29,Citation30]. Levels of these cytokines in plasma have been shown to increase in patients with CKD [Citation31,Citation32]. These cytokines activate nuclear factor-kappa B (NF-κB), which is a major signaling pathway that activates and promotes macrophage infiltration, further increasing the inflammatory pathway () [Citation33,Citation34]. Inhibition of the NF-κB signaling pathway was shown to inhibit renal injury, whereas activation of this pathway was shown to promote renal injury, including renal fibrosis [Citation33,Citation34].

3. Viral and non-viral vectors for renal fibrosis in vivo

Viral vectors and non-viral vectors have been studied as nano-sized carriers in gene therapy for renal fibrosis in vivo (). These vectors enhance delivery and increase the efficiency of gene transduction into the tubulointerstitial space and enhance the anti-fibrotic effects of gene therapy in vivo. The categories of vectors, genes, administration routes, and the effects of renal fibrosis in vivo are summarized in .

Table 1. Gene therapies using viral and non-viral vectors for the treatment of renal fibrosis.

Figure 2. Viral and non-viral vectors for renal fibrosis in vivo.

TNF: tumor necrosis factor; IL: interleukin; NF-κB: nuclear factor-kappa B.

Figure 2. Viral and non-viral vectors for renal fibrosis in vivo.TNF: tumor necrosis factor; IL: interleukin; NF-κB: nuclear factor-kappa B.

3.1. Viral vectors

Adenovirus and adenovirus-associated virus (AAV) vectors are the most widely used viral vectors in gene therapy for renal fibrosis in vivo, while only a few studies have reported on the role of lentiviral vectors. Many studies have targeted inhibition of the TGF-β1–Smad signaling pathway for the treatment of renal fibrosis. Various administration routes (intramuscle, intravenous, intra-arterial, intraperitoneal, intrarenal, intraureter) have been used for the delivery of genes to the tubulointerstitial space of the kidney.

3.1.1. Adenoviral and AAV vectors

Adenoviral vectors can deliver and transfect genes into both dividing and non-dividing cells [Citation55]. Adenoviral vectors are double-stranded, non-enveloped DNA viral vectors, 70–90 nm in diameter, with a genome of 36–38 kb [Citation55,Citation56]. Adenoviral vectors designed for expression of TGF-β1 type II receptor, which is a competitive inhibitor of TGF-β1, were injected into hindlimb muscles of a mouse model of diabetic nephropathy [Citation35]. Five weeks after administration, this gene therapy appeared to reduce fibrosis in both glomeruli and tubulointerstitial spaces [Citation35]. Another study reported that adenoviral vectors designed for expression of runt-related transcription factor 2 attenuated TGF-β1-induced Smad3 phosphorylation, and reduced expression levels of α-smooth muscle actin (α-SMA) and collagen I in the kidney of unilateral ureteral obstruction (UUO) mice produced by unilateral ureteral ligation, which is a representative animal model of renal fibrosis [Citation36,Citation57]. The administration of adenoviral vectors designed for expression of decorin, which is an inhibitor of TGF-β1, was found to inhibit expression levels of TGF-β1 mRNA and protein and reduce fibrosis in the kidney of a rat model of diabetic nephropathy [Citation37]. A study has reported on gene therapy that combined adenoviral vectors delivery and in vivo electroporation to deliver genes to the kidney [Citation38]. In that study, adenoviral vectors designed for expression of Smad7, which is an antagonist of TGF-β1–Smad signaling, were injected into the pelvic space of a rat model of renal fibrosis induced by UUO [Citation38]. Then, electroporation was performed on the kidneys. Ten electric pulses (50 m/s duration at 100 V) at a rate of 1 pulse/s were administrated to the kidney by a pair of electrode disks rigged on the tips of tweezers [Citation38]. That delivery system significantly over-expressed Smad7 in the kidney and inhibited renal fibrosis [Citation38]. Delivery of genes with adenoviral vectors to the kidney to modulate inflammatory signals and cytokines has been reported to inhibit renal fibrosis in vivo [Citation39Citation42]. Adenoviral vectors designed for expression of I-kappa-B-alpha (IκB-α), which is an inhibitor of NF-κB, were found to inhibit NF-κB activation by over-expression of IκB-α in the renal cortex and ameliorate tubulointerstitial injury characterized by fibrosis and infiltration of mononuclear cells in a rat model of renal fibrosis [Citation39]. Although adenoviral vectors have shown successful gene delivery to the kidney for the treatment of renal fibrosis, there is room for improvement as a carrier of gene therapy. The exogenous genes delivered using adenoviral vectors do not integrate into the host genome and do not replicate during cell division, resulting in short periods of gene expression of the targeted cells [Citation55]. To overcome this drawback of the adenoviral vector, AAV vectors have been developed [Citation58]. AAV vectors are single-stranded, non-enveloped DNA viral vectors, 18–26 nm in diameter, with a genome of 4–5 kb [Citation56,Citation58,Citation59]. AAV vectors can also transfect genes into both dividing and non-dividing cells, and may incorporate genes into the host genome. Therefore, the duration of gene expression delivered using AAV vectors is considered to be longer compared with that delivered by adenoviral vectors [Citation58]. AAV vectors were shown to have a low immunogenicity compared with adenoviral vectors [Citation58]. AAV vectors serotype 9, designed for expression of hepatocyte growth factor (HGF), which is an anti-fibrotic cytokine, showed remarkable attenuation of tubulointerstitial fibrosis in a mouse model of renal fibrosis [Citation40]. The administration of an AAV vector designed for expression of IL-10, which is an anti-inflammatory cytokine, was shown to over-express IL-10 in plasma and inhibit renal fibrosis by inhibiting infiltration of T lymphocytes and macrophages in a rat model of renal fibrosis [Citation41]. AAV vectors designed for expression of suppressor of cytokine signaling (SOCS)2 over-expressed SOCS2 in the kidney and inhibited renal fibrosis and inflammation in a rat model of diabetic nephropathy [Citation42]. The inhibitory effects on renal fibrosis by modulation of expression of the following genes by exogenous genes delivered with AAV vectors have been reported: adrenomedullin [Citation43]; endothelial nitric oxide synthase [Citation44]; Klotho [Citation45]; cytochrome P450 2J2 [Citation46]; and angiotensin II receptor 1 [Citation47].

3.1.2. Lentiviral vectors

Lentiviral vectors can transfect genes into both dividing cells and non-dividing cells and may incorporate genes into the host genome [Citation60]. The lentiviral vectors are enveloped, single-stranded RNA viral vectors, 80–130 nm in diameter, with a genome of 8–9 kb [Citation60]. Since lentiviral vectors are a recent development following on from adenoviral and AAV vectors, few studies have reported on lentiviral vectors as carriers of gene therapy for renal fibrosis in vivo. However, increasing interest in lentiviral vectors indicates the rise of a new field of research in lentiviral vectors for gene therapy of renal fibrosis [Citation48,Citation61]. Lentiviral vectors designed for expression of prohibitin, which is a pleiotropic protein for cellular proliferation, apoptosis, transcription, and mitochondria protein folding, were shown to ameliorate renal fibrosis in a mouse model of renal fibrosis induced by UUO [Citation48]. HIV-derived lentiviral vectors designed for expression of type II TGF-β receptor (TBRII) were reported to attenuate renal fibrosis better, as estimated by expression levels of extracellular matrix synthesis, such as fibronectin and collagen III, and expression of α-SMA in both cultured renal epithelial cells and renal fibroblasts compared with non-lentiviral constructs [Citation61]. However, the treatment effects on renal fibrosis using that lentiviral vector designed for the expression of type TBRII have not been investigated in vivo.

3.2. Non-viral vectors

There are several non-viral vectors that can deliver genes to the kidney (). Polyethylenimine (PEI) nanoparticles, liposome nanoparticles, and cationic gelatin nanoparticles have been demonstrated to show effective delivery of genes to the kidney and have promising treatment effects in animal models of renal fibrosis [Citation49Citation54].

3.2.1. PEI nanoparticles

PEI is a polymer, 50–100 nm in diameter, that is considered the preferable material for the preparation of non-viral vectors in terms of long-term safety and biocompatibility [Citation62,Citation63]. Small interfering RNA (siRNA) targeted paired box2 (PAX2) was reported to be delivered to the kidney with PEI nanoparticles via an intrarenal capsule injection in a mouse model of renal fibrosis induced by UUO [Citation49]. PAX2–siRNA–PEI nanoparticles inhibited PAX2 mRNA and PAX2 protein in the kidney, and ameliorated renal fibrosis [Citation49]. Delivery of microRNA-146a mimic with PEI nanoparticles was reported to over-express microRNA-146a in the fibrotic kidney induced by UUO, and inhibited renal fibrosis by inhibiting TGF-β1 and NF-κB signaling pathways in vivo [Citation49Citation54].

3.2.2. Liposome nanoparticles

Liposome nanoparticles, 100–150 nm in diameter, comprise phospholipids and cholesterol, which are the main components of the cell membrane, and therefore show high biocompatibility [Citation64]. Despite these advantages of liposome nanoparticles, few studies have reported on the validity of using liposome nanoparticles for gene delivery in the treatment of renal fibrosis in vivo. One study has reported that artificial viral envelope-type hemagglutinating virus of Japan (HVJ) liposome nanoparticles could deliver fluorescein isothiocyanate-labeled phosphorothioate non-targeted oligodeoxynucleotides to the nuclei of renal interstitial cells 10 min after transfection by retrograde injection through the ureter [Citation51]. However, treatment effects of genes delivered with HVJ liposome nanoparticles for renal fibrosis have not been investigated. These results suggest that liposome nanoparticles may quickly deliver genes to the kidney, and may be promising in gene therapy for renal fibrosis.

3.2.3. Gelatin nanoparticles

Gelatin is a protein derived from collagen [Citation65]. Cationic gelatin nanoparticles, 100–300 nm in diameter, which are produced by chemically introducing cations such as thylenediamine, putrescine, spermidine, or spermine to the carboxyl group of gelatin [Citation66], have been used in gene therapy for renal fibrosis in vivo [Citation52Citation54]. Plasmid DNA designed for the expression of TGF-β receptor (TBR) siRNA with cationized gelatin nanoparticles was administrated in the fibrotic kidney induced by UUO [Citation52]. These nanoparticles were shown to inhibit TBR expression and ameliorated fibrotic changes in the fibrotic kidney compared with naked plasmid DNA designed for the expression of TBR siRNA injection [Citation52]. Heat shock protein 47 (HSP47) siRNA with cationized gelatin nanoparticles was shown to knock down HSP47 expression and diminish renal fibrosis in a mouse model of renal fibrosis induced by UUO [Citation53]. Cationic gelatin nanoparticles incorporating plasmid DNA expressing matrix metalloprotease was shown to prevent renal fibrosis in a mouse model of diabetic nephropathy produced by intraperitoneal injection of streptozotocin, which causes damage to the pancreas and results in diabetic nephropathy [Citation54,Citation67].

4. Other methods of gene therapy for renal fibrosis in vivo

Electroporation and ultrasound methods are reported to deliver genes effectively to the kidney, and treatment effects have been demonstrated in renal fibrosis in vivo [Citation68,Citation69].

4.1. Electroporation

Electroporation is a transfection technique in which electric pulse waves are applied to target cells, creating micro-holes in the cell membrane to allow for the passage of exogenous genes into the cell. Plasmid DNA expressing HGF was injected into the tibialis anterior muscles and six electric pulses at 100 V were delivered by a stainless steel tweezer electrode placed in a transverse orientation relative to the muscle fiber. After transfection, plasma HGF levels increased and renal fibrosis was inhibited in a rat model of the fibrotic kidney induced by 5/6 nephrectomy [Citation68].

4.2. Ultrasound

Plasmid DNA expressing short hairpin RNA (shRNA) of connective tissue growth factor (CTGF) was loaded on to the surface of a cationic microbubble [Citation69]. The plasmid-carrying microbubbles were then administrated intravenously to mice and ultrasound was applied locally to the kidney treated with ureteral obstruction. This method exhibited reduced mRNA and protein levels of CTGF and inhibited fibrotic changes in the fibrotic kidney induced by UUO [Citation69]. Another study reported that delivery of plasmid DNA expressing shRNA of microRNA-433 to the kidney by ultrasound microbubble-mediated gene transfer suppressed the induction and progression of renal fibrosis in the fibrotic kidney induced by UUO [Citation15].

5. Summary

Nano-sized carriers such as viral and non-viral vectors in gene therapy for renal fibrosis in vivo have been developed. However, their long-term efficacy, side effects including toxicity and unexpected genomic DNA alternations, and their effects on other organs have not been fully investigated and require further study. Studies are also needed to develop nano-sized carriers that can exclusively deliver genes to the kidney.

Authors’ contributions

Haruhisa Miyazawa drafted the manuscript. Keiji Hirai, Susumu Ookawara, and Kenichi Ishibashi supervised the writing of the manuscript. Yoshiyuki Morishita conceived the content of the manuscript and supervised the writing of the manuscript. All authors contributed to the preparation of the manuscript and agree to be accountable for all aspects of the work.

Acknowledgments

The authors thank the members of the Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan.

Disclosure statement

No potential conflict of interest was reported by the authors.

Additional information

Funding

This work was partially supported by JSPS KAKENHI [grant number 17K09708].

Notes on contributors

Haruhisa Miyazawa

Haruhisa Miyazawa is an assistant professor of the Division of Nephrology, Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University. His research area is nephrology and dialysis.

Keiji Hirai

Keiji Hirai is an assistant professor of the Division of Nephrology, Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University. His research area is nephrology and dialysis.

Susumu Ookawara

Susumu Ookawara is an associate professor of the Division of Nephrology, Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University. His research area is nephrology and dialysis.

Kenichi Ishibashi

Kenichi Ishibashi is a professor of the Department of Medical Physiology, Meiji Pharmaceutical University. His research area is nephrology and water channels, aquaporins.

Yoshiyuki Morishita

Yoshiyuki Morishita is a professor of the Division of Nephrology, Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University. His research areas are nephrology, dialysis, gene delivery for the treatment of renal diseases, and analysis of microRNA profiling for the development of renal disease.

References

  • National Kidney Foundation. K/DOQI clinical practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Am J Kidney Dis. 2002;39:S1–S266. Epub 2002 Mar 21.
  • Hill NR, Fatoba ST, Oke JL, et al. Global prevalence of chronic kidney disease - a systematic review and meta-analysis. Plos One. 2016;11:1. Epub 2016 Jul 08.
  • Remuzzi G, Bertani T. Pathophysiology of progressive nephropathies. N Engl J Med. 1998;339:1448–8. Epub 1998 Nov 13.
  • Becker GJ, Hewitson TD. The role of tubulointerstitial injury in chronic renal failure. Curr Opin Nephrol Hypertens. 2000;9:133–138.
  • Humphreys BD, Lin S-L, Kobayashi A, et al. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol. 2010;176:85–97. Epub 2009 Dec 17.
  • Iwano M, Plieth D, Danoff TM, et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest. 2002;110:341–350. Epub 2002 Aug 7.
  • Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest. 2003;112:1776–1784. Epub 2003 Dec 18.
  • Li J, Qu X, Bertram JF. Endothelial-myofibroblast transition contributes to the early development of diabetic renal interstitial fibrosis in streptozotocin-induced diabetic mice. Am J Pathol. 2009;175:1380–1388. Epub 2009 Sep 5.
  • Broekema M, Harmsen MC, Van Luyn MJA, et al. Bone marrow-derived myofibroblasts contribute to the renal interstitial myofibroblast population and produce procollagen I after ischemia/reperfusion in rats. J Am Soc Nephrol. 2007;18:165–175. Epub 2006 Dec 1.
  • LeBleu VS, Taduri G, O’Connell J, et al. Origin and function of myofibroblasts in kidney fibrosis. Nat Med. 2013;19:1047–1053. Epub 2013 Jul 3.
  • Wong JKL, Mohseni R, Hamidieh AA, et al. Will nanotechnology bring new hope for gene delivery? Trends Biotechnol. 2017;35:434–451. Epub 2017 Jan 22.
  • Tang Y, Zeng Z, He X, et al. SiRNA crosslinked nanoparticles for the treatment of inflammation-induced liver injury. Adv Sci (Weinh). 2017;4:1600228. Epub 2017 Mar 3.
  • Chernousova S, Epple M. Live-cell imaging to compare the transfection and gene silencing efficiency of calcium phosphate nanoparticles and a liposomal transfection agent. Gene Ther. 2017. Epub 2017 Feb 22.
  • Natasha G, Gundogan B, Tan A, et al. Exosomes as immunotheranostic nanoparticles. Clin Ther. 2014;36:820–829. Epub 2014 May 28.
  • Campanholle G, Ligresti G, Gharib SA, et al. Cellular mechanisms of tissue fibrosis. 3. Novel mechanisms of kidney fibrosis. Am J Physiol Cell Physiol. 2013;304:C591–C603. Epub 2013 Jan 18.
  • Declèves A-E, Sharma K. New pharmacological treatments for improving renal outcomes in diabetes. Nat Rev Nephrol. 2010;6:371–380. Epub 2010 May 5.
  • Böttinger EP. TGF-beta in renal injury and disease. Semin Nephrol. 2007;27:309–320. Epub 2007 May 30.
  • Eddy AA. Molecular basis of renal fibrosis. Pediatr Nephrol. 2000;15:290–301. Epub 2001 Jan 10.
  • Boor P, Floege J. Chronic kidney disease growth factors in renal fibrosis. Clin Exp Pharmacol Physiol. 2011;38:441–450. Epub 2011 Feb 1.
  • Fujimoto M, Maezawa Y, Yokote K, et al. Mice lacking Smad3 are protected against streptozotocin-induced diabetic glomerulopathy. Biochem Biophys Res Commun. 2003;305:1002–1007. Epub 2003 May 28.
  • Sato M, Muragaki Y, Saika S, et al. Targeted disruption of TGF-beta1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J Clin Invest. 2003;112:1486–1494. Epub 2003 Nov 18.
  • Meng X-M, Huang XR, Xiao J, et al. Disruption of Smad4 impairs TGF-β/Smad3 and Smad7 transcriptional regulation during renal inflammation and fibrosis in vivo and in vitro. Kidney Int. 2012;81:266–279. Epub 2011 Nov 4.
  • Krensky AM, Ahn Y-T. Mechanisms of disease: regulation of RANTES (CCL5) in renal disease. Nat Clin Pract Nephrol. 2007;3:164–170. Epub 2007 Feb 27.
  • Guijarro C, Egido J. Transcription factor-kappa B (NF-kappa B) and renal disease. Kidney Int. 2001;59:415–424. Epub 2001 Feb 13.
  • Lange-Sperandio B, Trautmann A, Eickelberg O, et al. Leukocytes induce epithelial to mesenchymal transition after unilateral ureteral obstruction in neonatal mice. Am J Pathol. 2007;171:861–871. Epub 2007 Aug 7.
  • Sharma K, Jin Y, Guo J, et al. Neutralization of TGF-beta by anti-TGF-beta antibody attenuates kidney hypertrophy and the enhanced extracellular matrix gene expression in STZ-induced diabetic mice. Diabetes. 1996;45:522–530. Epub 1996 Apr 1.
  • Ziyadeh FN, Hoffman BB, Han DC, et al. Long-term prevention of renal insufficiency, excess matrix gene expression, and glomerular mesangial matrix expansion by treatment with monoclonal antitransforming growth factor-beta antibody in db/db diabetic mice. Proc Natl Acad Sci U S A. 2000;97:8015–8020. Epub 2000 Jun 22.
  • Lee SB, Kalluri R. Mechanistic connection between inflammation and fibrosis. Kidney Int Suppl. 2010;78:S22–S26. Epub 2010 Dec 1.
  • Imig JD, Ryan MJ. Immune and inflammatory role in renal disease. Compr Physiol. 2013;3:957–976. Epub 2013 May 31.
  • Van Linthout S, Miteva K, Tschöpe C. Crosstalk between fibroblasts and inflammatory cells. Cardiovasc Res. 2014;102:258–269. Epub 2014 Apr 15.
  • Herbelin A, Nguyen AT, Zingraff J, et al. Influence of uremia and hemodialysis on circulating interleukin-1 and tumor necrosis factor alpha. Kidney Int. 1990;37:116–125. Epub 1990 Jan 1.
  • Herbelin A, Ureña P, Nguyen AT, et al. Elevated circulating levels of interleukin-6 in patients with chronic renal failure. Kidney Int. 1991;39:954–960. Epub 1991 May 1.
  • Wilson HM, Chettibi S, Jobin C, et al. Inhibition of macrophage nuclear factor-kappaB leads to a dominant anti-inflammatory phenotype that attenuates glomerular inflammation in vivo. Am J Pathol. 2005;167:27–37. Epub 2005 Jun 24.
  • Anders HJ, Vielhauer V, Eis V, et al. Activation of toll-like receptor-9 induces progression of renal disease in MRL-Fas(lpr) mice. Faseb J. 2004;18:534–536. Epub 2004 Jan 22.
  • Kondo T, Takemura G, Kosai K, et al. Application of an adenoviral vector encoding soluble transforming growth factor-beta type II receptor to the treatment of diabetic nephropathy in mice. Clin Exp Pharmacol Physiol. 2008;35:1288–1293. Epub 2008 May 29.
  • Kim JI, Jang H-S, Jeong J-H, et al. Defect in Runx2 gene accelerates ureteral obstruction-induced kidney fibrosis via increased TGF-β signaling pathway. Biochim Biophys Acta. 2013;1832:1520–1527. Epub 2013 May 4.
  • Zhang Z, Wu F, Zheng F, et al. Adenovirus-mediated decorin gene transfection has therapeutic effects in a streptozocin-induced diabetic rat model. Nephron Exp Nephrol. 2010;116:e11–e21. Epub 2010 May 27.
  • Terada Y, Hanada S, Nakao A, et al. Gene transfer of Smad7 using electroporation of adenovirus prevents renal fibrosis in post-obstructed kidney. Kidney Int. 2002;61:S94–S98. Epub 2002 Feb 14.
  • Takase O, Hirahashi J, Takayanagi A, et al. Gene transfer of truncated IkappaBalpha prevents tubulointerstitial injury. Kidney Int. 2003;63:501–513. Epub 2003 Mar 13.
  • Schievenbusch S, Strack I, Scheffler M, et al. Combined paracrine and endocrine AAV9 mediated expression of hepatocyte growth factor for the treatment of renal fibrosis. Mol Ther. 2010;18:1302–1309. Epub 2010 Apr 29.
  • Mu W, Ouyang X, Agarwal A, et al. IL-10 suppresses chemokines, inflammation, and fibrosis in a model of chronic renal disease. J Am Soc Nephrol. 2005;16:3651–3660. Epub 2005 Oct 28.
  • Zhou Y, Lv C, Wu C, et al. Suppressor of cytokine signaling (SOCS) 2 attenuates renal lesions in rats with diabetic nephropathy. Acta Histochem. 2014;116:981–988. Epub 2014 May 8.
  • Wang C, Dobrzynski E, Chao J, et al. Adrenomedullin gene delivery attenuates renal damage and cardiac hypertrophy in Goldblatt hypertensive rats. Am J Physiol Renal Physiol. 2001;280:F964–F971. Epub 2001 May 16.
  • Savard S, Lavoie P, Villeneuve C, et al. eNOS gene delivery prevents hypertension and reduces renal failure and injury in rats with reduced renal mass. Nephrol Dial Transplant. 2012;27:2182–2190. Epub 2011 Nov 29.
  • Deng M, Luo Y, Li Y, et al. Klotho gene delivery ameliorates renal hypertrophy and fibrosis in streptozotocin-induced diabetic rats by suppressing the Rho-associated coiled-coil kinase signaling pathway. Mol Med Rep. 2015;12:45–54. Epub 2015 Feb 20.
  • Zhao G, Tu L, Li X, et al. Delivery of AAV2-CYP2J2 protects remnant kidney in the 5/6-nephrectomized rat via inhibition of apoptosis and fibrosis. Hum Gene Ther. 2012;23:688–699. Epub 2012 Jan 21.
  • Li X-G, Yan J-T, Xu X-Z, et al. Recombinant adeno-associated virus-mediated delivery of antisense angiotensin II receptor 1 gene attenuates hypertension development. Acta Pharmacol Sin. 2007;28:1737–1745. Epub 2007 Oct 26.
  • Zhou T-B, Qin Y-H, Lei F-Y, et al. Prohibitin attenuates oxidative stress and extracellular matrix accumulation in renal interstitial fibrosis disease. Plos One. 2013;8:e77187. Epub 2013 Nov 10.
  • Li L, Wu Y, Wang C, et al. Inhibition of PAX2 gene expression by siRNA (polyethylenimine) in experimental model of obstructive nephropathy. Ren Fail. 2012;34:1288–1296. Epub 2012 Oct 20.
  • Morishita Y, Imai T, Yoshizawa H, et al. Delivery of microRNA-146a with polyethylenimine nanoparticles inhibits renal fibrosis in vivo. Int J Nanomedicine. 2015;10:3475–3488. Epub 2015 May 23.
  • Tsujie M, Isaka Y, Ando Y, et al. Gene transfer targeting interstitial fibroblasts by the artificial viral envelope-type hemagglutinating virus of Japan liposome method. Kidney Int. 2000;57:1973–1980. Epub 2000 May 3.
  • Kushibiki T, Nagata-Nakajima N, Sugai M, et al. Enhanced anti-fibrotic activity of plasmid DNA expressing small interference RNA for TGF-beta type II receptor for a mouse model of obstructive nephropathy by cationized gelatin prepared from different amine compounds. J Control Release. 2006;110:610–617. Epub 2005 Dec 28.
  • Xia Z, Abe K, Furusu A, et al. Suppression of renal tubulointerstitial fibrosis by small interfering RNA targeting heat shock protein 47. Am J Nephrol. 2008;28:34–46. Epub 2007 Sep 25.
  • Aoyama T, Yamamoto S, Kanematsu A, et al. Local delivery of matrix metalloproteinase gene prevents the onset of renal sclerosis in streptozotocin-induced diabetic mice. Tissue Eng. 2003;9:1289–1299. Epub 2003 Dec 17.
  • Wold WSM, Toth K. Adenovirus vectors for gene therapy, vaccination and cancer gene therapy. Curr Gene Ther. 2013;13:421–433. Epub 2013 Nov 28.
  • Kamimura K, Suda T, Zhang G, et al. Advances in gene delivery systems. Pharmaceut Med. 2011;25:293–306. Epub 2011 Dec 28.
  • Chevalier RL, Forbes MS, Thornhill BA. Ureteral obstruction as a model of renal interstitial fibrosis and obstructive nephropathy. Kidney Int. 2009;75:1145–1152. Epub 2009 Apr 3.
  • Daya S, Berns KI. Gene therapy using adeno-associated virus vectors. Clin Microbiol Rev. 2008;21:583–593. Epub 2008 Oct 16.
  • Borel F, Kay MA, Mueller C. Recombinant AAV as a platform for translating the therapeutic potential of RNA interference. Mol Ther. 2014;22:692–701. Epub 2013 Dec 20.
  • Escors D, Breckpot K. Lentiviral vectors in gene therapy: their current status and future potential. Arch Immunol Ther Exp (Warsz). 2010;58:107–119. Epub 2010 Feb 10.
  • Yang T, Zhang B, Pat BK, et al. Lentiviral-mediated RNA interference against TGF-beta receptor type II in renal epithelial and fibroblast cell populations in vitro demonstrates regulated renal fibrogenesis that is more efficient than a nonlentiviral vector. J Biomed Biotechnol. 2010;2010:1–12. Epub 2010 Dec 15.
  • Lungwitz U, Breunig M, Blunk T, et al. Polyethylenimine-based non-viral gene delivery systems. Eur J Pharm Biopharm. 2005;60:247–266. Epub 2005 Jun 9.
  • Swami A, Kurupati RK, Pathak A, et al. A unique and highly efficient non-viral DNA/siRNA delivery system based on PEI-bisepoxide nanoparticles. Biochem Biophys Res Commun. 2007;362:835–841. Epub 2007 Sep 8.
  • Yoshizawa H, Morishita Y, Watanabe M, et al. TGF-β₁-siRNA delivery with nanoparticles inhibits peritoneal fibrosis. Gene Ther. 2015;22:333–340. Epub 2015 Jan 9.
  • Djagny VB, Wang Z, Xu S. Gelatin: a valuable protein for food and pharmaceutical industries: review. Crit Rev Food Sci Nutr. 2001;41:481–492. Epub 2001 Oct 11.
  • Azarmi S, Huang Y, Chen H, et al. Optimization of a two-step desolvation method for preparing gelatin nanoparticles and cell uptake studies in 143B osteosarcoma cancer cells. J Pharm Pharm Sci. 2006;9:124–132. Epub 2006 Jul 20.
  • Kitada M, Ogura Y, Koya D. Rodent models of diabetic nephropathy: their utility and limitations. Int J Nephrol Renovasc Dis. 2016;9:279–290. Epub 2016 Nov 25.
  • Tanaka T, Ichimaru N, Takahara S, et al. In vivo gene transfer of hepatocyte growth factor to skeletal muscle prevents changes in rat kidneys after 5/6 nephrectomy. Am J Transplant. 2002;2:828–836. Epub 2002 Oct 24.
  • Wei S, Xu C, Rychak JJ, et al. Short hairpin RNA knockdown of connective tissue growth factor by ultrasound-targeted microbubble destruction improves renal fibrosis. Ultrasound Med Biol. 2016;42:2926–2937. Epub 2016 Sep 7.