Background: The vascular endothelium serves as a semi-selective permeable barrier as a conduit for transport of fluid, solutes, and various cell populations between the vessel lumen and tissues. The endothelium thus has a dynamic role in the regulation of coagulation, immune system, lipid and electrolyte transport, as well as neurohumoral influences on vascular tone and end-organ injury to tissues such as the heart and kidney. Summary: Within this framework, pharmacologic strategies for heart and kidney diseases including blood pressure, glycemic control, and lipid reduction provide significant risk reduction, yet certain populations are at risk for substantial residual risk for disease progression and treatment resistance and often have unwanted off-target effects leaving the need for adjunct, alternative targeted therapies. Recent advances in techniques in sequencing and spatial transcriptomics have paved the way for the development of new therapies for targeting heart and kidney disease that include various gene, cell, and nano-based therapies. Cell-specific endothelium-specific targeting of viral vectors will enable their use for the treatment of heart and kidney diseases with gene therapy that can avoid unwanted off-target effects, improve treatment resistance, and reduce residual risk for disease progression. Key Messages: The vascular endothelium is an important therapeutic target for chronic kidney and cardiovascular diseases. Developing endothelial-specific gene therapies can benefit patients who develop resistance to current treatments.

Chronic kidney disease (CKD) is a global health burden that affects 850 million individuals, roughly 13% of the global population and exceeds that of diabetes [1, 2]. The dilemma regarding the management of CKD is the inordinate use of resources for the care of patients with CKD that progress to end stage and the care for accompanying comorbidities [3, 4]. Obesity and diabetes contribute significantly to the morbidity and mortality associated with the pre-dialysis population (e.g., CKD stage 1 through 4) [3‒5]. More specifically, cardiovascular diseases (CVDs) account for ∼40–50% of mortality in CKD and have a strong, continuous, graded relationship between decreases in estimated glomerular filtration rate and increasing incidents of CVD [6]. CVD risk factors in CKD range from high blood pressure, hyperglycemia, dyslipidemia, and tobacco use to non-traditional risk factors, such as vascular calcification and inflammation. High blood pressure remains the leading modifiable risk factor that can improve CVD outcomes [6]. Strategies to control blood pressure and those that target the renin-angiotensin-aldosterone system (RAAS) improve diabetic kidney disease outcomes to a greater extent than other anti-hypertensive regimens [7]. However, despite the widespread use of RAAS inhibition there is still a considerable “residual risk” for disease progression and enhanced risk for CVD. In this context, available therapies only provide a 20–30% risk reduction in CKD [4, 8] and there has been an active search for alternative treatment strategies from dual RAAS blockade to complementary glycemic control therapies such as sodium-glucose co-transporter-2 inhibition and glucagon-like peptide-1 agonism.

It is increasingly recognized that obesity not only is associated with an increased risk for CKD and CVD, but the presence of obesity enhances treatment resistance and residual risk for progression of disease. In the kidney, it is known that obesity alters intrarenal/intraglomerular hemodynamics by raising renal plasma flow and altering tubuloglomerular feedback mechanisms [5, 9‒11]. Obesity or the use of our Western diet has further shown to enhance the hyperfiltration mechanisms that lead to sodium retention, increased flow, and extracellular expansion [10]. However, from these observations it is difficult to distinguish between the confounding impact of associated hyperinsulinemia and insulin resistance, which have similar effects on vascular responses. Classically, insulin acts on the phosphatidylinositol 3-kinase (PI3K/Akt) pathway to facilitate glucose uptake and utilization [5]. However, impairments in insulin-dependent metabolic signaling contribute to reductions in bioavailable nitric oxide (NO), inflammation, oxidant stress, and growth and fibrotic pathways that trigger end-organ heart and kidney injuries.

Recent data indicate that in the kidney the glomerular endothelium, which is highly fenestrated and covered by a glycocalyx, is integral to the sieving properties of the glomerular filtration barrier and in the maintenance of the structure and function of podocytes that surround the glomerular capillaries. For instance, enhanced glomerular endothelium-related NO production increases blood flow and can alter the integrity of the filtration barrier, enhance filtration rate, and subsequent tubuloglomerular feedback mechanisms. Thereby, endothelial responses play a significant role in regulating kidney blood pressure and volume responses especially as it relates to the RAAS. In this regard, enhanced systemic and tissue RAAS in obesity occurs despite the salt and volume excess, which should suppress juxtaglomerular renin secretion. There is sufficient experimental evidence to support the concept that visceral adipose tissue enhances renin release, angiotensinogen, circulating aldosterone, and its mineralocorticoid receptor (MR) [9, 12‒14]. MR actions in heart failure and kidney diseases have been well defined clinically [14‒18]. Recent work has highlighted a role for MR on bioavailable NO and endothelial function as a link between heart and kidney diseases. Specifically, downstream actions of MR activation on the endothelial sodium channel (EnNaC) contribute to increased actin polymerization and cross-linking, reduced endothelial NO synthase (eNOS) activity and NO production leading to endothelial cell stiffening, impaired endothelial-dependent vessel relaxation, and target organ injury.

EnNaC as a molecular target: ENaC is known to regulate Na+ transport in kidney epithelial cells yet is also expressed in non-epithelial cells including endothelial cells (EnNaC) [4, 19]. ENaC consists of 3 subunits, α, β, and γ are members of the ENaC/degenerin family of ion channels, generally, all three subunits are essential for proper channel function and are expressed in vascular endothelial cells [19]. The distribution in kidney epithelial cells is pertinent to our understanding of ENaC regulation by flow-induced shear stress and mechanical forces. Work to date supports that western diet consumption increases aldosterone, which increases αEnNaC protein expression and its translocation to the cell membrane [20‒22], an effect mediated by MR activation in the endothelium [19]. EnNaC activation then results in polymerization of G- to F-actin [19] and reductions in bioavailable NO occur via negative regulation of eNOS during flow-mediated vasodilation [19, 22]. In this context, a number of studies support that EnNaC contributes to shear sensing in conduit and resistance arteries and regulates vascular endothelial stiffness [22, 23]. While vascular stiffening is associated with aging, it is independently associated with CKD and CVD [23] and is increasingly recognized as a therapeutic target for intervention.

Generally, endothelial stiffening is characterized by reduced NO bioavailability, increased expression of pro-inflammatory factors, and reduced endothelial-dependent vasodilation [19, 23]. This stiffness results from endothelial insults, which are dictated by a number of forces including epithelial cell myofibroblast activation and extracellular matrix stiffening [19, 20, 23]. Recent work supports that impaired endothelial NO production leads to extracellular matrix remodeling that contributes to endothelial stiffening and alterations in the endothelial glycocalyx that increase vessel wall permeability [21]. This relationship is dictated by applied tension and strain from derangements in cell-cell adhesions and the extracellular matrix via actin-myosin cytoskeletal rearrangements. Enhanced expression of endothelial cell adhesion molecules ICAM-1 (intercellular adhesion molecule-1) and VCAM-1 (vascular cell adhesion molecule-1) contribute to leukocyte/macrophage adhesion and infiltration that dictate endothelial stiffening [21, 22]. In this regard, consumption of a Western diet induces abnormal expression of tight junction proteins, including claudin-5 and activated endothelial cell MR, which in turn increases expression of VCAM-1, ICAM-1, and IL-6 that dictate glycocalyx degradation and stiffness.

Glycocalyx degradation EnNac and transglutaminases as a molecular target: In a well-developed glycocalyx, membrane expression of EnNaC is low, access of Na+ into the endothelial cells is limited, and NO release and vasodilation are maintained. In obesity, with higher vascular MR activity, membrane abundance of EnNaC is increased together with a deranged glycocalyx, facilitating Na+ entry into renal microcirculation endothelial cells and triggering the polymerization of G-actin to F-actin [21, 22]. This polymerization reduces caveolar eNOS release and bioavailable NO, which results in activation of TG2, collagen cross-linking, and endothelial stiffness [20‒22, 24]. In this regard, TG2 cross-links extracellular matrix proteins making them more resistant to breakdown and promote extracellular matrix remodeling. TG2 is expressed in the vasculature and kidney epithelial cells [19‒22, 24], where it is largely confined to the cytosol. Outside the cytosol, TG2 catalyzes a transamidation reaction [19, 25] enhancing cross-linking of extracellular matrix proteins. Recent work indicates that targeting ENaC and TG2 improves tight-junction protein expression, MMP-9 activation, and endothelium-dependent arterial relaxation suggesting their role in glycocalyx remodeling and increased endothelial permeability [22, 26]. Therefore, nanomechanics of endothelial stiffening in the microcirculation of the heart and kidney provide a rich environment for cell-specific molecular targeting.

The utility of targeting the endothelium has been limited by vectors that are non-selective and interact with numerous cell types that are permissive and allow transduction that limit their effectiveness in vascular-based disease such as in the heart and kidney. Gene therapy is an approach aiming to deliver or modify gene(s) in cells in order to alter their function and to gain a therapeutic effect. Currently, more than 20 gene therapy products have been approved by FDA or EMEA as gene medicines. Adenovirus, adeno-associated virus, or Lentivirus vectors are the most used gene therapy vehicles to deliver transgenes into the target tissue due to their efficacy and relative ease of large-scale, GMP-grade manufacturing. The vector of choice depends largely on the disease pathology, target tissue, the chosen delivery route, and desired duration of the transgene expression. Adenoviral vectors allow short-term transgene expression (weeks), and besides their current development or use as preventive vaccines, such as against COVID-19 [24], adenoviral vectors have been mostly developed for cancer therapy [27] and to treat ischemic diseases [28, 29]. In contrast to adenoviral vectors, lenitviral vectors and adeno-associated viral vectors allow more pro-longed transgene expression (months to years) and are more suitable for diseases requiring long-term therapeutic effects that can impact treatment resistance and disease progression.

All of these vector types have been tested for targeting the heart and kidney in animal models as gene therapy vectors. However, so far the main challenges have been a low efficacy and off-target expression of the transgene in the endothelium (reviewed in [30, 31]). In addition, due to the complex architecture of tissues like the heart and kidney and depending on the used delivery route and vector type/serotype variable results have been obtained. There are several choices for gene therapy vector delivery into a tissue such as the endothelium of the kidney, including systemic administration, anterograde delivery into the renal artery, retrograde administration, or intraparenchymal/subcapsular injection. Systemic delivery has been the most inefficient partly due to the vector sequestration into the liver and spleen. This could be circumvented by shielding the vectors with molecules, such as polyethylene glycol [32]. Also, certain adeno-associated viral serotypes have shown tropism to specific organs after systemic delivery. However, most of these studies have not been able to induce high transgene expression in the kidneys [31, 33, 34]. Of interest, the limited transduction of viral vectors following intravenous injection is also limited to the glomeruli [34, 35] and in the mouse kidney to the endothelial and mesangial cells of the glomerulus [35].

Direct delivery of gene therapy vectors to the kidneys has been a more promising delivery route to achieve high transduction efficiency. In a recent comparative study of three vector types, retrograde ureteral injections of the vectors to kidneys in mice were shown to result in a robust transduction of renal tubular cells [33] confirming previous observations [36‒38]. Higher transgene expression in off-target tissues was observed with adeno-associated viral than adenoviral or lenitviral vectors likely due to adeno-associated viral smaller capsid size [33]. Subcapsular injections were able to reduce the off-target expression, and AAV6.2 and AAV8 serotypes were shown to have the highest transduction efficiency in the kidneys [33]. Adenoviral vectors, however, only demonstrated transduction in the needle track and in addition showed damage to the renal parenchyma due to repeated injections [39]. To reduce the potential tissue damage, ultrasound-guided intraparenchymal injections of vectors have been tested and found to be feasible [40].

While most delivery methods that have been successful in the kidney, as an example, have resulted in transduction of the tubular epithelial cells, there are fewer reports of endothelial transduction. In context of endothelium targeting, slow infusion of the renal artery with adenoviral vector was shown to lead to a high transduction efficiency of the glomeruli in pigs [41] and in particular to the glomerular endothelium in rats [42]. As the glomerular barrier has been suggested to prevent virus vectors from reaching the kidney parenchyma, this could also be considered as a benefit to restrict vector entry into non-endothelial cells. Overall, improved retention of the vectors within the kidney and their cell-specific endothelial targeting are needed for the development of gene therapy for molecular targets such as EnNaC and TG2.

Targeting transduction would be ideal to redirect the virus vectors to a specific cell type, which would reduce off-target tissue expression. In addition, a combination of improved vector efficiency and cell-targeted delivery is likely to reduce immune responses to the vector or transgene due to lower doses required and limited biodistribution. One strategy to enable cellular targeting is to modify the surface molecules that interact with cellular receptors or molecules that facilitate viral entry. Modification of these viral proteins and thus a change in virus tropism has been achieved by introducing point mutations, using adapter molecules, inserting peptides or cysteine-moieties enabling coupling of the maleimide-activated ligands or pseudo-typing [42‒48]. Different serotypes of adeno-associated viral vectors exist naturally due to variations of their capsid proteins; however, these serotypes demonstrate a preference for a tissue and not specific cellular tropism. Much work on capsid engineering has been done, including using high-throughput, unbiased methods, such as directed evolution, to improve the specificity of adeno-associated vector to endothelial cells [43, 47, 49‒51]. However, this strategy is not feasible for all viral vectors; for example, for lentiviral vectors a change in the viral surface protein can negatively affect viral production [52]. We have used transferrin as a ligand to retarget adenoviral vectors across the endothelial monolayer [53], and αv integrin binding adenoviral vector was used to target kidney endothelium [54].

To circumvent limited viral tropism, other means of targeting transgene expression specifically to endothelial cells are necessary. Traditionally, one such method is the use of endothelial-specific promoters. An example is the FLT-1 promoter, which has been described in the context of Ad vectors to robustly and selectively drive the expression of a transgene in endothelial cells in contrast to ICAM-2 and vWF promoters [55]. Likewise, VE-cadherin promoter was used in an LV to drive the expression of the transgene in endothelial cells in the rat kidneys [56]. An additional advantage of using cell-type-specific promoters is the tendency for reduced liver expression, which most vectors sequester to [57]. However, cell-type-specific promoters have a tendency for lower transgene expression or “leakiness” in other cell types and this has limited their use. Other means of affecting endothelial targeting is via insertion of transcriptional regulatory elements to regulate the transgene expression. We have recently demonstrated such a strategy, which relied on the use of enhancers derived from endothelial-specific super enhancers [58]. Viral vectors containing this enhancer specifically activated reporter gene in endothelial cells and not in the other cell types. In addition, since this enhancer contained a hypoxia-inducible factor (HIF)-1a binding site, then reporter transgene was further increased in low oxygen condition. Hypoxia is a feature of some CVD and kidney diseases, making this vector an attractive option for endothelial-targeted viral vector-mediated therapy. Likewise, oxidative stress-regulated vectors may be useful for these diseases. A lentiviral vector containing an antioxidant response element induced transgene expression in cells with elevated nuclear factor E2 [59], which is a key transcription factor that facilitates intracellular redox balance and is activated in ischemia. Thus, in choosing an appropriate vector to target the endothelium in tissues like the kidney, many aspects need to be carefully considered including the choice of vector type, serotype, and design of the expression cassette. Although kidney injury has not been strongly on the gene therapy radar, encouragingly a number of groups have reported effective therapy mediated by gene therapy vectors in the kidney [30, 41, 60]. Furthermore, gene therapies targeting the kidney are likely to increase with advances in vector design and specificity that will overcome the wide cellular heterogeneity and architectural constraints of the kidney.

Recent advances on techniques, such as single-cell/-nuclei sequencing (scRNA-/snRNA-seq) and spatial transcriptomics, will pave the way for new therapies for endothelium-specific targeting of the heart and kidney. In sc/snRNA sequencing, cells or nuclei are isolated from the target tissue, allowing comparison of RNA transcripts within different cell types. Recent work has already shown the complexity and heterogeneity of kidney endothelial cells in adult mice, in which 24 endothelial cell subtypes were found, including 5 subtypes of glomerular endothelial cells alone [61]. Validation of these findings at the protein level is important in order to find the best targeting strategies for heart and kidney disease. As gene expression profiles of cells also change in different disease states, and most scRNA-seq studies have so far been performed in healthy animals, this should be taken into account when planning the endothelium targeting strategy. Gene expression profiles between human and mice likely also differ due to various factors such as different timelines of the disease development, genetics, age, and flow conditions. Besides sc/snRNA-seq, spatial transcriptomics, allowing detection of mRNAs spatially in the tissue context and comparison of different areas of the diseased tissue, will be an important novel tool to understand CKD pathology and to find the best targeting strategies of the endothelium.

The vascular endothelium should be thought of as a gatekeeper or sentinel to monitor and regulate not only vessel wall function but also target organ function like the heart and kidney. Within this framework, injury to the endothelium promotes pro-thrombotic-proinflammatory/oxidant pathways that impair vessel wall relaxation and stiffening as well as target organ injury. Current treatment options available for patients with CKD and CVD are limited to pharmacological compounds and achieve only a modest risk reduction in clinical trials, suggesting the need for adjunct and alternative therapies and targets. In this regard, recent advances in techniques, such as sc/snRNA sequencing and spatial transcriptomics, will pave the way for new therapies for targeting of CVD and CKD. The development of endothelial cell-specific targeting of virus vectors will enable their use for the treatment of heart and kidney diseases with gene therapy.

The authors have no conflicts of interest to declare.

This review was supported by the Research Council of Finland Flagship GeneCellNano program (Seppo Ylä-Herttuala, Nihay Laham-Karam, and Johanna P. Laakkonen) and research fellow (328835; Johanna P. Laakkonen) and the EU-funded ERC-advanced grant HeartGenes (884382; Seppo Ylä-Herttuala) and EU Horizon grant CardioReGenix (GA825670; Seppo Ylä-Herttuala). The funders had no role in the design, data collection, data analysis, and reporting of this study.

Nihay Laham-Karam, Johanna P. Laakkonen, and Adam Whaley-Connell conceptualized and planned the review. All authors (Nihay Laham-Karam, Johanna P. Laakkonen, Seppo Ylä-Herttuala, Annayya Aroor, Guanghong Jia, and Adam Whaley-Connell) wrote and/or edited the review.

Additional Information

Nihay Laham-Karam and Johanna P. Laakkonen should be considered as first authors.

1.
Jager
KJ
,
Kovesdy
C
,
Langham
R
,
Rosenberg
M
,
Jha
V
,
Zoccali
C
.
A single number for advocacy and communication-worldwide more than 850 million individuals have kidney diseases
.
Kidney Int
.
2019
;
96
(
5
):
1048
50
. .
2.
Hill
NR
,
Fatoba
ST
,
Oke
JL
,
Hirst
JA
,
O’Callaghan
CA
,
Lasserson
DS
, et al
.
Global prevalence of chronic kidney disease - a systematic review and meta-analysis
.
PLoS One
.
2016
;
11
(
7
):
e0158765
. .
3.
Tonneijck
L
,
Muskiet
MHA
,
Smits
MM
,
Van Bommel
EJ
,
Heerspink
HJL
,
Van Raalte
DH
, et al
.
Glomerular hyperfiltration in diabetes: mechanisms, clinical significance, and treatment
.
J Am Soc Nephrol
.
2017
;
28
(
4
):
1023
39
. .
4.
Vallon
V
,
Komers
R
.
Pathophysiology of the diabetic kidney
.
Compr Physiol
.
2011
;
1
(
3
):
1175
232
. .
5.
Whaley-Connell
A
,
Sowers
JR
.
Obesity and kidney disease: from population to basic science and the search for new therapeutic targets
.
Kidney Int
.
2017
;
92
(
2
):
313
23
. .
6.
Vaduganathan
M
,
Mensah
GA
,
Turco
JV
,
Fuster
V
,
Roth
GA
.
The global burden of cardiovascular diseases and risk: a compass for future health
.
J Am Coll Cardiol
.
2022
;
80
(
25
):
2361
71
. .
7.
Drawz
PE
,
Beddhu
S
,
Bignall
ONR
,
Cohen
JB
,
Flynn
JT
,
Ku
E
, et al
.
KDOQI US commentary on the 2021 KDIGO clinical practice guideline for the management of blood pressure in CKD
.
Am J Kidney Dis
.
2022
;
79
(
3
):
311
27
. .
8.
King
P
,
Peacock
I
,
Donnelly
R
.
The UK prospective diabetes study (UKPDS): clinical and therapeutic implications for type 2 diabetes
.
Br J Clin Pharmacol
.
1999
;
48
(
5
):
643
8
. .
9.
Hall
JE
,
Do Carmo
JM
,
Da Silva
AA
,
Wang
Z
,
Hall
ME
.
Obesity-induced hypertension: interaction of neurohumoral and renal mechanisms
.
Circ Res
.
2015
;
116
(
6
):
991
1006
. .
10.
Henegar
JR
,
Bigler
SA
,
Henegar
LK
,
Tyagi
SC
,
Hall
JE
.
Functional and structural changes in the kidney in the early stages of obesity
.
J Am Soc Nephrol
.
2001
;
12
(
6
):
1211
7
. .
11.
Hall
JE
,
Brands
MW
,
Dixon
WN
,
Smith
MJ
.
Obesity-induced hypertension. Renal function and systemic hemodynamics
.
Hypertension
.
1993
;
22
(
3
):
292
9
. .
12.
Giacchetti
G
,
Faloia
E
,
Mariniello
B
,
Sardu
C
,
Gatti
C
,
Camilloni
MA
, et al
.
Overexpression of the renin-angiotensin system in human visceral adipose tissue in normal and overweight subjects
.
Am J Hypertens
.
2002
;
15
(
5
):
381
8
. .
13.
Serazin-Leroy
V
,
Morot
M
,
De Mazancourt
P
,
Giudicelli
Y
.
Androgen regulation and site specificity of angiotensinogen gene expression and secretion in rat adipocytes
.
Am J Physiol Endocrinol Metab
.
2000
;
279
(
6
):
E1398
405
. .
14.
Dusserre
E
,
Moulin
P
,
Vidal
H
.
Differences in mRNA expression of the proteins secreted by the adipocytes in human subcutaneous and visceral adipose tissues
.
Biochim Biophys Acta
.
2000
;
1500
(
1
):
88
96
. .
15.
Brown
NJ
.
Contribution of aldosterone to cardiovascular and renal inflammation and fibrosis
.
Nat Rev Nephrol
.
2013
;
9
(
8
):
459
69
. .
16.
Calhoun
DA
,
Jones
D
,
Textor
S
,
Goff
DC
,
Murphy
TP
,
Toto
RD
, et al
.
Resistant hypertension: diagnosis, evaluation, and treatment. A scientific statement from the American Heart Association Professional Education Committee of the Council for High Blood Pressure Research
.
Hypertension
.
2008
;
51
(
6
):
1403
19
. .
17.
Epstein
M
,
Williams
GH
,
Weinberger
M
,
Lewin
A
,
Krause
S
,
Mukherjee
R
, et al
.
Selective aldosterone blockade with eplerenone reduces albuminuria in patients with type 2 diabetes
.
Clin J Am Soc Nephrol
.
2006
;
1
(
5
):
940
51
. .
18.
Mehdi
UF
,
Adams-Huet
B
,
Raskin
P
,
Vega
GL
,
Toto
RD
.
Addition of angiotensin receptor blockade or mineralocorticoid antagonism to maximal angiotensin-converting enzyme inhibition in diabetic nephropathy
.
J Am Soc Nephrol
.
2009
;
20
(
12
):
2641
50
. .
19.
Aroor
AR
,
Jia
G
,
Sowers
JR
.
Cellular mechanisms underlying obesity-induced arterial stiffness
.
Am J Physiol Regul Integr Comp Physiol
.
2018
;
314
(
3
):
R387
98
. .
20.
Jia
G
,
Aroor
AR
,
Sowers
JR
.
Arterial stiffness: a nexus between cardiac and renal disease
.
Cardiorenal Med
.
2014
;
4
(
1
):
60
71
. .
21.
Aroor
AR
,
Habibi
J
,
Nistala
R
,
Ramirez-Perez
FI
,
Martinez-Lemus
LA
,
Jaffe
IZ
, et al
.
Diet-induced obesity promotes kidney endothelial stiffening and fibrosis dependent on the endothelial mineralocorticoid receptor
.
Hypertension
.
2019
;
73
(
4
):
849
58
. .
22.
Jia
G
,
Habibi
J
,
Aroor
AR
,
Hill
MA
,
Yang
Y
,
Whaley-Connell
A
, et al
.
Epithelial sodium channel in aldosterone-induced endothelium stiffness and aortic dysfunction
.
Hypertension
.
2018
;
72
(
3
):
731
8
. .
23.
Jia
G
,
Aroor
AR
,
Hill
MA
,
Sowers
JR
.
Role of renin-angiotensin-aldosterone system activation in promoting cardiovascular fibrosis and stiffness
.
Hypertension
.
2018
;
72
(
3
):
537
48
. .
24.
Sadoff
J
,
Gray
G
,
Vandebosch
A
,
Cárdenas
V
,
Shukarev
G
,
Grinsztejn
B
, et al
.
Final analysis of efficacy and safety of single-dose Ad26.COV2.S
.
N Engl J Med
.
2022
;
386
(
9
):
847
60
. .
25.
Pavlov
TS
,
Staruschenko
A
.
Involvement of ENaC in the development of salt-sensitive hypertension
.
Am J Physiol Renal Physiol
.
2017
;
313
(
2
):
F135
40
. .
26.
Ramirez-Perez
FI
,
Cabral-Amador
FJ
,
Whaley-Connell
AT
,
Aroor
AR
,
Morales-Quinones
M
,
Woodford
ML
, et al
.
Cystamine reduces vascular stiffness in Western diet-fed female mice
.
Am J Physiol Heart Circ Physiol
.
2022
;
322
(
2
):
H167
80
. .
27.
Boorjian
SA
,
Alemozaffar
M
,
Konety
BR
,
Shore
ND
,
Gomella
LG
,
Kamat
AM
, et al
.
Intravesical nadofaragene firadenovec gene therapy for BCG-unresponsive non-muscle-invasive bladder cancer: a single-arm, open-label, repeat-dose clinical trial
.
Lancet Oncol
.
2021
;
22
(
1
):
107
17
. .
28.
Hartikainen
J
,
Hassinen
I
,
Hedman
A
,
Kivelä
A
,
Saraste
A
,
Knuuti
J
, et al
.
Adenoviral intramyocardial VEGF-DΔNΔC gene transfer increases myocardial perfusion reserve in refractory angina patients: a phase I/IIa study with 1-year follow-up
.
Eur Heart J
.
2017
;
38
(
33
):
2547
55
. .
29.
Pajula
J
,
Lähteenvuo
J
,
Lähteenvuo
M
,
Honkonen
K
,
Halonen
P
,
Hätinen
OP
, et al
.
Adenoviral VEGF-DΔN ΔC gene therapy for myocardial ischemia
.
Front Bioeng Biotechnol
.
2022
;
10
:
999226
. .
30.
Peek
JL
,
Wilson
MH
.
Gene therapy for kidney disease: targeting cystinuria
.
Curr Opin Nephrol Hypertens
.
2022
;
31
(
2
):
175
9
. .
31.
Davis
L
,
Park
F
.
Gene therapy research for kidney diseases
.
Physiol Genomics
.
2019
;
51
(
9
):
449
61
. .
32.
Kreppel
F
,
Gackowski
J
,
Schmidt
E
,
Kochanek
S
.
Combined genetic and chemical capsid modifications enable flexible and efficient de- and retargeting of adenovirus vectors
.
Mol Ther
.
2005
;
12
(
1
):
107
17
. .
33.
Rubin
JD
,
Nguyen
TV
,
Allen
KL
,
Ayasoufi
K
,
Barry
MA
.
Comparison of gene delivery to the kidney by adenovirus, adeno-associated virus, and lentiviral vectors after intravenous and direct kidney injections
.
Hum Gene Ther
.
2019
;
30
(
12
):
1559
71
. .
34.
Zincarelli
C
,
Soltys
S
,
Rengo
G
,
Rabinowitz
JE
.
Analysis of AAV serotypes 1-9 mediated gene expression and tropism in mice after systemic injection
.
Mol Ther
.
2008
;
16
(
6
):
1073
80
. .
35.
Lang
JF
,
Toulmin
SA
,
Brida
KL
,
Eisenlohr
LC
,
Davidson
BL
.
Standard screening methods underreport AAV-mediated transduction and gene editing
.
Nat Commun
.
2019
;
10
(
1
):
3415
0
. .
36.
Moullier
P
,
Friedlander
G
,
Calise
D
,
Ronco
P
,
Perricaudet
M
,
Ferry
N
.
Adenoviral-mediated gene transfer to renal tubular cells in vivo
.
Kidney Int
.
1994
;
45
(
4
):
1220
5
. .
37.
Gusella
GL
,
Fedorova
E
,
Marras
D
,
Klotman
PE
,
Klotman
ME
.
In vivo gene transfer to kidney by lentiviral vector
.
Kidney Int
.
2002
;
61
(
1 Suppl l
):
S32
S36
. .
38.
Takeda
SI
,
Takahashi
M
,
Mizukami
H
,
Kobayashi
E
,
Takeuchi
K
,
Hakamata
Y
, et al
.
Successful gene transfer using adeno-associated virus vectors into the kidney: comparison among adeno-associated virus serotype 1-5 vectors in vitro and in vivo
.
Nephron Exp Nephrol
.
2004
;
96
(
4
):
e119
e126
. .
39.
Watanabe
S
,
Ogasawara
T
,
Tamura
Y
,
Saito
T
,
Ikeda
T
,
Suzuki
N
, et al
.
Targeting gene expression to specific cells of kidney tubules in vivo, using adenoviral promoter fragments
.
PLoS One
.
2017
;
12
(
3
):
e0168638
. .
40.
Espana-Agusti
J
,
Tuveson
DA
,
Adams
DJ
,
Matakidou
A
.
A minimally invasive, lentiviral based method for the rapid and sustained genetic manipulation of renal tubules
.
Sci Rep
.
2015
;
5
:
11061
. .
41.
Heikkilä
P
,
Tibell
A
,
Morita
T
,
Chen
Y
,
Wu
G
,
Sado
Y
, et al
.
Adenovirus-mediated transfer of type IV collagen alpha5 chain cDNA into swine kidney in vivo: deposition of the protein into the glomerular basement membrane
.
Gene Ther
.
2001
;
8
(
11
):
882
90
. .
42.
Ye
X
,
Liu
XH
,
Li
Z
,
Ray
PE
.
Efficient gene transfer to rat renal glomeruli with recombinant adenoviral vectors
.
Hum Gene Ther
.
2001
;
12
(
2
):
141
8
. .
43.
Khachigian
LM
,
Varcoe
RL
,
Suoranta
T
,
Laham-Karam
N
,
Ylä-Herttuala
S
.
Gene therapeutic strategies for peripheral artery disease and new opportunities provided by adeno-associated virus vectors
.
Arterioscler Thromb Vasc Biol
.
2023
;
43
(
6
):
836
51
. .
44.
Gutierrez-Guerrero
A
,
Cosset
FL
,
Verhoeyen
E
.
Lentiviral vector pseudotypes: precious tools to improve gene modification of hematopoietic cells for research and gene therapy
.
Viruses
.
2020
;
12
(
9
):
1016
. .
45.
Syyam
A
,
Nawaz
A
,
Ijaz
A
,
Sajjad
U
,
Fazil
A
,
Irfan
S
, et al
.
Adenovirus vector system: construction, history and therapeutic applications
.
Biotechniques
.
2022
;
73
(
6
):
297
305
. .
46.
Weklak
D
,
Pembaur
D
,
Koukou
G
,
Jönsson
F
,
Hagedorn
C
,
Kreppel
F
.
Genetic and chemical capsid modifications of adenovirus vectors to modulate vector-host interactions
.
Viruses
.
2021
;
13
(
7
):
1300
. .
47.
Denby
L
,
Work
LM
,
Seggern
DJV
,
Wu
E
,
McVey
JH
,
Nicklin
SA
, et al
.
Development of renal-targeted vectors through combined in vivo phage display and capsid engineering of adenoviral fibers from serotype 19p
.
Mol Ther
.
2007
;
15
(
9
):
1647
54
. .
48.
Nicklin
SA
,
White
SJ
,
Watkins
SJ
,
Hawkins
RE
,
Baker
AH
.
Selective targeting of gene transfer to vascular endothelial cells by use of peptides isolated by phage display
.
Circulation
.
2000
;
102
(
2
):
231
7
. .
49.
Krolak
T
,
Chan
KY
,
Kaplan
L
,
Huang
Q
,
Wu
J
,
Zheng
Q
, et al
.
A high-efficiency AAV for endothelial cell transduction throughout the central nervous system
.
Nat Cardiovasc Res
.
2022
;
1
(
4
):
389
400
, .
50.
Liu
YB
,
Xu
BC
,
Chen
YT
,
Yuan
X
,
Liu
JY
,
Liu
T
, et al
.
Directed evolution of AAV accounting for long-term and enhanced transduction of cardiovascular endothelial cells in vivo
.
Mol Ther Methods Clin Dev
.
2021
;
22
:
148
61
. .
51.
White
SJ
,
Nicklin
SA
,
Büning
H
,
Brosnan
MJ
,
Leike
K
,
Papadakis
ED
, et al
.
Targeted gene delivery to vascular tissue in vivo by tropism-modified adeno-associated virus vectors
.
Circulation
.
2004
;
109
(
4
):
513
9
. .
52.
Boudeffa
D
,
Bertin
B
,
Biek
A
,
Mormin
M
,
Leseigneur
F
,
Galy
A
, et al
.
Toward a scalable purification protocol of GaLV-TR-pseudotyped lentiviral vectors
.
Hum Gene Ther Methods
.
2019
;
30
(
5
):
153
71
. .
53.
Laakkonen
JP
,
Engler
T
,
Romero
IA
,
Weksler
B
,
Couraud
PO
,
Kreppel
F
, et al
.
Transcellular targeting of fiber- and hexon-modified adenovirus vectors across the brain microvascular endothelial cells in vitro
.
PLoS One
.
2012
;
7
(
9
):
e45977
. .
54.
McDonald
GA
,
Zhu
G
,
Li
Y
,
Kovesdi
I
,
Wickham
TJ
,
Sukhatme
VP
.
Efficient adenoviral gene transfer to kidney cortical vasculature utilizing a fiber modified vector
.
J Gene Med
.
1999
;
1
(
2
):
103
10
. .
55.
Nicklin
SA
,
Reynolds
PN
,
Brosnan
MJ
,
White
SJ
,
Curiel
DT
,
Dominiczak
AF
, et al
.
Analysis of cell-specific promoters for viral gene therapy targeted at the vascular endothelium
.
Hypertension
.
2001
;
38
(
1
):
65
70
. .
56.
Cao
J
,
Sodhi
K
,
Inoue
K
,
Quilley
J
,
Rezzani
R
,
Rodella
L
, et al
.
Lentiviral-human heme oxygenase targeting endothelium improved vascular function in angiotensin II animal model of hypertension
.
Hum Gene Ther
.
2011
;
22
(
3
):
271
82
. .
57.
Powell
SK
,
Rivera-Soto
R
,
Gray
SJ
.
Viral expression cassette elements to enhance transgene target specificity and expression in gene therapy
.
Discov Med
.
2015
;
19
(
102
):
49
57
.
58.
Mushimiyimana
I
,
Niskanen
H
,
Beter
M
,
Laakkonen
JP
,
Kaikkonen
MU
,
Ylä-Herttuala
S
, et al
.
Characterization of a functional endothelial super-enhancer that regulates ADAMTS18 and angiogenesis
.
Nucleic Acids Res
.
2021
;
49
(
14
):
8078
96
. .
59.
Hurttila
H
,
Koponen
JK
,
Kansanen
E
,
Jyrkkänen
HK
,
Kivelä
A
,
Kylätie
R
, et al
.
Oxidative stress-inducible lentiviral vectors for gene therapy
.
Gene Ther
.
2008
;
15
(
18
):
1271
9
. .
60.
Ding
WY
,
Kuzmuk
V
,
Hunter
S
,
Lay
A
,
Hayes
B
,
Beesley
M
, et al
.
Adeno-associated virus gene therapy prevents progression of kidney disease in genetic models of nephrotic syndrome
.
Sci Transl Med
.
2023
;
15
(
708
):
eabc8226
. .
61.
Dumas
SJ
,
Meta
E
,
Borri
M
,
Goveia
J
,
Rohlenova
K
,
Conchinha
NV
, et al
.
Single-cell RNA sequencing reveals renal endothelium heterogeneity and metabolic adaptation to water deprivation
.
J Am Soc Nephrol
.
2020
;
31
(
1
):
118
38
. .